1
|
Huang HY, Salinas S, Cornell J, Udoh IB, Shen Y, Zhou M. CCR5 regulates Aβ 1-42-induced learning and memory deficits in mice. Neurobiol Learn Mem 2024; 208:107890. [PMID: 38215963 DOI: 10.1016/j.nlm.2024.107890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/02/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
C-C chemokine receptor 5 (CCR5) is a chemokine receptor involved in immune responses and a co-receptor for HIV infection. Recently, CCR5 has also been reported to play a role in synaptic plasticity, learning and memory, and cognitive deficits associated with normal aging, traumatic brain injury (TBI), and HIV-associated neurocognitive disorder (HAND). In contrast, the role of CCR5 in cognitive deficits associated with other disorders, including Alzheimer's disease (AD), is much less understood. Studies have reported an increase in expression of CCR5 or its ligands in both AD patients and AD rodent models, suggesting a correlation between AD and CCR5 expression. However, whether blocking CCR5 in specific brain regions, such as the hippocampus, could improve memory deficits in AD mouse models is unknown. To study the potential causal role of CCR5 in cognitive deficits in AD, we injected soluble Aβ1-42 or a control (Aβ42-1) oligomers in the dorsal CA1 region of the hippocampus and found that Aβ1-42 injection resulted in severe memory impairment in the object place recognition (OPR) and novel object recognition (NOR) tests. Aβ1-42 injection caused an increase in Ccr5, Ccl3, and Ccl4 in the dorsal hippocampus, and the expression levels of CCR5 and its ligands remained elevated at 2 weeks after Aβ1-42 injection. Knocking down Ccr5 in the CA1 region of dorsal hippocampus reversed the increase in microglia number and size in dorsal CA1 and rescued memory deficits. These results indicate that CCR5 plays an important role in modulating Aβ1-42-induced learning and memory deficits, and suggest that CCR5 antagonists may serve as a potential treatment to improve cognitive deficits associated with AD.
Collapse
Affiliation(s)
- Hou-Yuan Huang
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Shelbi Salinas
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Jessica Cornell
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Iquo-Bella Udoh
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Yang Shen
- Neurobiology, Psychiatry and Psychology Departments & Integrative Center for Learning and Memory, UCLA, Los Angeles, CA, USA
| | - Miou Zhou
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
2
|
Tournier BB, Sorce S, Marteyn A, Ghidoni R, Benussi L, Binetti G, Herrmann FR, Krause K, Zekry D. CCR5 deficiency: Decreased neuronal resilience to oxidative stress and increased risk of vascular dementia. Alzheimers Dement 2024; 20:124-135. [PMID: 37489764 PMCID: PMC10917026 DOI: 10.1002/alz.13392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
INTRODUCTION As the chemokine receptor5 (CCR5) may play a role in ischemia, we studied the links between CCR5 deficiency, the sensitivity of neurons to oxidative stress, and the development of dementia. METHODS Logistic regression models with CCR5/apolipoprotein E (ApoE) polymorphisms were applied on a sample of 205 cognitively normal individuals and 189 dementia patients from Geneva. The impact of oxidative stress on Ccr5 expression and cell death was assessed in mice neurons. RESULTS CCR5-Δ32 allele synergized with ApoEε4 as risk factor for dementia and specifically for dementia with a vascular component. We confirmed these results in an independent cohort from Italy (157 cognitively normal and 620 dementia). Carriers of the ApoEε4/CCR5-Δ32 genotype aged ≥80 years have an 11-fold greater risk of vascular-and-mixed dementia. Oxidative stress-induced cell death in Ccr5-/- mice neurons. DISCUSSION We propose the vulnerability of CCR5-deficient neurons in response to oxidative stress as possible mechanisms contributing to dementia.
Collapse
Affiliation(s)
- Benjamin B. Tournier
- Department of PsychiatryGeneva University Hospitals and University of GenevaGenevaSwitzerland
| | - Silvia Sorce
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Antoine Marteyn
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
- Division of GeriatricsDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
- Division of Internal Medicine for the AgedDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| | - Roberta Ghidoni
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Luisa Benussi
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - François R Herrmann
- Division of GeriatricsDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| | - Karl‐Heinz Krause
- Department of Pathology and ImmunologyFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Dina Zekry
- Division of Internal Medicine for the AgedDepartment of Rehabilitation and GeriatricsGeneva University HospitalsThônexSwitzerland
| |
Collapse
|
3
|
Ma W, Liu A, Wu X, Gao L, Chen J, Wu H, Liu M, Fan Y, Peng L, Yang J, Kong J, Li B, Ji Z, Dong Y, Luo S, Song J, Bao F. The intricate role of CCL5/CCR5 axis in Alzheimer disease. J Neuropathol Exp Neurol 2023; 82:894-900. [PMID: 37769321 PMCID: PMC10587995 DOI: 10.1093/jnen/nlad071] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
The morbidity and mortality associated with Alzheimer disease (AD), one of the most common neurodegenerative diseases, are increasing each year. Although both amyloid β and tau proteins are known to be involved in AD pathology, their detailed functions in the pathogenesis of the disease are not fully understood. There is increasing evidence that neuroinflammation contributes to the development and progression of AD, with astrocytes, microglia, and the cytokines and chemokines they secrete acting coordinately in these processes. Signaling involving chemokine (C-C motif) ligand 5 (CCL5) and its main receptor C-C chemokine receptor 5 (CCR5) plays an important role in normal physiologic processes as well as pathologic conditions such as neurodegeneration. In recent years, many studies have shown that the CCL5/CCR5 axis plays a major effect in the pathogenesis of AD, but there are also a few studies that contradict this. In short, the role of CCL5/CCR5 axis in the pathogenesis of AD is still intricate. This review summarizes the structure, distribution, physiologic functions of the CCL5/CCR5 axis, and the progress in understanding its involvement in the pathogenesis of AD.
