101
|
Ray RS, Katyal A. Myeloperoxidase: Bridging the gap in neurodegeneration. Neurosci Biobehav Rev 2016; 68:611-620. [PMID: 27343997 DOI: 10.1016/j.neubiorev.2016.06.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 10/21/2022]
Abstract
Neurodegenerative conditions present a group of complex disease pathologies mostly due to unknown aetiology resulting in neuronal death and permanent neurological disability. Any undesirable stress to the brain, disrupts homeostatic balance, through a remarkable convergence of pathophysiological changes and immune dysregulation. The crosstalk between inflammatory and oxidative mechanisms results in the release of neurotoxic mediators apparently spearheaded by myeloperoxidase derived from activated microglia, astrocytes, neurons as well as peripheral inflammatory cells. These isolated entities combinedly have the potential to flare up and contribute significantly to neuropathology and disease progression. Recent, clinicopathological evidence support the association of myeloperoxidase and its cytotoxic product, hypochlorous acid in a plethora of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Multiple sclerosis, Stroke, Epilepsy etc. But the biochemical and mechanistic insights into myeloperoxidase mediated neuroinflammation and neuronal death is still an uncharted territory. The current review outlines the emerging recognition of myeloperoxidase in neurodegeneration, which may offer novel therapeutic and diagnostic targets for neurodegenerative disorders.
Collapse
Affiliation(s)
- R S Ray
- Dr. B.R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, North Campus, Delhi 110 007, India.
| | - Anju Katyal
- Dr. B.R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, North Campus, Delhi 110 007, India.
| |
Collapse
|
102
|
Liver X receptors regulate cerebrospinal fluid production. Mol Psychiatry 2016; 21:844-56. [PMID: 26324101 DOI: 10.1038/mp.2015.133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/23/2015] [Accepted: 07/28/2015] [Indexed: 11/08/2022]
Abstract
Of the two isoforms of Liver X receptor (LXR), LXRβ has been shown to have major effects in the central nervous system (CNS) and on the regulation of aquaporins while LXRα has its most marked effects on cholesterol homeostasis. Both receptors have immunomodulatory functions. In LXRαβ knockout (ko) mice, the CNS phenotype is much more severe than in the LXRβ ko mice, suggesting a contribution of LXRα in CNS functions. One of the most striking abnormalities in the brains of LXRαβ ko mice is the occlusion of the lateral ventricles with age. In the present study, we have found by immunohistochemical staining that both LXRα and LXRβ are expressed in the cell nuclei of the epithelium of the choroid plexus and in the ependymal cells surrounding the lateral ventricles. The two receptors regulate several genes and can compensate for each other on expression of genes involved in structural integrity (E-cadherin, P-cadherin and β-catenin) and function (aquaporin 1 and carbonic anhydrase IX). Aquaporin 4 (AQ4) is not expressed in the choroid plexus but is expressed in the astrocytic end feet and ependymal cells. AQP4 expression was increased in white matter around lateral ventricles but not in neurons of LXRαβ ko mice. The data show that LXR is a regulator of cerebrospinal fluid (CSF) both at the choroid plexus and at the astrocytic end feet and defects in the synthesis of cerebrospinal fluid may be targeted by LXR agonists to facilitate CSF production, turnover and clearance in CNS diseases.
Collapse
|
103
|
Abstract
PURPOSE OF REVIEW This article evaluates recent experimental and human evidence regarding the involvement of lipids, lipoproteins, and apolipoproteins in neurodegenerative diseases, and reviews the current literature of the effects of cholesterol-lowering treatment on cognition. RECENT FINDINGS Plasma levels of traditional lipids and lipoproteins are not consistently associated with risk of dementia even though low plasma levels of apolipoprotein E, through unknown mechanisms, robustly predict future dementia. Experimental evidence suggests neuroprotective roles of several brain and cerebrospinal fluid apolipoproteins. Whether plasma levels of apolipoprotein E, or any other apolipoprotein with possible central nervous system and/or blood-brain barrier functions (apolipoproteins J, A-I, A-II, A-IV, D, C-I, and C-III) may become accessible biomarker components that improve risk prediction for dementia together with genetic risk variants and cardiovascular risk factors remains to be determined. SUMMARY Apolipoproteins with well established functions in peripheral lipid metabolism may play important roles for brain vascular health and Alzheimer's disease pathophysiology. Experimental work on lipids, lipoproteins, and apolipoproteins in the central nervous system together with robust prospective human studies will help to substantiate the drug target potential of these lipid components.
Collapse
Affiliation(s)
- Cheryl L Wellington
- aDepartment of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada bDepartment of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospitals cFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
104
|
Courtney R, Landreth GE. LXR Regulation of Brain Cholesterol: From Development to Disease. Trends Endocrinol Metab 2016; 27:404-414. [PMID: 27113081 PMCID: PMC4986614 DOI: 10.1016/j.tem.2016.03.018] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/07/2023]
Abstract
Liver X receptors (LXRs) are master regulators of cholesterol homeostasis and inflammation in the central nervous system (CNS). The brain, which contains a disproportionately large amount of the body's total cholesterol (∼25%), requires a complex and delicately balanced cholesterol metabolism to maintain neuronal function. Dysregulation of cholesterol metabolism has been implicated in numerous neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Due to their cholesterol-sensing and anti-inflammatory activities, LXRs are positioned centrally in the everyday maintenance of CNS function. This review focuses on recent research into the role of LXRs in the CNS during normal development and homeostasis and in disease states.
Collapse
Affiliation(s)
- Rebecca Courtney
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Gary E Landreth
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
105
|
Du SQ, Wang XR, Xiao LY, Tu JF, Zhu W, He T, Liu CZ. Molecular Mechanisms of Vascular Dementia: What Can Be Learned from Animal Models of Chronic Cerebral Hypoperfusion? Mol Neurobiol 2016; 54:3670-3682. [PMID: 27206432 DOI: 10.1007/s12035-016-9915-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/03/2016] [Indexed: 01/06/2023]
Abstract
Vascular dementia (VD) is defined as a progressive neurodegenerative disease of cognitive decline, attributable to cerebrovascular factors. Numerous studies have demonstrated that chronic cerebral hypoperfusion (CCH) is associated with the initiation and progression of VD and Alzheimer's disease (AD). Suitable animal models were established to replicate such pathological condition in experimental research, which contributes largely to comprehending causal relationships between CCH and cognitive impairment. The most widely used experimental model of VD and CCH is permanent bilateral common carotid artery occlusion in rats. In CCH models, changes of learning and memory, cerebral blood flow (CBF), energy metabolism, and neuropathology initiated by ischemia were revealed. However, in order to achieve potential therapeutic targets, particular mechanisms in cognitive and neuropathological changes from CCH to dementia should be investigated. Recent studies have shown that hypoperfusion resulted in a chain of disruption of homeostatic interactions, including oxidative stress, neuroinflammation, neurotransmitter system dysfunction, mitochondrial dysfunction, disturbance of lipid metabolism, and alterations of growth factors. Evidence from experimental studies that elucidate the damaging effects of such imbalances suggests their critical roles in the pathogenesis of VD. The present review provides a summary of the achievements in mechanisms made with the CCH models, permits an understanding of the causative role played by CCH in VD, and highlights preventative and therapeutic prospects.
Collapse
Affiliation(s)
- Si-Qi Du
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Ling-Yong Xiao
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Jian-Feng Tu
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Wen Zhu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Tian He
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Cun-Zhi Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China.
| |
Collapse
|
106
|
Sandoval-Hernández AG, Restrepo A, Cardona-Gómez GP, Arboleda G. LXR activation protects hippocampal microvasculature in very old triple transgenic mouse model of Alzheimer’s disease. Neurosci Lett 2016; 621:15-21. [DOI: 10.1016/j.neulet.2016.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/30/2016] [Accepted: 04/04/2016] [Indexed: 02/07/2023]
|
107
|
Wu CH, Chen CC, Lai CY, Hung TH, Lin CC, Chao M, Chen SF. Treatment with TO901317, a synthetic liver X receptor agonist, reduces brain damage and attenuates neuroinflammation in experimental intracerebral hemorrhage. J Neuroinflammation 2016; 13:62. [PMID: 26968836 PMCID: PMC4788882 DOI: 10.1186/s12974-016-0524-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/03/2016] [Indexed: 11/10/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) induces a series of inflammatory processes that contribute to neuronal damage and neurological deterioration. Liver X receptors (LXRs) are nuclear receptors that negatively regulate transcriptional processes involved in inflammatory responses, but their role in the pathology following ICH remains unclear. The present study investigated the neuroprotective effects and anti-inflammatory actions of TO901317, a synthetic LXR agonist, in a model of collagenase-induced ICH and in microglial cultures. Methods Mice subjected to collagenase-induced ICH injury were injected with either TO901317 (30 mg/kg) or vehicle 10 min after ICH and subsequently daily for 2 days. Behavioral studies, histology analysis, and assessments of hematoma volumes, brain water content, and blood-brain barrier (BBB) permeability were performed. The protein expression of LXR-α, LXR-β, ATP binding cassette transporter-1 (ABCA-1), and inflammatory molecules was analyzed. The anti-inflammatory mechanism of TO901317 was investigated in cultured microglia that were stimulated with either lipopolysaccharide (LPS) or thrombin. Results ICH induced an increase in LXR-α protein levels in the hemorrhagic hemisphere at 6 h whereas LXR-β expression remained unaffected. Both LXR-α and LXR-β were expressed in neurons and microglia in the peri-ICH region and but rarely in astrocytes. TO901317 significantly attenuated functional deficits and brain damage up to 28 days post-ICH. TO901317 also reduced neuronal death, BBB disruption, and brain edema at day 4 post-ICH. These changes were associated with marked reductions in microglial activation, neutrophil infiltration, and expression levels of inflammatory mediators at 4 and 7 days. However, TO901317 had no effect on matrix metalloproteinase-9 activity. In BV2 microglial cultures, TO901317 attenuated LPS- and thrombin-stimulated nitric oxide production and reduced LPS-induced p38, JNK, MAPK, and nuclear factor-kappa B (NF-κB) signaling. Moreover, delaying administration of TO901317 to 3 h post-ICH reduced brain tissue damage and neuronal death. Conclusions Our results suggest that enhancing LXR activation may provide a potential therapy for ICH by modulating the cytotoxic functions of microglia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0524-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chun-Hu Wu
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chai-You Lai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Taipei and College of Medicine, Chang Gung University, Taipei, Taiwan, Republic of China
| | - Chao-Chang Lin
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Min Chao
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China.