Collapse
Affiliation(s)
- Weijiang Ma
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Aihua Liu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
- Yunnan Province Key Laboratory of Children’s Major Diseases Research, The Affiliated Children Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Xinya Wu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Li Gao
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jingjing Chen
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Hanxin Wu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Meixiao Liu
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Yuxin Fan
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Li Peng
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jiaru Yang
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jing Kong
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Bingxue Li
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Zhenhua Ji
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Yan Dong
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Suyi Luo
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Jieqin Song
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Fukai Bao
- Evidence-Based Medicine Team, Faculty of Basic Medical Sciences, The Institute for Tropical Medicine, Kunming Medical University, Kunming, Yunnan, China
- Yunnan Province Key Laboratory of Children’s Major Diseases Research, The Affiliated Children Hospital, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
4
|
Kim B, Vasanthakumar A, Li QS, Nudelman KN, Risacher SL, Davis JW, Idler K, Lee J, Seo SW, Waring JF, Saykin AJ, Nho K, for the Alzheimer's Disease Neuroimaging Initiative (ADNI). Integrative analysis of DNA methylation and gene expression identifies genes associated with biological aging in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12354. [PMID: 36187194 PMCID: PMC9489162 DOI: 10.1002/dad2.12354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022]
Abstract
Introduction The acceleration of biological aging is a risk factor for Alzheimer's disease (AD). Here, we performed weighted gene co-expression network analysis (WGCNA) to identify modules and dysregulated genesinvolved in biological aging in AD. Methods We performed WGCNA to identify modules associated with biological clocks and hub genes of the module with the highest module significance. In addition, we performed differential expression analysis and association analysis with AD biomarkers. Results WGCNA identified five modules associated with biological clocks, with the module designated as "purple" showing the strongest association. Functional enrichment analysis revealed that the purple module was related to cell migration and death. Ten genes were identified as hub genes in purple modules, of which CX3CR1 was downregulated in AD and low levels of CX3CR1 expression were associated with AD biomarkers. Conclusion Network analysis identified genes associated with biological clocks, which suggests the genetic architecture underlying biological aging in AD. Highlights Examine links between Alzheimer's disease (AD) peripheral transcriptome and biological aging changes.Weighted gene co-expression network analysis (WGCNA) found five modules related to biological aging.Among the hub genes of the module, CX3CR1 was downregulated in AD.The CX3CR1 expression level was associated with cognitive performance and brain atrophy.
Collapse
Affiliation(s)
- Bo‐Hyun Kim
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Samsung Alzheimer Research CenterSamsung Medical CenterSeoulRepublic of Korea
- Department of Health Sciences and TechnologySHAISTSungkyunkwan UniversitySeoulRepublic of Korea
| | | | - Qingqin S. Li
- Neuroscience Therapeutic AreaJanssen Research & Development, LLCTitusvilleNew JerseyUSA
| | - Kelly N.H. Nudelman
- National Centralized Repository for Alzheimer's Disease and Related DementiasIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Shannon L. Risacher
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Kenneth Idler
- Genomics Research CenterAbbVieNorth ChicagoIllinoisUSA
| | - Jong‐Min Lee
- Department of Biomedical EngineeringHanyang UniversitySeoulRepublic of Korea
| | - Sang Won Seo
- Samsung Alzheimer Research CenterSamsung Medical CenterSeoulRepublic of Korea
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
- Department of Health Sciences and TechnologySHAISTSungkyunkwan UniversitySeoulRepublic of Korea
| | | | - Andrew J. Saykin
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kwangsik Nho
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
5
|
Role of Chemokines in the Development and Progression of Alzheimer's Disease. J Mol Neurosci 2022; 72:1929-1951. [PMID: 35821178 PMCID: PMC9392685 DOI: 10.1007/s12031-022-02047-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurogenerative disorder manifested by gradual memory loss and cognitive decline due to profound damage of cholinergic neurons. The neuropathological hallmarks of AD are intracellular deposits of neurofibrillary tangles (NFTs) and extracellular aggregates of amyloid β (Aβ). Mounting evidence indicates that intensified neuroinflammatory processes play a pivotal role in the pathogenesis of AD. Chemokines serve as signaling molecules in immune cells but also in nerve cells. Under normal conditions, neuroinflammation plays a neuroprotective role against various harmful factors. However, overexpression of chemokines initiates disruption of the integrity of the blood–brain barrier, facilitating immune cells infiltration into the brain. Then activated adjacent glial cells–astrocytes and microglia, release massive amounts of chemokines. Prolonged inflammation loses its protective role and drives an increase in Aβ production and aggregation, impairment of its clearance, or enhancement of tau hyperphosphorylation, contributing to neuronal loss and exacerbation of AD. Moreover, chemokines can be further released in response to growing deposits of toxic forms of Aβ. On the other hand, chemokines seem to exert multidimensional effects on brain functioning, including regulation of neurogenesis and synaptic plasticity in regions responsible for memory and cognitive abilities. Therefore, underexpression or complete genetic ablation of some chemokines can worsen the course of AD. This review covers the current state of knowledge on the role of particular chemokines and their receptors in the development and progression of AD. Special emphasis is given to their impact on forming Aβ and NFTs in humans and in transgenic murine models of AD.
Collapse
|
6
|
Feng YQ, Xu ZZ, Wang YT, Xiong Y, Xie W, He YY, Chen L, Liu GY, Li X, Liu J, Wu Q. Targeting C–C Chemokine Receptor 5: Key to Opening the Neurorehabilitation Window After Ischemic Stroke. Front Cell Neurosci 2022; 16:876342. [PMID: 35573839 PMCID: PMC9095921 DOI: 10.3389/fncel.2022.876342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the world’s second major cause of adult death and disability, resulting in the destruction of brain tissue and long-term neurological impairment; induction of neuronal plasticity can promote recovery after stroke. C–C chemokine receptor 5 (CCR5) can direct leukocyte migration and localization and is a co-receptor that can mediate human immunodeficiency virus (HIV) entry into cells. Its role in HIV infection and immune response has been extensively studied. Furthermore, CCR5 is widely expressed in the central nervous system (CNS), is engaged in various physiological activities such as brain development, neuronal differentiation, communication, survival, and learning and memory capabilities, and is also involved in the development of numerous neurological diseases. CCR5 is differentially upregulated in neurons after stroke, and the inhibition of CCR5 in specific regions of the brain promotes motor and cognitive recovery. The mechanism by which CCR5 acts as a therapeutic target to promote neurorehabilitation after stroke has rarely been systematically reported yet. Thus, this review aims to discuss the function of CCR5 in the CNS and the mechanism of its effect on post-stroke recovery by regulating neuroplasticity and the inflammatory response to provide an effective basis for clinical rehabilitation after stroke.
Collapse
|
7
|
Bauss J, Morris M, Shankar R, Olivero R, Buck LN, Stenger CL, Hinds D, Mills J, Eby A, Zagorski JW, Smith C, Cline S, Hartog NL, Chen B, Huss J, Carcillo JA, Rajasekaran S, Bupp CP, Prokop JW. CCR5 and Biological Complexity: The Need for Data Integration and Educational Materials to Address Genetic/Biological Reductionism at the Interface of Ethical, Legal, and Social Implications. Front Immunol 2021; 12:790041. [PMID: 34925370 PMCID: PMC8674737 DOI: 10.3389/fimmu.2021.790041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/18/2021] [Indexed: 01/02/2023] Open
Abstract
In the age of genomics, public understanding of complex scientific knowledge is critical. To combat reductionistic views, it is necessary to generate and organize educational material and data that keep pace with advances in genomics. The view that CCR5 is solely the receptor for HIV gave rise to demand to remove the gene in patients to create host HIV resistance, underestimating the broader roles and complex genetic inheritance of CCR5. A program aimed at providing research projects to undergraduates, known as CODE, has been expanded to build educational material for genes such as CCR5 in a rapid approach, exposing students and trainees to large bioinformatics databases and previous experiments for broader data to challenge commitment to biological reductionism. Our students organize expression databases, query environmental responses, assess genetic factors, generate protein models/dynamics, and profile evolutionary insights into a protein such as CCR5. The knowledgebase generated in the initiative opens the door for public educational information and tools (molecular videos, 3D printed models, and handouts), classroom materials, and strategy for future genetic ideas that can be distributed in formal, semiformal, and informal educational environments. This work highlights that many factors are missing from the reductionist view of CCR5, including the role of missense variants or expression of CCR5 with neurological phenotypes and the role of CCR5 and the delta32 variant in complex critical care patients with sepsis. When connected to genomic stories in the news, these tools offer critically needed Ethical, Legal, and Social Implication (ELSI) education to combat biological reductionism.