| |
Collapse
|
108
|
Anastasio TJ. Temporal-logic analysis of microglial phenotypic conversion with exposure to amyloid-β. MOLECULAR BIOSYSTEMS 2016; 11:434-53. [PMID: 25406664 DOI: 10.1039/c4mb00457d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer Disease (AD) remains a leading killer with no adequate treatment. Ongoing research increasingly implicates the brain's immune system as a critical contributor to AD pathogenesis, but the complexity of the immune contribution poses a barrier to understanding. Here I use temporal logic to analyze a computational specification of the immune component of AD. Temporal logic is an extension of logic to propositions expressed in terms of time. It has traditionally been used to analyze computational specifications of complex engineered systems but applications to complex biological systems are now appearing. The inflammatory component of AD involves the responses of microglia to the peptide amyloid-β (Aβ), which is an inflammatory stimulus and a likely causative AD agent. Temporal-logic analysis of the model provides explanations for the puzzling findings that Aβ induces an anti-inflammatory and well as a pro-inflammatory response, and that Aβ is phagocytized by microglia in young but not in old animals. To potentially explain the first puzzle, the model suggests that interferon-γ acts as an "autocrine bridge" over which an Aβ-induced increase in pro-inflammatory cytokines leads to an increase in anti-inflammatory mediators also. To potentially explain the second puzzle, the model identifies a potential instability in signaling via insulin-like growth factor 1 that could explain the failure of old microglia to phagocytize Aβ. The model predicts that augmentation of insulin-like growth factor 1 signaling, and activation of protein kinase C in particular, could move old microglia from a neurotoxic back toward a more neuroprotective and phagocytic phenotype.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Computational Neurobiology Laboratory, Department of Molecular and Integrative Physiology, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
109
|
Bao X, Cai Y, Wang Y, Zhao J, He X, Yu D, Huang J, Jing S, Du Z, Yang T, Warner M, Gustafsson JA, Fan X. Liver X Receptor β Is Involved in Formalin-Induced Spontaneous Pain. Mol Neurobiol 2016; 54:1467-1481. [PMID: 26846362 DOI: 10.1007/s12035-016-9737-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/20/2016] [Indexed: 12/30/2022]
Abstract
Increasing evidence indicates that the liver X receptor(LXR) β modulates inflammatory pain. However, the molecular mechanisms through which LXRβ modulates pain are unclear. Here, we found that LXRβ-null mice responded more strongly to acute noxious stimuli than wild-type (WT) littermates (in the hot plate and Hargreaves tests) and had augmented tonic inflammatory pain (in the formalin test). This increased reactivity to inflammatory pain was accompanied by enhanced formalin-evoked Fos and pERK staining of second-order nociceptive neurons. Immunohistochemistry showed that the expression of CGRP, SP, and IB4 was increased in the lamina I-II of the lumbar dorsal horns in formalin-injected LXRβ knockout (KO) mice compared with the WT controls. In addition, LXRβ deletion in the mice enhanced the formalin-induced inflammation with more activated microglia and astrocytes in the spinal cord. Furthermore, the levels of pro-inflammatory cytokines (IL-1β ,TNF-α) as well as NFκB in the formalin-injected paw were elevated by the loss of LXRβ. Taken together, these data indicate that LXRβ is involved in acute as well as inflammatory pain, and thus, it may be considered as a new target for the development of analgesics.
Collapse
Affiliation(s)
- Xiaohang Bao
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China.,Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Yulong Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Ying Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Jinghui Zhao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Xie He
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Dan Yu
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Jing Huang
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Sheng Jing
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Zhiyong Du
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | - Tiande Yang
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China.
| | - Margaret Warner
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77054, USA
| | - Jan-Ake Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77054, USA. .,Department of Biosciences and Nutrition, Karolinska Institute, Novum, Novum, 141 86, Sweden.
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
110
|
Sandoval-Hernández A, Contreras MJ, Jaramillo J, Arboleda G. Regulation of Oligodendrocyte Differentiation and Myelination by Nuclear Receptors: Role in Neurodegenerative Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:287-310. [DOI: 10.1007/978-3-319-40764-7_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
111
|
Sandoval-Hernández AG, Buitrago L, Moreno H, Cardona-Gómez GP, Arboleda G. Role of Liver X Receptor in AD Pathophysiology. PLoS One 2015; 10:e0145467. [PMID: 26720273 PMCID: PMC4697813 DOI: 10.1371/journal.pone.0145467] [Citation(s) in RCA: 33] [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: 05/29/2015] [Accepted: 12/03/2015] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease (AD) is the major cause of dementia worldwide. The pharmacological activation of nuclear receptors (Liver X receptors: LXRs or Retinoid X receptors: RXR) has been shown to induce overexpression of the ATP-Binding Cassette A1 (ABCA1) and Apolipoprotein E (ApoE), changes that are associated with improvement in cognition and reduction of amyloid beta pathology in amyloidogenic AD mouse models (i.e. APP, PS1: 2tg-AD). Here we investigated whether treatment with a specific LXR agonist has a measurable impact on the cognitive impairment in an amyloid and Tau AD mouse model (3xTg-AD: 12-months-old; three months treatment). The data suggests that the LXR agonist GW3965 is associated with increased expression of ApoE and ABCA1 in the hippocampus and cerebral cortex without a detectable reduction of the amyloid load. We also report that most cells overexpressing ApoE (86±12%) are neurons localized in the granular cell layer of the hippocampus and entorhinal cortex. In the GW3965 treated 3xTg-AD mice we also observed reduction in astrogliosis and increased number of stem and proliferating cells in the subgranular zone of the dentate gyrus. Additionally, we show that GW3965 rescued hippocampus long term synaptic plasticity, which had been disrupted by oligomeric amyloid beta peptides. The effect of GW3965 on synaptic function was protein synthesis dependent. Our findings identify alternative functional/molecular mechanisms by which LXR agonists may exert their potential benefits as a therapeutic strategy against AD.
Collapse
Affiliation(s)
| | - Luna Buitrago
- Departments of Neurology and Physiology/Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Herman Moreno
- Departments of Neurology and Physiology/Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Gloria Patricia Cardona-Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Calle 70, No. 52–21, Medellín, Colombia
| | - Gonzalo Arboleda
- Grupo de Muerte Celular, Instituto de Genética Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
112
|
Graham A. Mitochondrial regulation of macrophage cholesterol homeostasis. Free Radic Biol Med 2015; 89:982-92. [PMID: 26416507 DOI: 10.1016/j.freeradbiomed.2015.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/28/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022]
Abstract
This review explores the relationship between mitochondrial structure and function in the regulation of macrophage cholesterol metabolism and proposes that mitochondrial dysfunction contributes to loss of the elegant homeostatic mechanisms which normally maintain cellular sterol levels within defined limits. Mitochondrial sterol 27-hydroxylase (CYP27A1) can generate oxysterol activators of liver X receptors which heterodimerise with retinoid X receptors, enhancing the transcription of ATP binding cassette transporters (ABCA1, ABCG1, and ABCG4), that can remove excess cholesterol via efflux to apolipoproteins A-1, E, and high density lipoprotein, and inhibit inflammation. The activity of CYP27A1 is regulated by the rate of supply of cholesterol substrate to the inner mitochondrial membrane, mediated by a complex of proteins. The precise identity of this dynamic complex remains controversial, even in steroidogenic tissues, but may include steroidogenic acute regulatory protein and the 18 kDa translocator protein, together with voltage-dependent anion channels, ATPase AAA domain containing protein 3A, and optic atrophy type 1 proteins. Certainly, overexpression of StAR and TSPO proteins can enhance macrophage cholesterol efflux to apoA-I and/or HDL, while perturbations in mitochondrial function, or changes in the expression of mitochondrial fusion proteins, alter the efficiency of cholesterol efflux. Molecules which can sustain or improve mitochondrial function or increase the activity of the protein complex involved in cholesterol transfer may have utility in resolving the problem of dysregulated macrophage cholesterol homeostasis, a condition which may contribute to inflammation, atherosclerosis, nonalcoholic steatohepatitis, osteoblastic bone resorption, and some disorders of the central nervous system.
Collapse
Affiliation(s)
- Annette Graham
- Department of Life Sciences, School of Health and Life Sciences, and Institute for Applied Health Research, Glasgow Caledonian University, 70 Cowcaddens Road, Glasgow G4 0BA, United Kingdom.
| |
Collapse
|
113
|
Kata D, Földesi I, Feher LZ, Hackler L, Puskas LG, Gulya K. Rosuvastatin enhances anti-inflammatory and inhibits pro-inflammatory functions in cultured microglial cells. Neuroscience 2015; 314:47-63. [PMID: 26633263 DOI: 10.1016/j.neuroscience.2015.11.053] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 12/20/2022]
Abstract
Microglial activation results in profound morphological, functional and gene expression changes that affect the pro- and anti-inflammatory mechanisms of these cells. Although statins have beneficial effects on inflammation, they have not been thoroughly investigated for their ability to affect microglial functions. Therefore the effects of rosuvastatin, one of the most commonly prescribed drugs in cardiovascular therapy, either alone or in combination with bacterial lipopolysaccharide (LPS), were profiled in pure microglial cultures derived from the forebrains of 18-day-old rat embryos. To reveal the effects of rosuvastatin on a number of pro- and anti-inflammatory mechanisms, we performed morphometric, functional and gene expression studies relating to cell adhesion and proliferation, phagocytosis, pro- and anti-inflammatory cytokine (IL-1β, tumor necrosis factor α (TNF-α) and IL-10, respectively) production, and the expression of various inflammation-related genes, including those related to the above morphological parameters and cellular functions. We found that microglia could be an important therapeutic target of rosuvastatin. In unchallenged (control) microglia, rosuvastatin inhibited proliferation and cell adhesion, but promoted microspike formation and elevated the expression of certain anti-inflammatory genes (Cxcl1, Ccl5, Mbl2), while phagocytosis or pro- and anti-inflammatory cytokine production were unaffected. Moreover, rosuvastatin markedly inhibited microglial activation in LPS-challenged cells by affecting both their morphology and functions as it inhibited LPS-elicited phagocytosis and inhibited pro-inflammatory cytokine (IL-1β, TNF-α) production, concomitantly increasing the level of IL-10, an anti-inflammatory cytokine. Finally, rosuvastatin beneficially and differentially affected the expression of a number of inflammation-related genes in LPS-challenged cells by inhibiting numerous pro-inflammatory and stimulating several anti-inflammatory genes. Since the microglia could elicit pro-inflammatory responses leading to neurodegeneration, it is important to attenuate such mechanisms and promote anti-inflammatory properties, and develop prophylactic therapies. By beneficially regulating both pro- and anti-inflammatory microglial functions, rosuvastatin may be considered as a prophylactic agent in the prevention of inflammation-related neurological disorders.