Collapse
Affiliation(s)
- Jacob Bauss
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Michele Morris
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States
| | - Rama Shankar
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Rosemary Olivero
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Infectious Disease, Helen DeVos Children's Hospital, Grand Rapids, MI, United States
| | - Leah N Buck
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Department of Mathematics, University of North Alabama, Florence, AL, United States
| | - Cynthia L Stenger
- Department of Mathematics, University of North Alabama, Florence, AL, United States
| | - David Hinds
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States
| | - Joshua Mills
- Department of Biology, Grand Valley State University, Allendale, MI, United States
| | - Alexandra Eby
- Department of Science, Davenport University, Grand Rapids, MI, United States
| | - Joseph W Zagorski
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
| | - Caitlin Smith
- Department of Biology, Athens State University, Athens, AL, United States
| | - Sara Cline
- Department of Biology, Athens State University, Athens, AL, United States
| | - Nicholas L Hartog
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Allergy & Immunology, Spectrum Health, Grand Rapids, MI, United States
| | - Bin Chen
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - John Huss
- Department of Philosophy, The University of Akron, Akron, OH, United States
| | - Joseph A Carcillo
- Department of Critical Care Medicine and Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Office of Research, Spectrum Health, Grand Rapids, MI, United States.,Pediatric Intensive Care Unit, Helen DeVos Children's Hospital, Grand Rapids, MI, United States
| | - Caleb P Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Medical Genetics, Spectrum Health, Grand Rapids, MI, United States
| | - Jeremy W Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
8
|
Rather MA, Khan A, Alshahrani S, Rashid H, Qadri M, Rashid S, Alsaffar RM, Kamal MA, Rehman MU. Inflammation and Alzheimer's Disease: Mechanisms and Therapeutic Implications by Natural Products. Mediators Inflamm 2021; 2021:9982954. [PMID: 34381308 PMCID: PMC8352708 DOI: 10.1155/2021/9982954] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/24/2021] [Accepted: 07/10/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with no clear causative event making the disease difficult to diagnose and treat. The pathological hallmarks of AD include amyloid plaques, neurofibrillary tangles, and widespread neuronal loss. Amyloid-beta has been extensively studied and targeted to develop an effective disease-modifying therapy, but the success rate in clinical practice is minimal. Recently, neuroinflammation has been focused on as the event in AD progression to be targeted for therapies. Various mechanistic pathways including cytokines and chemokines, complement system, oxidative stress, and cyclooxygenase pathways are linked to neuroinflammation in the AD brain. Many cells including microglia, astrocytes, and oligodendrocytes work together to protect the brain from injury. This review is focused to better understand the AD inflammatory and immunoregulatory processes to develop novel anti-inflammatory drugs to slow down the progression of AD.
Collapse
Affiliation(s)
- Mashoque Ahmad Rather
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Tamil Nadu 608002, India
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hina Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Marwa Qadri
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Rana M. Alsaffar
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
9
|
Lack of Association Between the CCR5-delta32 Polymorphism and Neurodegenerative Disorders. Alzheimer Dis Assoc Disord 2021; 34:244-247. [PMID: 31972607 DOI: 10.1097/wad.0000000000000367] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Recent studies have suggested that diminished Ccr5 functioning has an effect on synaptic plasticity and hippocampal memory in mouse models. CCR5-delta32, a 32-bp frameshift deletion in human CCR5 encoding a nonfunctional receptor, has been reported to have a protective effect against human immunodeficiency virus infection but its role as a modifier of neurodegenerative disease has been minimally explored. We investigated whether the CCR5-delta32 polymorphism could have an effect in the context of human neurodegenerative diseases. METHODS We examined the frequency of the CCR5-delta32 polymorphism in a large and well-characterized cohort including 1425 patients with neurodegenerative dementias and 2032 controls. RESULTS We did not observe a significant association between the CCR5-delta32 polymorphism and any of the neurodegenerative diseases screened in this study. However, we observed an earlier age of onset among neurodegenerative disease patients carrying the CCR5-delta32 allele. CONCLUSIONS Although our findings were inconclusive, the earlier age of onset observed among neurodegenerative disease patients carrying the CCR5-delta32 allele suggests that the deletion may have a detrimental effect in the context of neurodegeneration.