Collapse
Affiliation(s)
- D Kata
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - I Földesi
- Department of Laboratory Medicine, University of Szeged, Szeged, Hungary
| | | | | | | | - K Gulya
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary.
| |
Collapse
|
114
|
Sandoval-Hernández AG, Hernández HG, Restrepo A, Muñoz JI, Bayon GF, Fernández AF, Fraga MF, Cardona-Gómez GP, Arboleda H, Arboleda GH. Liver X Receptor Agonist Modifies the DNA Methylation Profile of Synapse and Neurogenesis-Related Genes in the Triple Transgenic Mouse Model of Alzheimer's Disease. J Mol Neurosci 2015; 58:243-53. [PMID: 26553261 DOI: 10.1007/s12031-015-0665-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022]
Abstract
The liver X receptor agonist, GW3965, improves cognition in Alzheimer's disease (AD) mouse models. Here, we determined if short-term GW3965 treatment induces changes in the DNA methylation state of the hippocampus, which are associated with cognitive improvement. Twenty-four-month-old triple-transgenic AD (3xTg-AD) mice were treated with GW3965 (50 mg/kg/day for 6 days). DNA methylation state was examined by modified bisulfite conversion and hybridization on Illumina Infinium Methylation BeadChip 450 k arrays. The Morris water maze was used for behavioral analysis. Our results show in addition to improvement in cognition methylation changes in 39 of 13,715 interrogated probes in treated 3xTg-AD mice compared with untreated 3xTg-AD mice. These changes in methylation probes include 29 gene loci. Importantly, changes in methylation status were mainly from synapse-related genes (SYP, SYN1, and DLG3) and neurogenesis-associated genes (HMGB3 and RBBP7). Thus, our results indicate that liver X receptors (LXR) agonist treatment induces rapid changes in DNA methylation, particularly in loci associated with genes involved in neurogenesis and synaptic function. Our results suggest a new potential mechanism to explain the beneficial effect of GW3965.
Collapse
Affiliation(s)
- A G Sandoval-Hernández
- Grupo de Muerte Celular, Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
| | - H G Hernández
- Grupo de Neurociencias, Universidad Nacional, Bogotá, Colombia
| | - A Restrepo
- Grupo de Muerte Celular, Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
| | - J I Muñoz
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - G F Bayon
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, Spain
| | - A F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, Spain
| | - M F Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, Spain
| | - G P Cardona-Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - H Arboleda
- Grupo de Neurociencias, Universidad Nacional, Bogotá, Colombia
| | - Gonzalo H Arboleda
- Grupo de Muerte Celular, Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia.
- Departamento de Patología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
115
|
Lin CY, Vedin LL, Steffensen KR. The emerging roles of liver X receptors and their ligands in cancer. Expert Opin Ther Targets 2015; 20:61-71. [PMID: 26788589 DOI: 10.1517/14728222.2015.1081169] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Liver X receptors (LXRs) are nuclear receptors with well-known functions in cholesterol transport, fatty acid and glucose metabolism, and modulation of immune responses. Natural and synthetic ligands have been identified and are under development for the treatment of metabolic and inflammatory conditions and diseases. There is mounting evidence pointing to functional roles for LXRs in a variety of malignancies and the potential therapeutic efficacy of their ligands. AREAS COVERED This review summarizes the discovery and characterization of LXRs and their ligands, surveys their effects and mechanisms of action in cell-based and animal models of cancer, and proposes the future direction of basic and translational studies of LXRs and their ligands in cancer research and therapeutics. EXPERT OPINION Targeting LXRs is a promising strategy for cancer treatment, particularly for those cancers which do not have effective treatment options. Key questions remain, however, regarding the specific mechanisms of action, effects on other target cells within the tumor microenvironment, and receptor status in patient populations. Moreover, LXR ligands optimized for disease-specific functions and cancer-related endpoints are currently not available. These issues represent both challenges and significant opportunities for future research and development efforts.
Collapse
Affiliation(s)
- Chin-Yo Lin
- a 1 University of Houston, Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry , Houston, TX, USA
| | - Lise-Lotte Vedin
- b 2 Karolinska University Hospital Huddinge, Karolinska Institutet, Division of Clinical Chemistry, Department of Laboratory Medicine , SE-141 86 Stockholm, Sweden
| | - Knut R Steffensen
- b 2 Karolinska University Hospital Huddinge, Karolinska Institutet, Division of Clinical Chemistry, Department of Laboratory Medicine , SE-141 86 Stockholm, Sweden
| |
Collapse
|
116
|
Corona AW, Kodoma N, Casali BT, Landreth GE. ABCA1 is Necessary for Bexarotene-Mediated Clearance of Soluble Amyloid Beta from the Hippocampus of APP/PS1 Mice. J Neuroimmune Pharmacol 2015; 11:61-72. [PMID: 26175148 DOI: 10.1007/s11481-015-9627-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/07/2015] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (AD) is characterized by impaired clearance of amyloid beta (Aβ) peptides, leading to the accumulation of Aβ in the brain and subsequent neurodegeneration and cognitive impairment. ApoE plays a critical role in the proteolytic degradation of soluble forms of Aβ. This effect is dependent upon lipidation of ApoE by ABCA1-mediated transfer of phospholipids and cholesterol. ApoE and ABCA1 are induced by the action of the RXR agonist, bexarotene. We have previously shown that bexarotene reduces Aβ levels in AD mouse models and we have hypothesized that this effect requires ABCA1-mediated lipidation of ApoE. To test this hypothesis, we crossed ABCA1-deficient (ABCA1 KO) mice with the APP/PS1 model of AD. Aged ABCA1 WT and ABCA1 KO APP/PS1 mice were treated for 7 days with vehicle or bexarotene (100 mg/kg/day). Bexarotene reduced levels of soluble Aβ 1-40 and 1-42 in the hippocampus of ABCA1 WT but not ABCA1 KO APP/PS1 mice. In contrast, insoluble levels of Aβ, and plaque loads were unaffected by bexarotene in this study. ABCA1 KO mice had increased levels of inflammation compared with ABCA1 WT mice. Bexarotene also increased most inflammatory gene markers evaluated. The effect of bexarotene on microglial inflammatory profiles, however, was independent of ABCA1 genotype. Importantly, bexarotene ameliorated deficits in novel object recognition in ABCA1 WT but not ABCA1 KO APP/PS1 mice. These data indicate that ABCA1-induced lipidation of ApoE is necessary for the ability of bexarotene to clear hippocampal soluble Aβ and ameliorate cognitive deficits.
Collapse
Affiliation(s)
- Angela W Corona
- Department of Neurosciences School of Medicine, Case Western Reserve University, 2210 Circle Dr., Rm E649, Cleveland, OH, 44106-4928, USA
| | - Nathan Kodoma
- Department of Neurosciences School of Medicine, Case Western Reserve University, 2210 Circle Dr., Rm E649, Cleveland, OH, 44106-4928, USA
| | - Brad T Casali
- Department of Neurosciences School of Medicine, Case Western Reserve University, 2210 Circle Dr., Rm E649, Cleveland, OH, 44106-4928, USA
| | - Gary E Landreth
- Department of Neurosciences School of Medicine, Case Western Reserve University, 2210 Circle Dr., Rm E649, Cleveland, OH, 44106-4928, USA.
| |
Collapse
|
117
|
Skerrett R, Pellegrino MP, Casali BT, Taraboanta L, Landreth GE. Combined Liver X Receptor/Peroxisome Proliferator-activated Receptor γ Agonist Treatment Reduces Amyloid β Levels and Improves Behavior in Amyloid Precursor Protein/Presenilin 1 Mice. J Biol Chem 2015; 290:21591-602. [PMID: 26163517 DOI: 10.1074/jbc.m115.652008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is characterized by the extracellular accumulation of amyloid β (Aβ), which is accompanied by a robust inflammatory response in the brain. Both of these pathogenic processes are regulated by nuclear receptors, including the liver X receptors (LXRs) and peroxisome-proliferator receptor γ (PPARγ). Agonists of LXRs have been demonstrated previously to reduce Aβ levels and improve cognitive deficits in AD mouse models by inducing the transcription and lipidation of apolipoprotein E (apoE). Agonists targeting PPARγ reduce the microglial expression of proinflammatory genes and have also been shown to modulate apoE expression. Here we investigate whether a combination therapy with both LXR and PPARγ agonists results in increased benefits in an AD mouse model. We found that the LXR agonist GW3965 and the PPARγ agonist pioglitazone were individually able to increase the levels of apoE and related genes, decrease the expression of proinflammatory genes, and facilitate Aβ decreases in the hippocampus. Combined treatment with both agonists provoked a further increase in the expression of apoE and a decrease in the soluble and deposited forms of Aβ. The decrease in plaques was associated with increased colocalization between microglia and plaques. In addition, the PPARγ agonist in the combined treatment paradigm was able to counteract the elevation in plasma triglycerides that is a side effect of LXR agonist treatment. These results suggest that combined LXR/PPARγ agonist treatment merits further investigation for the treatment of AD.
Collapse
Affiliation(s)
- Rebecca Skerrett
- From the Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106 and
| | - Mateus P Pellegrino
- the School of Medicine, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
| | - Brad T Casali
- From the Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106 and
| | - Laura Taraboanta
- From the Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106 and
| | - Gary E Landreth
- From the Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106 and
| |
Collapse
|
118
|
Gamba P, Testa G, Gargiulo S, Staurenghi E, Poli G, Leonarduzzi G. Oxidized cholesterol as the driving force behind the development of Alzheimer's disease. Front Aging Neurosci 2015; 7:119. [PMID: 26150787 PMCID: PMC4473000 DOI: 10.3389/fnagi.2015.00119] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/03/2015] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD), the most common neurodegenerative disorder associated with dementia, is typified by the pathological accumulation of amyloid Aβ peptides and neurofibrillary tangles (NFT) within the brain. Considerable evidence indicates that many events contribute to AD progression, including oxidative stress, inflammation, and altered cholesterol metabolism. The brain’s high lipid content makes it particularly vulnerable to oxidative species, with the consequent enhancement of lipid peroxidation and cholesterol oxidation, and the subsequent formation of end products, mainly 4-hydroxynonenal and oxysterols, respectively from the two processes. The chronic inflammatory events observed in the AD brain include activation of microglia and astrocytes, together with enhancement of inflammatory molecule and free radical release. Along with glial cells, neurons themselves have been found to contribute to neuroinflammation in the AD brain, by serving as sources of inflammatory mediators. Oxidative stress is intimately associated with neuroinflammation, and a vicious circle has been found to connect oxidative stress and inflammation in AD. Alongside oxidative stress and inflammation, altered cholesterol metabolism and hypercholesterolemia also significantly contribute to neuronal damage and to progression of AD. Increasing evidence is now consolidating the hypothesis that oxidized cholesterol is the driving force behind the development of AD, and that oxysterols are the link connecting the disease to altered cholesterol metabolism in the brain and hypercholesterolemia; this is because of the ability of oxysterols, unlike cholesterol, to cross the blood brain barrier (BBB). The key role of oxysterols in AD pathogenesis has been strongly supported by research pointing to their involvement in modulating neuroinflammation, Aβ accumulation, and cell death. This review highlights the key role played by cholesterol and oxysterols in the brain in AD pathogenesis.