Collapse
|
10
|
Ping S, Qiu X, Kyle M, Zhao LR. Brain-derived CCR5 Contributes to Neuroprotection and Brain Repair after Experimental Stroke. Aging Dis 2021; 12:72-92. [PMID: 33532129 PMCID: PMC7801286 DOI: 10.14336/ad.2020.0406] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 02/04/2023] Open
Abstract
Chemokine (C-C motif) receptor 5 (CCR5) is expressed not only in the immune cells but also in cerebral cells such as neurons, glia, and vascular cells. Stroke triggers high expression of CCR5 in the brain. However, the role of CCR5 in stroke remains unclear. In this study, using bone marrow chimeras we have determined the involvement of brain-derived or bone marrow-derived CCR5 in neuroprotection and brain repair after experimental stroke. CCR5-/- mice that received either wild-type (WT) or CCR5-/- bone marrow transplantation showed larger infarction sizes than the WT mice that received either WT or CCR5-/- bone marrow transplantation in both the acute (48h) and subacute (2 months) phases after cerebral cortical ischemia, suggesting that the lack of CCR5 in the brain leads to severe brain damage after stroke. However, the lack of CCR5 in the bone marrow-derived cells did not affect infarction size. The impairments of somatosensory-motor function and motor coordination were exacerbated in the mice lacking CCR5 in the brain. At 2 months post-stroke, increased degenerative neurons, decreased dendrites and synapses, decreased Iba1+ microglia/ macrophages, reduced myelination and CNPase+ oligodendrocytes in the peri-infarct cortex were observed in the mice lacking CCR5 in the brain. These pathological changes are significantly correlated with the increased infarction size and exacerbated neurological deficits. These data suggest that brain-derived CCR5 plays a key role in neuroprotection and brain repair in the subacute phase of stroke. This study reveals a novel role of CCR5 in stroke, which sheds new light on post-stroke pathomechanism.
Collapse
Affiliation(s)
- Suning Ping
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| | - Xuecheng Qiu
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| | - Michele Kyle
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| |
Collapse
|
11
|
Necula D, Riviere-Cazaux C, Shen Y, Zhou M. Insight into the roles of CCR5 in learning and memory in normal and disordered states. Brain Behav Immun 2021; 92:1-9. [PMID: 33276089 DOI: 10.1016/j.bbi.2020.11.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/25/2020] [Accepted: 11/28/2020] [Indexed: 12/21/2022] Open
Abstract
As cognitive impairments continue to rise in prevalence, there is an urgent need to understand the mechanisms of learning and memory in normal and disordered states. C-C chemokine receptor 5 (CCR5) has been implicated in the regulation of multiple forms of learning and memory via its regulation on learning-related cell signaling and neuronal plasticity. As a chemokine receptor and a co-receptor for HIV, CCR5's role in immune response and HIV-associated neurocognitive disorder (HAND) has been widely studied. In contrast, CCR5 is less understood in cognitive deficits associated with other disorders, including Alzheimer's disease (AD), stroke and certain psychiatric disorders. A broad overview of the present literature shows that CCR5 acts as a potent suppressor of synaptic plasticity and learning and memory, although a few studies have reported the opposite effect of CCR5 in stroke or AD animal models. By summarizing the current literature of CCR5 in animal and human studies of cognition, this review aims to provide a comprehensive overview of the role of CCR5 in learning and memory in both normal and disordered states and to discuss the possibility of CCR5 suppression as an effective therapeutic to alleviate cognitive deficits in HAND, AD, and stroke.
Collapse
Affiliation(s)
- Deanna Necula
- Department of Neuroscience, UCSF, San Francisco, CA, USA
| | - Cecile Riviere-Cazaux
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Yang Shen
- Neurobiology, Psychiatry and Psychology Departments & Integrative Center for Learning and Memory, UCLA, Los Angeles, CA, USA
| | - Miou Zhou
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
12
|
Rubio T, Felipo V, Tarazona S, Pastorelli R, Escudero-García D, Tosca J, Urios A, Conesa A, Montoliu C. Multi-omic analysis unveils biological pathways in peripheral immune system associated to minimal hepatic encephalopathy appearance in cirrhotic patients. Sci Rep 2021; 11:1907. [PMID: 33479266 PMCID: PMC7820002 DOI: 10.1038/s41598-020-80941-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/23/2020] [Indexed: 01/29/2023] Open
Abstract
Patients with liver cirrhosis may develop minimal hepatic encephalopathy (MHE) which affects their quality of life and life span. It has been proposed that a shift in peripheral inflammation triggers the appearance of MHE. However, the mechanisms involved in this immune system shift remain unknown. In this work we studied the broad molecular changes involved in the induction of MHE with the goal of identifying (1) altered genes and pathways in peripheral blood cells associated to the appearance of MHE, (2) serum metabolites and cytokines with modified levels in MHE patients and (3) MHE-regulated immune response processes related to changes in specific serum molecules. We adopted a multi-omic approach to profile the transcriptome, metabolome and a panel of cytokines of blood samples taken from cirrhotic patients with or without MHE. Transcriptomic analysis supports the hypothesis of alternations in the Th1/Th2 and Th17 lymphocytes cell populations as major drivers of MHE. Cluster analysis of serum molecules resulted in six groups of chemically similar compounds, suggesting that functional modules operate during the induction of MHE. Finally, the multi-omic integrative analysis suggested a relationship between cytokines CCL20, CX3CL1, CXCL13, IL-15, IL-22 and IL-6 with alteration in chemotaxis, as well as a link between long-chain unsaturated phospholipids and the increased fatty acid transport and prostaglandin production. We found altered immune pathways that may collectively contribute to the mild cognitive impairment phenotype in MHE. Our approach is able to combine extracellular and intracellular information, opening new insights to the understanding of the disease.