Collapse
Affiliation(s)
- Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin Orbassano, Torino, Italy
| |
Collapse
|
119
|
Savonenko AV, Melnikova T, Wang Y, Ravert H, Gao Y, Koppel J, Lee D, Pletnikova O, Cho E, Sayyida N, Hiatt A, Troncoso J, Davies P, Dannals RF, Pomper MG, Horti AG. Cannabinoid CB2 Receptors in a Mouse Model of Aβ Amyloidosis: Immunohistochemical Analysis and Suitability as a PET Biomarker of Neuroinflammation. PLoS One 2015; 10:e0129618. [PMID: 26086915 PMCID: PMC4472959 DOI: 10.1371/journal.pone.0129618] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 05/11/2015] [Indexed: 11/18/2022] Open
Abstract
In Alzheimer's disease (AD), one of the early responses to Aβ amyloidosis is recruitment of microglia to areas of new plaque. Microglial receptors such as cannabinoid receptor 2 (CB2) might be a suitable target for development of PET radiotracers that could serve as imaging biomarkers of Aβ-induced neuroinflammation. Mouse models of amyloidosis (J20APPswe/ind and APPswe/PS1ΔE9) were used to investigate the cellular distribution of CB2 receptors. Specificity of CB2 antibody (H60) was confirmed using J20APPswe/ind mice lacking CB2 receptors. APPswe/PS1ΔE9 mice were used in small animal PET with a CB2-targeting radiotracer, [11C]A836339. These studies revealed increased binding of [11C]A836339 in amyloid-bearing mice. Specificity of the PET signal was confirmed in a blockade study with a specific CB2 antagonist, AM630. Confocal microscopy revealed that CB2-receptor immunoreactivity was associated with astroglial (GFAP) and, predominantly, microglial (CD68) markers. CB2 receptors were observed, in particular, in microglial processes forming engulfment synapses with Aβ plaques. In contrast to glial cells, neuron (NeuN)-derived CB2 signal was equal between amyloid-bearing and control mice. The pattern of neuronal CB2 staining in amyloid-bearing mice was similar to that in human cases of AD. The data collected in this study indicate that Aβ amyloidosis without concomitant tau pathology is sufficient to activate CB2 receptors that are suitable as an imaging biomarker of neuroinflammation. The main source of enhanced CB2 PET binding in amyloid-bearing mice is increased CB2 immunoreactivity in activated microglia. The presence of CB2 immunoreactivity in neurons does not likely contribute to the enhanced CB2 PET signal in amyloid-bearing mice due to a lack of significant neuronal loss in this model. However, significant loss of neurons as seen at late stages of AD might decrease the CB2 PET signal due to loss of neuronally-derived CB2. Thus this study in mouse models of AD indicates that a CB2-specific radiotracer can be used as a biomarker of neuroinflammation in the early preclinical stages of AD, when no significant neuronal loss has yet developed.
Collapse
Affiliation(s)
- Alena V. Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Departments of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- * E-mail: (AGH); (AS)
| | - Tatiana Melnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yuchuan Wang
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Hayden Ravert
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yongjun Gao
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Jeremy Koppel
- Litwin-Zucker Research Center, Feinstein Institute for Medical Research, North-Shore Long Island Jewish Health System, Manhasset, NY, United States of America
| | - Deidre Lee
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Olga Pletnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Eugenia Cho
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Nuzhat Sayyida
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Andrew Hiatt
- MAPP Biopharmaceutical Inc, San-Diego, CA, United States of America
| | - Juan Troncoso
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Departments of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Peter Davies
- Litwin-Zucker Research Center, Feinstein Institute for Medical Research, North-Shore Long Island Jewish Health System, Manhasset, NY, United States of America
| | - Robert F. Dannals
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Andrew G. Horti
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- * E-mail: (AGH); (AS)
| |
Collapse
|
120
|
Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP. Neuroinflammation in Alzheimer's disease. Lancet Neurol 2015; 14:388-405. [PMID: 25792098 DOI: 10.1016/s1474-4422(15)70016-5] [Citation(s) in RCA: 3843] [Impact Index Per Article: 427.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing evidence suggests that Alzheimer's disease pathogenesis is not restricted to the neuronal compartment, but includes strong interactions with immunological mechanisms in the brain. Misfolded and aggregated proteins bind to pattern recognition receptors on microglia and astroglia, and trigger an innate immune response characterised by release of inflammatory mediators, which contribute to disease progression and severity. Genome-wide analysis suggests that several genes that increase the risk for sporadic Alzheimer's disease encode factors that regulate glial clearance of misfolded proteins and the inflammatory reaction. External factors, including systemic inflammation and obesity, are likely to interfere with immunological processes of the brain and further promote disease progression. Modulation of risk factors and targeting of these immune mechanisms could lead to future therapeutic or preventive strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Michael T Heneka
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany; German Center for Neurodegnerative Diseases (DZNE), Bonn, Germany.
| | - Monica J Carson
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California, Riverside, CA, USA
| | - Joseph El Khoury
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Gary E Landreth
- Alzheimer Research Laboratory, Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Andreas H Jacobs
- Department of Geriatrics, Johanniter Hospital, Bonn, Germany; European Institute for Molecular Imaging (EIMI) at the Westfalian Wilhelms University (WWU), Münster, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Javier Vitorica
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Cientificas Universidad de Sevilla, Sevilla, Spain
| | - Richard M Ransohoff
- Department of Neuroscience, Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Karl Herrup
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong
| | - Sally A Frautschy
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, the Geriatric, Research, and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Bente Finsen
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, Basque Foundation for Science (IKERBASQUE), Bilbao, Spain; Department of Neurosciences, University of the Basque Country UPV/EHU (Euskal Herriko Unibertsitatea/Universidad del País Vasco) and CIBERNED (Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas), Leioa, Spain
| | - Koji Yamanaka
- Research Institute of Environmental Medicine, Nagoya University/RIKEN Brain Science Institute, Wako-shi, Japan
| | - Jari Koistinaho
- Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Eicke Latz
- German Center for Neurodegnerative Diseases (DZNE), Bonn, Germany; Institute of Innate Immunity, University of Bonn, Bonn, Germany; Department of InfectiousDiseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Annett Halle
- Max-Planck Research Group Neuroimmunology, Center of Advanced European Studies and Research (CAESAR), Bonn, Germany
| | - Gabor C Petzold
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany; German Center for Neurodegnerative Diseases (DZNE), Bonn, Germany
| | - Terrence Town
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Dave Morgan
- Department of Molecular Pharmacology and Physiology, Byrd Alzheimer's Institute, University of South Florida College of Medicine, Tampa, FL, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - V Hugh Perry
- School of Biological Sciences, Southampton General Hospital, Southampton, UK
| | - Clive Holmes
- Clinical and Experimental Science, University of Southampton, Southampton, UK; Memory Assessment and Research Centre, Moorgreen Hospital, Southern Health Foundation Trust, Southampton, UK
| | - Nicolas G Bazan
- Louisiana State University Neuroscience Center of Excellence, Louisiana State University Health Sciences Center School of Medicine in New Orleans, LA, USA
| | - David J Brooks
- Division of Experimental Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Stéphane Hunot
- Centre National de la Recherche Scientifique (CNRS), UMR 7225, Experimental Therapeutics of Neurodegeneration, Paris, France
| | - Bertrand Joseph
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Nikolaus Deigendesch
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Olga Garaschuk
- Institute of Physiology II, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Erik Boddeke
- Department of Neuroscience, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | | | - John C Breitner
- Centre for Studies on Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, and the McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Greg M Cole
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, the Geriatric, Research, and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Douglas T Golenbock
- Department of InfectiousDiseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Markus P Kummer
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
121
|
Tai LM, Ghura S, Koster KP, Liakaite V, Maienschein‐Cline M, Kanabar P, Collins N, Ben‐Aissa M, Lei AZ, Bahroos N, Green SJ, Hendrickson B, Van Eldik LJ, LaDu MJ. APOE-modulated Aβ-induced neuroinflammation in Alzheimer's disease: current landscape, novel data, and future perspective. J Neurochem 2015; 133:465-88. [PMID: 25689586 PMCID: PMC4400246 DOI: 10.1111/jnc.13072] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 01/12/2023]
Abstract
Chronic glial activation and neuroinflammation induced by the amyloid-β peptide (Aβ) contribute to Alzheimer's disease (AD) pathology. APOE4 is the greatest AD-genetic risk factor; increasing risk up to 12-fold compared to APOE3, with APOE4-specific neuroinflammation an important component of this risk. This editorial review discusses the role of APOE in inflammation and AD, via a literature review, presentation of novel data on Aβ-induced neuroinflammation, and discussion of future research directions. The complexity of chronic neuroinflammation, including multiple detrimental and beneficial effects occurring in a temporal and cell-specific manner, has resulted in conflicting functional data for virtually every inflammatory mediator. Defining a neuroinflammatory phenotype (NIP) is one way to address this issue, focusing on profiling the changes in inflammatory mediator expression during disease progression. Although many studies have shown that APOE4 induces a detrimental NIP in peripheral inflammation and Aβ-independent neuroinflammation, data for APOE-modulated Aβ-induced neuroinflammation are surprisingly limited. We present data supporting the hypothesis that impaired apoE4 function modulates Aβ-induced effects on inflammatory receptor signaling, including amplification of detrimental (toll-like receptor 4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways. To ultimately develop APOE genotype-specific therapeutics, it is critical that future studies define the dynamic NIP profile and pathways that underlie APOE-modulated chronic neuroinflammation. In this editorial review, we present data supporting the hypothesis that impaired apoE4 function modulates Aβ-induced effects on inflammatory receptor signaling, including amplification of detrimental (TLR4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways, resulting in an adverse NIP that causes neuronal dysfunction. NIP, Neuroinflammatory phenotype; P.I., pro-inflammatory; A.I., anti-inflammatory.