Collapse
Affiliation(s)
- Teresa Rubio
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Sonia Tarazona
- Departamento de Estadística e Investigación Operativa Aplicadas y Calidad, Universitat Politècnica de València, Valencia, Spain
| | - Roberta Pastorelli
- Protein and Metabolite Biomarkers Unit, Laboratory of Mass Spectrometry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Desamparados Escudero-García
- Unidad de Digestivo, Hospital Clínico de Valencia, Departamento Medicina, Universidad de Valencia, Valencia, Spain
| | - Joan Tosca
- Unidad de Digestivo, Hospital Clínico de Valencia, Valencia, Spain
| | - Amparo Urios
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
- Neurological Impairment Laboratory, Fundación Investigación Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain
| | - Ana Conesa
- Microbiology and Cell Science Department, Institute for Food and Agricultural Sciences, Genetics Institute, University of Florida, Gainesville, USA.
| | - Carmina Montoliu
- Neurological Impairment Laboratory, Fundación Investigación Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria-INCLIVA, Valencia, Spain
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
13
|
Wang H, Shen Y, Chuang H, Chiu C, Ye Y, Zhao L. Neuroinflammation in Alzheimer's Disease: Microglia, Molecular Participants and Therapeutic Choices. Curr Alzheimer Res 2020; 16:659-674. [PMID: 31580243 DOI: 10.2174/1567205016666190503151648] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/21/2019] [Accepted: 04/30/2019] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is the world's most common dementing illness. It is pathologically characterized by β-amyloid accumulation, extracellular senile plaques and intracellular neurofibrillary tangles formation, and neuronal necrosis and apoptosis. Neuroinflammation has been widely recognized as a crucial process that participates in AD pathogenesis. In this review, we briefly summarized the involvement of microglia in the neuroinflammatory process of Alzheimer's disease. Its roles in the AD onset and progression are also discussed. Numerous molecules, including interleukins, tumor necrosis factor alpha, chemokines, inflammasomes, participate in the complex process of AD-related neuroinflammation and they are selectively discussed in this review. In the end of this paper from an inflammation- related perspective, we discussed some potential therapeutic choices.
Collapse
Affiliation(s)
- Haijun Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yin Shen
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haoyu Chuang
- Department of Neurosurgery, Tainan Municipal An-Nan Hospital, Tainan, Taiwan.,Department of Neurosurgery, China Medical University Bei-Gang Hospital, Yun-Lin, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan
| | - Chengdi Chiu
- School of Medicine, China Medical University, Taichung, Taiwan.,Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan
| | - Youfan Ye
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Zhao
- Department of Infectious Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
14
|
Insights into the Gene Expression Profiles of Active and Restricted Red/Green-HIV + Human Astrocytes: Implications for Shock or Lock Therapies in the Brain. J Virol 2020; 94:JVI.01563-19. [PMID: 31896591 DOI: 10.1128/jvi.01563-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022] Open
Abstract
A significant number of people living with human immunodeficiency virus type 1 (HIV-1) suffer from HIV-associated neurocognitive disorders (HAND). Many previous studies investigating HIV in astrocytes as a heterogenous population have established the relevance of astrocytes to HIV-associated neuropathogenesis. However, these studies were unable to differentiate the state of infection, i.e., active or latent, or to evaluate how this affects astrocyte biology. In this study, the pseudotyped doubly labeled fluorescent reporter red/green (R/G)-HIV-1 was used to identify and enrich restricted and active populations of HIV+ astrocytes based on the viral promoter activity. Here, we report that the majority of human astrocytes restricted R/G-HIV-1 gene expression early during infection and were resistant to reactivation by vorinostat and interleukin 1β. However, actively infected astrocytes were inducible, leading to increased expression of viral proteins upon reactivation. R/G-HIV-1 infection also significantly decreased the cell proliferation and glutamate clearance ability of astrocytes, which may contribute to excitotoxicity. Moreover, transcriptome analyses to compare gene expression patterns of astrocyte harboring active versus restricted long terminal repeats (LTRs) revealed that the gene expression patterns were similar and that the active population demonstrated more widespread and robust changes. Our data suggest that harboring the HIV genome profoundly alters astrocyte biology and that strategies that keep the virus latent (e.g., block and lock) or those that reactivate the latent virus (e.g., shock and kill) would be detrimental to astrocyte function and possibly augment their contributions to HAND.IMPORTANCE More than 36 million people are living with HIV-1 worldwide, and despite antiretroviral therapy, 30 to 50% of the people living with HIV-1 suffer from mild to moderate neurocognitive disorders. HIV-1 reservoirs in the central nervous system (CNS) are challenging to address due to low penetration of antiretroviral drugs, lack of resident T cells, and permanent integration of provirus into neural cells such as microglia and astrocytes. Several studies have shown astrocyte dysfunction during HIV-1 infection. However, little is known about how HIV-1 latency affects their function. The significance of our research is in identifying that the majority of HIV+ astrocytes restrict HIV expression and were resistant to reactivation. Further, simply harboring the HIV genome profoundly altered astrocyte biology, resulting in a proinflammatory phenotype and functional changes. In this context, therapeutic strategies to reactivate or silence astrocyte HIV reservoirs, without excising proviral DNA, will likely lead to detrimental neuropathological outcomes during HIV CNS infection.
Collapse
|
15
|
Ellwanger JH, Kaminski VDL, Chies JA. What we say and what we mean when we say redundancy and robustness of the chemokine system - how CCR5 challenges these concepts. Immunol Cell Biol 2019; 98:22-27. [PMID: 31613403 DOI: 10.1111/imcb.12291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
Abstract
Classically, robustness and redundancy are features that define the intricate and connected functioning of the chemokine system. Following these ideas, the lack of a particular chemokine or chemokine receptor could be compensated by the presence of other molecules with similar functions, ensuring robustness to the systems. Although these concepts are generally accepted, they represent an oversimplification of a highly complex system that works in a context-dependent manner. Based on different studies, and taking as an example the chemokine receptor CCR5 (C-C chemokine receptor type 5) and the genetic variant CCR5Δ32, which on several occasions challenge the classical concepts of redundancy and robustness of the chemokine system, we will discuss and question this general and oversimplified point of view.
Collapse
Affiliation(s)
- Joel H Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Valéria de L Kaminski
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - José Ab Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
16
|
Li T, Zhu J. Entanglement of CCR5 and Alzheimer's Disease. Front Aging Neurosci 2019; 11:209. [PMID: 31447666 PMCID: PMC6692443 DOI: 10.3389/fnagi.2019.00209] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022] Open
Abstract
Although the mechanisms of Alzheimer's disease are diverse and unclear, the past 20 years have witnessed the unprecedented development of the AD inflammation theory. As a key inflammatory receptor family, the C-C chemokine receptor family is a remarkable participant in the cause of Alzheimer's disease; of this family, CCR5 is the most widely studied. CCR5 is an essential entrance when HIV infects immune cells and is also involved in other inflammatory and immune activities. New evidence on the inevitably intertwined link between Alzheimer's disease and CCR5 indicates that CCR5 accelerates the development of Alzheimer's disease, and few studies disputed it. The role of CCR5 in Alzheimer's disease remains elusive. However, as the research progresses, this intricate relationship will gradually be uncovered.
Collapse
Affiliation(s)
- Tianwen Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Institutes of Brain Science, Shanghai, China
| |
Collapse
|
17
|
Zuena AR, Casolini P, Lattanzi R, Maftei D. Chemokines in Alzheimer's Disease: New Insights Into Prokineticins, Chemokine-Like Proteins. Front Pharmacol 2019; 10:622. [PMID: 31231219 PMCID: PMC6568308 DOI: 10.3389/fphar.2019.00622] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder characterized by the presence of β-amyloid aggregates deposited as senile plaques and by the presence of neurofibrillary tangles of tau protein. To date, there is a broad consensus on the idea that neuroinflammation is one of the most important component in Alzheimer’s disease pathogenesis. Chemokines and their receptors, beside the well-known role in the immune system, are widely expressed in the nervous system, where they play a significant role in the neuroinflammatory processes. Prokineticins are a new family of chemokine-like molecules involved in numerous physiological and pathological processes including immunity, pain, inflammation, and neuroinflammation. Prokineticin 2 (PROK2) and its receptors PKR1 and PKR2 are widely expressed in the central nervous system in both neuronal and glial cells. In Alzheimer’s disease, PROK2 sustains the neuroinflammatory condition and contributes to neurotoxicity, since its expression is strongly upregulated by amyloid-β peptide and reversed by the PKR antagonist PC1. This review aims to summarize the current knowledge on the neurotoxic and/or neuroprotective function of chemokines in Alzheimer’s disease, focusing on the prokineticin system: it represents a new field of investigation that can stimulate the research of innovative pharmacotherapeutic strategies.