Collapse
Affiliation(s)
- Leon M. Tai
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Shivesh Ghura
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Kevin P. Koster
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | | | | | - Pinal Kanabar
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | - Nicole Collins
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Manel Ben‐Aissa
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Arden Zhengdeng Lei
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | - Neil Bahroos
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | | | - Bill Hendrickson
- UIC Research Resources CenterUniversity of IllinoisChicagoIllinoisUSA
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| |
Collapse
|
122
|
Theofilopoulos S, Arenas E. Liver X receptors and cholesterol metabolism: role in ventral midbrain development and neurodegeneration. F1000PRIME REPORTS 2015; 7:37. [PMID: 26097711 PMCID: PMC4447034 DOI: 10.12703/p7-37] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of the ventral midbrain is orchestrated by a number of cell-extrinsic and -intrinsic factors that control critical processes, such as the patterning of the neural tube along the main body axis and the specification of diverse neuronal cell types in distinct positions of the neural tube. Subsequently, the regulation of neurogenesis and survival— acquire particular relevance in order to define the final size of diverse neuronal populations. In a series of studies during the last few years, we have identified liver X receptors (LXRs) as critical regulators of ventral midbrain development. Moreover, specific cholesterol derivatives present in the midbrain or in the cerebrospinal fluid were identified as LXR ligands, capable of specifically and selectively regulating neurogenesis and the survival of distinct neuronal populations, including midbrain dopamine neurons. These studies have shown that cholesterol derivatives are an entirely new class of factors capable of regulating both neuronal survival and neurogenesis, thus providing a direct link between cholesterol metabolism and brain development. In addition, LXRs and cholesterol metabolism were found to play a critical role in regulating the balance between neuronal survival and death in diverse midbrain neuronal populations. In this review, we will focus on these two aspects and on the possible role of cholesterol metabolism and LXRs in neurodegeneration.
Collapse
|
123
|
Abstract
Members of the nuclear receptor superfamily of ligand-dependent transcription factors carry out vital cellular functions and are highly druggable therapeutic targets. Liver X receptors (LXRs) are nuclear receptor family members that function in cholesterol transport, glucose metabolism and the modulation of inflammatory responses. There is now accumulating evidence to support the involvement of LXRs in a variety of malignancies and the potential efficacy of their ligands in these diseases. This Review summarizes the discovery and characterization of LXRs and their ligands, their effects and mechanisms in preclinical cancer models, and the future directions of basic and translational LXR research in cancer therapeutics.
Collapse
Affiliation(s)
- Chin-Yo Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5056, USA
| | - Jan-Åke Gustafsson
- 1] Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5056, USA. [2] Department of Biosciences and Nutrition at NOVUM, Karolinska Institutet, Huddinge SE-141 83, Sweden
| |
Collapse
|
124
|
Abstract
Dyslipidaemia is a major risk factor for cardiovascular diseases. Pharmacological lowering of LDL-C levels using statins reduces cardiovascular risk. However, a substantial residual risk persists especially in patients with type 2 diabetes mellitus. Because of the inverse association observed in epidemiological studies of HDL-C with the risk for cardiovascular diseases, novel therapeutic strategies to raise HDL-C levels or improve HDL functionality are developed as complementary therapy for cardiovascular diseases. However, until now most therapies targeting HDL-C levels failed in clinical trials because of side effects or absence of clinical benefits. This chapter will highlight the emerging small molecules currently developed and tested in clinical trials to pharmacologically modulate HDL-C and functionality including new CETP inhibitors (anacetrapib, evacetrapib), novel PPAR agonists (K-877, CER-002, DSP-8658, INT131 and GFT505), LXR agonists (ATI-111, LXR-623, XL-652) and RVX-208.
Collapse
|
125
|
Gooshe M, Abdolghaffari AH, Gambuzza ME, Rezaei N. The role of Toll-like receptors in multiple sclerosis and possible targeting for therapeutic purposes. Rev Neurosci 2014; 25:713-39. [PMID: 24914714 DOI: 10.1515/revneuro-2014-0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023]
Abstract
The interaction between the immune and nervous systems suggests invaluable mechanisms for several pathological conditions, especially neurodegenerative disorders. Multiple sclerosis (MS) is a potentially disabling chronic autoimmune disease, characterized by chronic inflammation and neurodegenerative pathology of the central nervous system. Toll-like receptors (TLRs) are an important family of receptors involved in host defense and in recognition of invading pathogens. The role of TLRs in the pathogenesis of autoimmune disorders such as MS is only starting to be uncovered. Recent studies suggest an ameliorative role of TLR3 and a detrimental role of other TLRs in the onset and progression of MS and experimental autoimmune encephalomyelitis, a murine model of MS. Thus, modulating TLRs can represent an innovative immunotherapeutic approach in MS therapy. This article outlines the role of these TLRs in MS, also discussing TLR-targeted agonist or antagonists that could be used in the different stages of the disease.
Collapse
|
126
|
Skerrett R, Malm T, Landreth G. Nuclear receptors in neurodegenerative diseases. Neurobiol Dis 2014; 72 Pt A:104-16. [PMID: 24874548 PMCID: PMC4246019 DOI: 10.1016/j.nbd.2014.05.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/14/2014] [Accepted: 05/17/2014] [Indexed: 01/04/2023] Open
Abstract
Nuclear receptors have generated substantial interest in the past decade as potential therapeutic targets for the treatment of neurodegenerative disorders. Despite years of effort, effective treatments for progressive neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and ALS remain elusive, making non-classical drug targets such as nuclear receptors an attractive alternative. A substantial literature in mouse models of disease and several clinical trials have investigated the role of nuclear receptors in various neurodegenerative disorders, most prominently AD. These studies have met with mixed results, yet the majority of studies in mouse models report positive outcomes. The mechanisms by which nuclear receptor agonists affect disease pathology remain unclear. Deciphering the complex signaling underlying nuclear receptor action in neurodegenerative diseases is essential for understanding this variability in preclinical studies, and for the successful translation of nuclear receptor agonists into clinical therapies.
Collapse
Affiliation(s)
- Rebecca Skerrett
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Tarja Malm
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; A.I. Virtanen Institute for Molecular Sciences, Department of Neurobiology, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
127
|
Koldamova R, Fitz NF, Lefterov I. ATP-binding cassette transporter A1: from metabolism to neurodegeneration. Neurobiol Dis 2014; 72 Pt A:13-21. [PMID: 24844148 PMCID: PMC4302328 DOI: 10.1016/j.nbd.2014.05.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/01/2014] [Accepted: 05/06/2014] [Indexed: 01/04/2023] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol efflux to lipid-free apolipoprotein A-I (apoA-I) and apolipoprotein E (apoE). ABCA1 is an essential regulator of high density lipoproteins (HDL) and reverse cholesterol transport - a role that determines its importance for atherosclerosis. Over the last 10 years studies have provided convincing evidence that ABCA1, via its control of apoE lipidation, also has a role in Alzheimer's disease (AD). A series of reports have revealed a significant impact of ABCA1 on Aβ deposition and clearance in AD model mice, as well as an association of common and rare ABCA1 gene variants with the risk for AD. Since APOE is the major genetic risk factor for late onset AD, the regulation of apoE level or its functionality by ABCA1 may prove significant for AD pathogenesis. ABCA1 is transcriptionally regulated by Liver X Receptors (LXR) and Retinoic X Receptors (RXR) which provides a starting point for drug discovery and development of synthetic LXR and RXR agonists for treatment of metabolic and neurodegenerative disorders. This review summarizes the recent results of research on ABCA1, particularly relevant to atherosclerosis and AD.
Collapse
Affiliation(s)
- Radosveta Koldamova
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Nicholas F Fitz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Iliya Lefterov
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
128
|
Tai LM, Koster KP, Luo J, Lee SH, Wang YT, Collins NC, Ben Aissa M, Thatcher GRJ, LaDu MJ. Amyloid-β pathology and APOE genotype modulate retinoid X receptor agonist activity in vivo. J Biol Chem 2014; 289:30538-30555. [PMID: 25217640 DOI: 10.1074/jbc.m114.600833] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous data demonstrate that bexarotene (Bex), retinoid X receptor (RXR) agonist, reduces soluble and insoluble amyloid-β (Aβ) in Alzheimer disease (AD)-transgenic mice either by increasing the levels of mouse apolipoprotein E (apoE) or increasing ABCA1/ABCG1-induced apoE lipoprotein association/lipidation. However, although the mechanism of action of RXR agonists remains unclear, a major concern for their use is human (h)-APOE4, the greatest AD genetic risk factor. If APOE4 imparts a toxic gain-of-function, then increasing apoE4 may increase soluble Aβ, likely the proximal AD neurotoxin. If the APOE4 loss-of-function is lipidation of apoE4, then induction of ABCA1/ABCG1 may be beneficial. In novel EFAD-Tg mice (overexpressing h-Aβ42 with h-APOE), levels of soluble Aβ (Aβ42 and oligomeric Aβ) are highest in E4FAD hippocampus (HP) > E3FAD-HP > E4FAD cortex (CX) > E3FAD-CX, whereas levels of lipoprotein-associated/lipidated apoE have the opposite pattern (6 months). In E4FAD-HP, short-term RXR agonist treatment (Bex or LG100268; 5.75-6 months) increased ABCA1, apoE4 lipoprotein-association/lipidation, and apoE4/Aβ complex, decreased soluble Aβ, and increased PSD95. In addition, hydrogel delivery, which mimics low sustained release, was equally effective as gavage for Bex and LG100268. RXR agonists induced no beneficial effects in the E4FAD-HP in a prevention protocol (5-6 months) and actually increased soluble Aβ levels in E3FAD-CX and E4FAD-CX with the short-term protocol, possibly the result of systemic hepatomegaly. Thus, RXR agonists address the loss-of-function associated with APOE4 and exacerbated by Aβ pathology, i.e. low levels of apoE4 lipoprotein association/lipidation. Further studies are vital to address whether RXR agonists are an APOE4-specific AD therapeutic and the systemic side effects that limit translational application.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jia Luo
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and University of Illinois at Chicago, Chicago, Illinois 60612
| | - Sue H Lee
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and University of Illinois at Chicago, Chicago, Illinois 60612
| | - Yue-Ting Wang
- UICentre (Drug Discovery at UIC), University of Illinois at Chicago, Chicago, Illinois 60612
| | - Nicole C Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Manel Ben Aissa
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and University of Illinois at Chicago, Chicago, Illinois 60612
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612.
| |
Collapse
|
129
|
Gadde S, Even-Or O, Kamaly N, Hasija A, Gagnon PG, Adusumilli KH, Erakovic A, Pal AK, Zhang XQ, Kolishetti N, Shi J, Fisher EA, Farokhzad OC. Development of therapeutic polymeric nanoparticles for the resolution of inflammation. Adv Healthc Mater 2014; 3:1448-1456. [PMID: 24659608 DOI: 10.1002/adhm.201300688] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/19/2014] [Indexed: 11/09/2022]
Abstract
Liver X receptors (LXRs) attenuate inflammation by modulating the expression of key inflammatory genes, making LXRs and their ligands particularly attractive candidates for therapeutic intervention in cardiovascular, metabolic, and/or inflammatory diseases. Herein, enhanced proresolving activity of polymeric nanoparticles (NPs) containing the synthetic LXR agonist GW3965 (LXR-NPs) is demonstrated, developed from a combinatorial library of more than 70 formulations with variations in critical physicochemical parameters. In vitro studies on peritoneal macrophages confirm that LXR-NPs are significantly more effective than the free agonist at downregulating pro-inflammatory mediators (MCP-1 and TNFα), as well as inducing the expression of LXR target genes (ABCA1 and SREBP1c). Through a zymosan-induced acute peritonitis in vivo model, LXR-NPs are found to be more efficient than free GW3965 at limiting the recruitment of polymononuclear neutrophils (50% vs 17%), suppressing the gene expression and secretion of pro-inflammatory factors MCP-1 and TNFα in peritoneal macrophages, and decreasing the resolution interval up to 4 h. Furthermore, LXR-NPs suppress the secretion of MCP-1 and TNFα by monocytes and macrophages more efficiently than the commercial drug dexamethasone. Overall, these findings demonstrate that LXR-NPs are capable of promoting resolution of inflammation and highlight the prospect of LXR-based nanotherapeutics for inflammatory diseases.
Collapse
Affiliation(s)
- Suresh Gadde
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Orli Even-Or
- Department of Cell Biology and the Leon H. Charney, Division of Cardiology, Department of Medicine, New York University School of Medicine; New York NY 10016 USA
| | - Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Apoorva Hasija
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Philippe G. Gagnon
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Krishna H. Adusumilli
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Andrea Erakovic
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Anoop K. Pal
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Xue-Qing Zhang
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Nagesh Kolishetti
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Jinjun Shi
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
| | - Edward A. Fisher
- Department of Cell Biology and the Leon H. Charney, Division of Cardiology, Department of Medicine, New York University School of Medicine; New York NY 10016 USA
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School; Boston MA 02115 USA
- King Abdulaziz University; Jeddah 21589 Saudi Arabia
| |
Collapse
|
130
|
Malek G, Lad EM. Emerging roles for nuclear receptors in the pathogenesis of age-related macular degeneration. Cell Mol Life Sci 2014; 71:4617-36. [PMID: 25156067 DOI: 10.1007/s00018-014-1709-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/14/2014] [Accepted: 08/18/2014] [Indexed: 12/20/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in the elderly in the Western world. Over the last 30 years, our understanding of the pathogenesis of the disease has grown exponentially thanks to the results of countless epidemiology, genetic, histological, and biochemical studies. This information, in turn, has led to the identification of multiple biologic pathways potentially involved in development and progression of AMD, including but not limited to inflammation, lipid and extracellular matrix dysregulation, and angiogenesis. Nuclear receptors are a superfamily of transcription factors that have been shown to regulate many of the pathogenic pathways linked with AMD and as such they are emerging as promising targets for therapeutic intervention. In this review, we will present the fundamental phenotypic features of AMD and discuss our current understanding of the pathobiological disease mechanisms. We will introduce the nuclear receptor superfamily and discuss the current literature on their effects on AMD-related pathophysiology.
Collapse
Affiliation(s)
- Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, AERI Room 4006, Durham, NC, 27710, USA,
| | | |
Collapse
|
131
|
Hottman DA, Chernick D, Cheng S, Wang Z, Li L. HDL and cognition in neurodegenerative disorders. Neurobiol Dis 2014; 72 Pt A:22-36. [PMID: 25131449 DOI: 10.1016/j.nbd.2014.07.015] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 06/26/2014] [Accepted: 07/28/2014] [Indexed: 12/12/2022] Open
Abstract
High-density lipoproteins (HDLs) are a heterogeneous group of lipoproteins composed of various lipids and proteins. HDL is formed both in the systemic circulation and in the brain. In addition to being a crucial player in the reverse cholesterol transport pathway, HDL possesses a wide range of other functions including anti-oxidation, anti-inflammation, pro-endothelial function, anti-thrombosis, and modulation of immune function. It has been firmly established that high plasma levels of HDL protect against cardiovascular disease. Accumulating evidence indicates that the beneficial role of HDL extends to many other systems including the central nervous system. Cognition is a complex brain function that includes all aspects of perception, thought, and memory. Cognitive function often declines during aging and this decline manifests as cognitive impairment/dementia in age-related and progressive neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. A growing concern is that no effective therapy is currently available to prevent or treat these devastating diseases. Emerging evidence suggests that HDL may play a pivotal role in preserving cognitive function under normal and pathological conditions. This review attempts to summarize recent genetic, clinical and experimental evidence for the impact of HDL on cognition in aging and in neurodegenerative disorders as well as the potential of HDL-enhancing approaches to improve cognitive function.
Collapse
Affiliation(s)
- David A Hottman
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dustin Chernick
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Shaowu Cheng
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhe Wang
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
132
|
Abstract
Blood cholesterol levels are not consistently elevated in subjects with age-related cognitive decline, although epidemiological studies suggest that Alzheimer's disease and cardiovascular diseases share common risk factors. These include the presence of an unusual genetic variant, the APOE4 (apolipoprotein E4) allele, which modulates LDL (low-density lipoproteins) metabolism, increases free radical formation and reduces plasma antioxidant concentrations. Together, these risk factors support a mechanism for increased LDL circulation time and free radical modification of LDL. Plasma oxycholesterols, hydroxylated metabolites of cholesterol, are carried by oxidized LDL, and elevated lipids in mid-life are associated with increased long-term risk of dementia. Although brain cholesterol metabolism is segregated from the systemic circulation, during oxidative stress, plasma oxycholesterols could have damaging effects on BBB (blood–brain barrier) function and consequently on neuronal cells. Cholesterol-lowering drugs such as statins may prevent the modifications to LDL in mid-life and might show beneficial effects in later life.
Collapse
|
133
|
Irwin RW, Solinsky CM, Brinton RD. Frontiers in therapeutic development of allopregnanolone for Alzheimer's disease and other neurological disorders. Front Cell Neurosci 2014; 8:203. [PMID: 25126056 PMCID: PMC4115668 DOI: 10.3389/fncel.2014.00203] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/04/2014] [Indexed: 01/08/2023] Open
Abstract
Allopregnanolone (Allo), a neurosteroid, has emerged as a promising promoter of endogenous regeneration in brain. In a mouse model of Alzheimer’s disease, Allo induced neurogenesis, oligodendrogenesis, white matter generation and cholesterol homeostasis while simultaneously reducing β-amyloid and neuroinflammatory burden. Allo activates signaling pathways and gene expression required for regeneration of neural stem cells and their differentiation into neurons. In parallel, Allo activates systems to sustain cholesterol homeostasis and reduce β-amyloid generation. To advance Allo into studies for chronic human neurological conditions, we examined translational and clinical parameters: dose, regimen, route, formulation, outcome measures, and safety regulations. A treatment regimen of once per week at sub-sedative doses of Allo was optimal for regeneration and reduction in Alzheimer’s pathology. This regimen had a high safety profile following chronic exposure in aged normal and Alzheimer’s mice. Formulation of Allo for multiple routes of administration has been developed for both preclinical and clinical testing. Preclinical evidence for therapeutic efficacy of Allo spans multiple neurological diseases including Alzheimer’s, Parkinson’s, multiple sclerosis, Niemann-Pick, diabetic neuropathy, status epilepticus, and traumatic brain injury. To successfully translate Allo as a therapeutic for multiple neurological disorders, it will be necessary to tailor dose and regimen to the targeted therapeutic mechanisms and disease etiology. Treatment paradigms conducted in accelerated disease models in young animals have a low probability of successful translation to chronic diseases in adult and aged humans. Gender, genetic risks, stage and burden of disease are critical determinants of efficacy. This review focuses on recent advances in development of Allo for Alzheimer’s disease (AD) that have the potential to accelerate therapeutic translation for multiple unmet neurological needs.
Collapse
Affiliation(s)
- Ronald W Irwin
- Department of Pharmacology and Pharmaceutical Sciences, Pharmaceutical Sciences Center, School of Pharmacy, University of Southern California Los Angeles, CA, USA
| | - Christine M Solinsky
- Clinical and Experimental Therapeutics Program, School of Pharmacy, University of Southern California Los Angeles, CA, USA
| | - Roberta Diaz Brinton
- Department of Pharmacology and Pharmaceutical Sciences, Pharmaceutical Sciences Center, School of Pharmacy, University of Southern California Los Angeles, CA, USA ; Department of Neurology, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
134
|
Anti-ApoE antibody given after plaque onset decreases Aβ accumulation and improves brain function in a mouse model of Aβ amyloidosis. J Neurosci 2014; 34:7281-92. [PMID: 24849360 DOI: 10.1523/jneurosci.0646-14.2014] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Apolipoprotein E (apoE) is the strongest known genetic risk factor for late onset Alzheimer's disease (AD). It influences amyloid-β (Aβ) clearance and aggregation, which likely contributes in large part to its role in AD pathogenesis. We recently found that HJ6.3, a monoclonal antibody against apoE, significantly reduced Aβ plaque load when given to APPswe/PS1ΔE9 (APP/PS1) mice starting before the onset of plaque deposition. To determine whether the anti-apoE antibody HJ6.3 affects Aβ plaques, neuronal network function, and behavior in APP/PS1 mice after plaque onset, we administered HJ6.3 (10 mg/kg/week) or PBS intraperitoneally to 7-month-old APP/PS1 mice for 21 weeks. HJ6.3 mildly improved spatial learning performance in the water maze, restored resting-state functional connectivity, and modestly reduced brain Aβ plaque load. There was no effect of HJ6.3 on total plasma cholesterol or cerebral amyloid angiopathy. To investigate the underlying mechanisms of anti-apoE immunotherapy, HJ6.3 was applied to the brain cortical surface and amyloid deposition was followed over 2 weeks using in vivo imaging. Acute exposure to HJ6.3 affected the course of amyloid deposition in that it prevented the formation of new amyloid deposits, limited their growth, and was associated with occasional clearance of plaques, a process likely associated with direct binding to amyloid aggregates. Topical application of HJ6.3 for only 14 d also decreased the density of amyloid plaques assessed postmortem. Collectively, these studies suggest that anti-apoE antibodies have therapeutic potential when given before or after the onset of Aβ pathology.
Collapse
|
135
|
Sodhi RK, Singh N. Liver X receptor agonist T0901317 reduces neuropathological changes and improves memory in mouse models of experimental dementia. Eur J Pharmacol 2014; 732:50-9. [DOI: 10.1016/j.ejphar.2014.03.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 03/13/2014] [Accepted: 03/20/2014] [Indexed: 01/08/2023]
|
136
|
Sallam T, Ito A, Rong X, Kim J, van Stijn C, Chamberlain BT, Jung ME, Chao LC, Jones M, Gilliland T, Wu X, Su GL, Tangirala RK, Tontonoz P, Hong C. The macrophage LBP gene is an LXR target that promotes macrophage survival and atherosclerosis. J Lipid Res 2014; 55:1120-30. [PMID: 24671012 PMCID: PMC4031943 DOI: 10.1194/jlr.m047548] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/22/2014] [Indexed: 01/25/2023] Open
Abstract
The liver X receptors (LXRs) are members of the nuclear receptor superfamily that regulate sterol metabolism and inflammation. We sought to identify previously unknown genes regulated by LXRs in macrophages and to determine their contribution to atherogenesis. Here we characterize a novel LXR target gene, the lipopolysaccharide binding protein (LBP) gene. Surprisingly, the ability of LXRs to control LBP expression is cell-type specific, occurring in macrophages but not liver. Treatment of macrophages with oxysterols or loading with modified LDL induces LBP in an LXR-dependent manner, suggesting a potential role for LBP in the cellular response to cholesterol overload. To investigate this further, we performed bone marrow transplant studies. After 18 weeks of Western diet feeding, atherosclerotic lesion burden was assessed revealing markedly smaller lesions in the LBP(-/-) recipients. Furthermore, loss of bone marrow LBP expression increased apoptosis in atherosclerotic lesions as determined by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Supporting in vitro studies with isolated macrophages showed that LBP expression does not affect cholesterol efflux but promotes the survival of macrophages in the setting of cholesterol loading. The LBP gene is a macrophage-specific LXR target that promotes foam cell survival and atherogenesis.
Collapse
Affiliation(s)
- Tamer Sallam
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| | - Ayaka Ito
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| | - Xin Rong
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| | - Jason Kim
- Department of Medicine, Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA
| | - Caroline van Stijn
- Department of Medicine, Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA
| | - Brian T Chamberlain
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA
| | - Michael E Jung
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA
| | - Lily C Chao
- Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA
| | - Marius Jones
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| | - Thomas Gilliland
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| | - XiaoHui Wu
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Grace L Su
- Medical Service, Department of Veterans Affairs Medical Center, Ann Arbor, MI Department of Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Rajendra K Tangirala
- Department of Medicine, Division of Endocrinology, University of California, Los Angeles, Los Angeles, CA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| | - Cynthia Hong
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
137
|
Tice CM, Noto PB, Fan KY, Zhuang L, Lala DS, Singh SB. The Medicinal Chemistry of Liver X Receptor (LXR) Modulators. J Med Chem 2014; 57:7182-205. [PMID: 24832115 DOI: 10.1021/jm500442z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Colin M. Tice
- Vitae Pharmaceuticals Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Paul B. Noto
- Vitae Pharmaceuticals Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Kristi Yi Fan
- Vitae Pharmaceuticals Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Linghang Zhuang
- Vitae Pharmaceuticals Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Deepak S. Lala
- Vitae Pharmaceuticals Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Suresh B. Singh
- Vitae Pharmaceuticals Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| |
Collapse
|
138
|
Hong C, Tontonoz P. Liver X receptors in lipid metabolism: opportunities for drug discovery. Nat Rev Drug Discov 2014; 13:433-44. [DOI: 10.1038/nrd4280] [Citation(s) in RCA: 401] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
139
|
Wong BX, Hung YH, Bush AI, Duce JA. Metals and cholesterol: two sides of the same coin in Alzheimer's disease pathology. Front Aging Neurosci 2014; 6:91. [PMID: 24860500 PMCID: PMC4030154 DOI: 10.3389/fnagi.2014.00091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/28/2014] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease. It begins years prior to the onset of clinical symptoms, such as memory loss and cognitive decline. Pathological hallmarks of AD include the accumulation of β-amyloid in plaques and hyperphosphorylated tau in neurofibrillary tangles. Copper, iron, and zinc are abnormally accumulated and distributed in the aging brain. These metal ions can adversely contribute to the progression of AD. Dysregulation of cholesterol metabolism has also been implicated in the development of AD pathology. To date, large bodies of research have been carried out independently to elucidate the role of metals or cholesterol on AD pathology. Interestingly, metals and cholesterol affect parallel molecular and biochemical pathways involved in AD pathology. The possible links between metal dyshomeostasis and altered brain cholesterol metabolism in AD are reviewed.
Collapse
Affiliation(s)
- Bruce X Wong
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Ya Hui Hung
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Ashley I Bush
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - James A Duce
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia ; School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds Leeds, North Yorkshire, UK
| |
Collapse
|
140
|
Testa G, Gamba P, Badilli U, Gargiulo S, Maina M, Guina T, Calfapietra S, Biasi F, Cavalli R, Poli G, Leonarduzzi G. Loading into nanoparticles improves quercetin's efficacy in preventing neuroinflammation induced by oxysterols. PLoS One 2014; 9:e96795. [PMID: 24802026 PMCID: PMC4011877 DOI: 10.1371/journal.pone.0096795] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/11/2014] [Indexed: 12/31/2022] Open
Abstract
Chronic inflammatory events appear to play a fundamental role in Alzheimer's disease (AD)-related neuropathological changes, and to result in neuronal dysfunction and death. The inflammatory responses observed in the AD brain include activation and proliferation of glial cells, together with up-regulation of inflammatory mediators and of free radicals. Along with glial cells, neurons themselves can also react and contribute to neuroinflammatory changes in the AD brain, by serving as sources of inflammatory mediators. Because excess cholesterol cannot be degraded in the brain, it must be excreted from that organ as cholesterol oxidation products (oxysterols), in order to prevent its accumulation. Among risk factors for this neurodegenerative disease, a mechanistic link between altered cholesterol metabolism and AD has been suggested; oxysterols appear to be the missing linkers between the two, because of their neurotoxic effects. This study shows that 24-hydroxycholesterol, 27-hydroxycholesterol, and 7β-hydroxycholesterol, the three oxysterols potentially implicated in AD pathogenesis, induce some pro-inflammatory mediator expression in human neuroblastoma SH-SY5Y cells, via Toll-like receptor-4/cyclooxygenase-2/membrane bound prostaglandin E synthase (TLR4/COX-2/mPGES-1); this clearly indicates that oxysterols may promote neuroinflammatory changes in AD. To confirm this evidence, cells were incubated with the anti-inflammatory flavonoid quercetin; remarkably, its anti-inflammatory effects in SH-SY5Y cells were enhanced when it was loaded into β-cyclodextrin-dodecylcarbonate nanoparticles, versus cells pretreated with free quercetin. The goal of loading quercetin into nanoparticles was to improve its permeation across the blood-brain barrier into the brain, and its bioavailability to reach target cells. The findings show that this drug delivery system might be a new therapeutic strategy for preventing or reducing AD progression.
Collapse
Affiliation(s)
- Gabriella Testa
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Ulya Badilli
- Department of Pharmaceutical Technology, University of Ankara, Ankara, Turkey
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Marco Maina
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Tina Guina
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Simone Calfapietra
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
- * E-mail:
| |
Collapse
|
141
|
Yang H, Zheng S, Qiu Y, Yang Y, Wang C, Yang P, Li Q, Lei B. Activation of liver X receptor alleviates ocular inflammation in experimental autoimmune uveitis. Invest Ophthalmol Vis Sci 2014; 55:2795-804. [PMID: 24699378 DOI: 10.1167/iovs.13-13323] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE To investigate whether a synthetic LXR agonist TO901317 (TO90) ameliorates ocular inflammation in a mouse model of experimental autoimmune uveitis (EAU) and to explore its underlying mechanism. METHODS EAU was induced with subcutaneous injection of IRBP161-180 peptide (SGIPYIISYLHPGNTILHVD) in B10.RIII mice. TO90 (50 mg/kg/d) or vehicle was administrated orally for successive 16 days or 8 days as prevention or effector phase, respectively. The severity of EAU was evaluated with clinical and histological scores. The levels of LXRs, NF-κB subunit p65, and an LXR target gene ABCA1 in the retina were detected with real-time PCR and Western blotting. The expressions of proinflammatory genes, including TNF-α, IL-1β, IL-6, MCP-1, IFN-γ, and IL-17, were detected by real-time PCR. IRBP-specific lymphocyte proliferation was detected by MTT. Intracellular IFN-γ and IL-17 in CD4(+) T cells were measured by flow cytometry. RESULTS We found both LXRα and LXRβ were expressed in mouse retina. After administering TO90 orally to B10.RIII mice, the expression of LXRα but not LXRβ was upregulated in the naïve mice. Compared with naïve mice, LXRα expression was increased in vehicle and TO90-treated EAU mice, but the LXRβ expression was unchanged. The protein level of ABCA1 was enhanced in TO90-treated naïve and EAU mice but was unchanged in vehicle-treated EAU mice, suggesting activation of LXRα by TO90 is ligand dependent. TO90-mediated activation of LXRα improved the clinical and morphological scores in EAU mice. Meanwhile, activation of LXRα decreased the expressions of proinflammatory cytokines, including TNF-α, IL-1β, IL-6, MCP-1, IFN-γ, and IL-17 in the retina. TO90 treatment inhibited IRBP-specific immune responses. The proportions of Th1 and Th17 expressing IFN-γ and IL-17 were reduced in TO90-treated EAU mice in both prevention and effector phases. Furthermore, TO90 significantly downregulated the expressions of an NF-κB subunit p65 at the protein and mRNA levels. CONCLUSIONS TO90 activates LXRα and potently attenuates ocular inflammation in EAU. Alleviation of ocular inflammation could partially result from inhibition of the NF-κB signaling pathway. TO90 reduces IFN-γ and IL-17 expression in both prevention and treatment scenarios. Our data suggest that the LXR agonist may become a novel class of therapeutic agent for autoimmune uveitis.
Collapse
Affiliation(s)
- Hongxia Yang
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Guillot-Sestier MV, Town T. Innate immunity in Alzheimer's disease: a complex affair. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:593-607. [PMID: 23574177 DOI: 10.2174/1871527311312050008] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by three major histopathological hallmarks: β-amyloid deposits, neurofibrillary tangles and gliosis. While neglected for decades, the neuroinflammatory processes coordinated by microglia are now accepted as etiologic events in AD evolution. Microglial cells are found in close vicinity to amyloid plaques and display various activation phenotypes determined by the expression of a wide range of cytokines, chemokines, and innate immune surface receptors. During the development of AD pathology, microglia fail to restrict amyloid plaques and may contribute to neurotoxicity and cognitive deficit. Nevertheless, under specific activation states, microglia can participate in cerebral amyloid clearance. This review focuses on the complex relationship between microglia and Aβ pathology, and highlights both deleterious and beneficial roles of microglial activation states in the context of AD. A deeper understanding of microglial biology will hopefully pave the way for next-generation AD therapeutic approaches aimed at harnessing these enigmatic innate immune cells of the central nervous system.
Collapse
Affiliation(s)
- Marie-Victoire Guillot-Sestier
- Regenerative Medicine Institute Neural Program, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Steven Spielberg Building Room 345, Los Angeles, CA 90048, USA
| | | |
Collapse
|
143
|
Huang C. Natural modulators of liver X receptors. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2014; 12:76-85. [DOI: 10.1016/s2095-4964(14)60013-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
144
|
El Kharrassi Y, Samadi M, Lopez T, Nury T, El Kebbaj R, Andreoletti P, El Hajj HI, Vamecq J, Moustaid K, Latruffe N, El Kebbaj MS, Masson D, Lizard G, Nasser B, Cherkaoui-Malki M. Biological activities of Schottenol and Spinasterol, two natural phytosterols present in argan oil and in cactus pear seed oil, on murine miroglial BV2 cells. Biochem Biophys Res Commun 2014; 446:798-804. [PMID: 24582563 DOI: 10.1016/j.bbrc.2014.02.074] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/18/2014] [Indexed: 12/17/2022]
Abstract
The objective of this study was to evaluate the biological activities of the major phytosterols present in argan oil (AO) and in cactus seed oil (CSO) in BV2 microglial cells. Accordingly, we first determined the sterol composition of AO and CSO, showing the presence of Schottenol and Spinasterol as major sterols in AO. While in CSO, in addition to these two sterols, we found mainly another sterol, the Sitosterol. The chemical synthesis of Schottenol and Spinasterol was performed. Our results showed that these two phytosterols, as well as sterol extracts from AO or CSO, are not toxic to microglial BV2 cells. However, treatments by these phytosterols impact the mitochondrial membrane potential. Furthermore, both Schottenol and Spinasterol can modulate the gene expression of two nuclear receptors, liver X receptor (LXR)-α and LXRβ, their target genes ABCA1 and ABCG1. Nonetheless, only Schottenol exhibited a differential activation vis-à-vis the nuclear receptor LXRβ. Thus Schottenol and Spinasterol can be considered as new LXR agonists, which may play protective roles by the modulation of cholesterol metabolism.
Collapse
Affiliation(s)
- Youssef El Kharrassi
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France; Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26000 Settat, Morocco
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Department of Chemistry, Université de Lorraine, Metz, France
| | | | - Thomas Nury
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France
| | - Riad El Kebbaj
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France; Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26000 Settat, Morocco
| | - Pierre Andreoletti
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France
| | - Hammam I El Hajj
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France
| | - Joseph Vamecq
- INSERM and HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Khadija Moustaid
- Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26000 Settat, Morocco
| | - Norbert Latruffe
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France
| | - M'Hammed Saïd El Kebbaj
- Laboratoire de recherche sur les Lipoprotéines et l'Athérosclérose, Faculté des Sciences Ben M'sik, Avenue Cdt Driss El Harti BP. 7955, Université Hassan II-Mohammedia-Casablanca, Morocco
| | | | - Gérard Lizard
- Université de Bourgogne, Laboratoire Bio-PeroxIL, EA7270, Dijon F-21000, France
| | - Boubker Nasser
- Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP 577, 26000 Settat, Morocco
| | | |
Collapse
|
145
|
Irwin RW, Brinton RD. Allopregnanolone as regenerative therapeutic for Alzheimer's disease: Translational development and clinical promise. Prog Neurobiol 2014; 113:40-55. [PMID: 24044981 PMCID: PMC10124616 DOI: 10.1016/j.pneurobio.2013.08.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/22/2013] [Accepted: 08/25/2013] [Indexed: 10/26/2022]
Abstract
Herein, we review a translational development plan to advance allopregnanolone to the clinic as a regenerative therapeutic for neurodegenerative diseases, in particular Alzheimer's. Allopregnanolone, an endogenous neurosteroid that declines with age and neurodegenerative disease, was exogenously administered and assessed for safety and efficacy to promote neuro-regeneration, cognitive function and reduction of Alzheimer's pathology. Allopregnanolone-induced neurogenesis correlated with restoration of learning and memory function in a mouse model of Alzheimer's disease and was comparably efficacious in aged normal mice. Critical to success was a dosing and treatment regimen that was consistent with the temporal requirements of systems biology of regeneration in brain. A treatment regimen that adhered to regenerative requirements of brain was also efficacious in reducing Alzheimer's pathology. With an optimized dosing and treatment regimen, chronic allopregnanolone administration promoted neurogenesis, oligodendrogenesis, reduced neuroinflammation and beta-amyloid burden while increasing markers of white matter generation and cholesterol homeostasis. Allopregnanolone meets three of the four drug-like physicochemical properties described by Lipinski's rule that predict the success rate of drugs in development for clinical trials. Pharmacokinetic and pharmacodynamic outcomes, securing GMP material, development of clinically translatable formulations and acquiring regulatory approval are discussed. Investigation of allopregnanolone as a regenerative therapeutic has provided key insights into mechanistic targets for neurogenesis and disease modification, dosing requirements, optimal treatment regimen, route of administration and the appropriate formulation necessary to advance to proof of concept clinical studies to determine efficacy of allopregnanolone as a regenerative and disease modifying therapeutic for Alzheimer's disease.
Collapse
|
146
|
Wang Q, Wang S, Shi Y, Yao M, Hou L, Jiang L. Reduction of Liver X Receptor β expression in primary rat neurons by antisense oligodeoxynucleotides decreases secreted amyloid β levels. Neurosci Lett 2014; 561:146-50. [PMID: 24394905 DOI: 10.1016/j.neulet.2013.12.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/26/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Ligand-activated Liver X Receptor (LXR) is known to increase cholesterol efflux from cells and reduce the production of amyloid β (Aβ) from amyloid-beta precursor protein (APP). However, little is known about the effects of LXRβ, one subtype of LXR, on endogenous Aβ. In this study, we show that LXRβ inactivation with specific antisense oligodeoxynucleotides (As-ODN) significantly reduced secreted Aβ and decreased mRNA levels of APP(751+770), and α-, β-secretase (ADAM10, BACE1) in primary rat neurons. We also show that As-ODN down-regulated the LXR responsive genes ABCA1 and HMG-CoA reductase (HMGCR). These changes are associated with decreased cellular cholesterol levels. The effect of LXRβ inactivation on Aβ levels is likely due to the alteration of cholesterol production and APP processing. Thus, our data suggest that LXRβ has an important function in cholesterol homeostasis and endogenous Aβ maintenance in neurons.
Collapse
Affiliation(s)
- Qie Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Suling Wang
- Hebei Province Blood Center, Shijiazhuang, China
| | - Yun Shi
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Min Yao
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Lianguo Hou
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Lingling Jiang
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
147
|
Parikh M, Patel K, Soni S, Gandhi T. Liver X Receptor: A Cardinal Target for Atherosclerosis and Beyond. J Atheroscler Thromb 2014. [DOI: 10.5551/jat.19778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
148
|
Dynamic changes of Apo A1 mediated by LXR/RXR/ABCA1 pathway in brains of the aging rats with cerebral hypoperfusion. Brain Res Bull 2013; 100:84-92. [PMID: 24291698 DOI: 10.1016/j.brainresbull.2013.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 11/20/2022]
Abstract
Cerebral hypoperfusion or aging often results into the disturbances of cholesterol and lipoprotein, which have been tightly associated with numerous neurological and psychiatric diseases, such as vascular dementia. The pathway of liver X receptor-β (LXR-β)/retinoic X receptor-α (RXR-α)/ABCA1 plays a vital role in lipoprotein metabolism. However, there were no reports about the relationship between the signal molecules of the pathway and lipoprotein homeostasis in cerebral hypoperfusion models. Therefore, we aimed to detect the expression of the pathway molecules in the aging rat models of chronic cerebral hypoperfusion and to explore its underlying mechanism. The model with cerebral hypoperfusion was established by ligating of the bilateral common carotid arteries (2VO). The temporal blood flow in the model rats was significantly decreased 14 d, 21 d and 28 d after 2VO compared with the control. The serum levels of high-density lipoprotein (HDL) and total cholesterol (TC) were reached a peak at 14 d, then, they were gradually decreased. The changes of LXR-β, RXR-α, ABCA1 and apolipoprotein A1 (apo A1) of the pathway were consistent with the changes of HDL and TC. We conclude that LXR-β/RXR-α/ABCA1 and downstream genes apo A1 undergo dynamic changes during the process of cerebral hypoperfusion. The LXR-β/RXR-α/ABCA1 mediated apo A1 cholesterol may play a protective effect, and the effect only exists in a certain period of time.
Collapse
|
149
|
Rong X, Albert CJ, Hong C, Duerr MA, Chamberlain BT, Tarling EJ, Ito A, Gao J, Wang B, Edwards PA, Jung ME, Ford DA, Tontonoz P. LXRs regulate ER stress and inflammation through dynamic modulation of membrane phospholipid composition. Cell Metab 2013; 18:685-97. [PMID: 24206663 PMCID: PMC3889491 DOI: 10.1016/j.cmet.2013.10.002] [Citation(s) in RCA: 236] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/27/2013] [Accepted: 09/26/2013] [Indexed: 12/11/2022]
Abstract
The fatty acyl composition of phospholipids determines the biophysical character of membranes and impacts the function of membrane proteins. Here, we define a nuclear receptor pathway for the dynamic modulation of membrane composition in response to changes in cellular lipid metabolism. Ligand activation of liver X receptors (LXRs) preferentially drives the incorporation of polyunsaturated fatty acids into phospholipids through induction of the remodeling enzyme Lpcat3. Promotion of Lpcat3 activity ameliorates endoplasmic reticulum (ER) stress induced by saturated free fatty acids in vitro or by hepatic lipid accumulation in vivo. Conversely, Lpcat3 knockdown in liver exacerbates ER stress and inflammation. Mechanistically, Lpcat3 modulates inflammation both by regulating inflammatory kinase activation through changes in membrane composition and by affecting substrate availability for inflammatory mediator production. These results outline an endogenous mechanism for the preservation of membrane homeostasis during lipid stress and identify Lpcat3 as an important mediator of LXR effects on metabolism.
Collapse
Affiliation(s)
- Xin Rong
- Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Spann NJ, Glass CK. Sterols and oxysterols in immune cell function. Nat Immunol 2013; 14:893-900. [PMID: 23959186 DOI: 10.1038/ni.2681] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 07/02/2013] [Indexed: 12/12/2022]
Abstract
Intermediates in the cholesterol-biosynthetic pathway and oxysterol derivatives of cholesterol regulate diverse cellular processes. Recent studies have expanded the appreciation of their roles in controlling the functions of cells of the innate and adaptive immune systems. Here we review recent literature reporting on the biological functions of sterol intermediates and oxysterols, acting through transcription factors such as the liver X receptors (LXRs), sterol regulatory element-binding proteins (SREBPs) and the G protein-coupled receptor EBI2, in regulating the differentiation and population expansion of cells of the innate and adaptive immune systems, their responses to inflammatory mediators, their effects on the phagocytic functions of macrophages and their effects on antiviral activities and the migration of immune cells. Such findings have raised many new questions about the production of endogenous bioactive sterols and oxysterols and their mechanisms of action in the immune system.
Collapse
Affiliation(s)
- Nathanael J Spann
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | | |
Collapse
|