Collapse
Affiliation(s)
- Anna Rita Zuena
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University of Rome, Rome, Italy
| | - Paola Casolini
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University of Rome, Rome, Italy
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology "Vittorio Erspamer," Sapienza University of Rome, Rome, Italy
| | - Daniela Maftei
- Department of Biochemical Sciences "Alessandro Rossi Fanelli," Sapienza University of Rome, Rome, Italy
| |
Collapse
|
18
|
Sui Y, Zhang Y, Dong C, Xu B, Sun X. The small molecular CCR3 antagonist YM344031 attenuates neurodegenerative pathologies and improves learning and memory performance in a mouse model of Alzheimer's disease. Brain Res 2019; 1719:1-10. [PMID: 31121157 DOI: 10.1016/j.brainres.2019.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/03/2019] [Accepted: 05/19/2019] [Indexed: 12/13/2022]
Abstract
The chemokine C-C receptor 3 (CCR3) plays a role in the pathogenesis of Alzheimer's disease (AD). Based on our previous observations that deletion of CCR3 prevented neurodegenerative pathologies in amyloid precursor protein/presenilin 1 (APP/PS1) double-transgenic mice, we hypothesize that CCR3 antagonists may provide therapeutic benefits to AD. To this end, we examined the effect of the brain-penetrable CCR3 antagonist, YM344031, on AD-related pathologies in APP/PS1 double transgenic mice. Treatment of 10-month-old APP/PS1 double-transgenic mice with YM344031 (50 mg/kg, b.i.d.) for two months resulted in dramatic decreases in β-amyloid deposition, tau hyperphosphorylation and synaptic loss in the forebrain, significant attenuation of microgliosis and astrogliosis, and marked improvement of spatial learning and memory performance compared with the vehicle-treated mice. These results support CCR3 antagonism as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Yi Sui
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Yao Zhang
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Chunyao Dong
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Bing Xu
- Department of Neurology, Shenyang First People's Hospital, Shenyang Brain Institute, Shenyang Medical College Affiliated Shenyang Brain Hospital, Shenyang 110047, China
| | - Xiaohong Sun
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang 110004, China.
| |
Collapse
|
19
|
CCR5 gene editing – Revisiting pros and cons of CCR5 absence. INFECTION GENETICS AND EVOLUTION 2019; 68:218-220. [DOI: 10.1016/j.meegid.2018.12.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/19/2018] [Accepted: 12/23/2018] [Indexed: 01/08/2023]
|
20
|
Jorda A, Cauli O, Santonja JM, Aldasoro M, Aldasoro C, Obrador E, Vila JM, Mauricio MD, Iradi A, Guerra-Ojeda S, Marchio P, Valles SL. Changes in Chemokines and Chemokine Receptors Expression in a Mouse Model of Alzheimer's Disease. Int J Biol Sci 2019; 15:453-463. [PMID: 30745834 PMCID: PMC6367555 DOI: 10.7150/ijbs.26703] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/21/2018] [Indexed: 11/18/2022] Open
Abstract
The amyloid precursor protein plus presenilin-1 (APP/PS1) mice are a frequently-used model for Alzheimer's disease studies (AD). However, the data relevant to which proteins are involved in inflammatory mechanism are not sufficiently well-studied using the AD mouse model. Using behavioral studies, quantitative RT-PCR and Western-blot techniques, significant findings were determined by the expression of proteins involved in inflammation comparing APP/PS1 and Wild type mice. Increased GFAP expression could be associated with the elevation in number of reactive astrocytes. IL-3 is involved in inflammation and ABDF1 intervenes normally in the transport across cell membranes and both were found up-regulated in APP/PS1 mice compared to Wild type mice. Furthermore, CCR5 expression was decreased and both CCL3 and CCL4 chemokines were highly expressed indicating a possible gliosis and probably an increase in chemotaxis from lymphocytes and T cell generation. We also noted for the first time, a CCR8 increase expression with diminution of its CCL1 chemokine, both normally involved in protection from bacterial infection and demyelination. Control of inflammatory proteins will be the next step in understanding the progression of AD and also in determining the mechanisms that can develop in this disease.
Collapse
Affiliation(s)
- Adrián Jorda
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Omar Cauli
- Faculty of Surgery and Chiropody, University of Valencia. Spain
| | | | - Martin Aldasoro
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Constanza Aldasoro
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Elena Obrador
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Jose Ma Vila
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | | | - Antonio Iradi
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Patricia Marchio
- Department of Physiology, School of Medicine, University of Valencia. Spain
| | - Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia. Spain
| |
Collapse
|
21
|
Guedes JR, Lao T, Cardoso AL, El Khoury J. Roles of Microglial and Monocyte Chemokines and Their Receptors in Regulating Alzheimer's Disease-Associated Amyloid-β and Tau Pathologies. Front Neurol 2018; 9:549. [PMID: 30158892 PMCID: PMC6104478 DOI: 10.3389/fneur.2018.00549] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023] Open
Abstract
Chemokines and their receptors have been shown to affect amyloid-β (Aβ) and tau pathologies in mouse models of Alzheimer's disease (AD) by regulating microglia and monocyte-associated neuroinflammation, microglial movement and monocyte recruitment into the brain. These cells in turn can promote and mediate Aβ phagocytosis and degradation and tau phosphorylation. In this review we discuss published work in this field in mouse models of AD and review what is known about the contributions of microglial and monocyte chemokines and their receptors to amyloid and tau pathologies. We focus on the roles of the chemokine/chemokine receptor pairs CCL2/CCR2, CX3CL1/CX3CR1, CCL5/CCR5, CXCL10/CXCR3 and CXCL1/CXCR2, highlighting important knowledge gaps in this field. A full understanding of the functions of chemokines and their receptors in AD may guide the development of novel immunotherapies for this devastating disease.
Collapse
Affiliation(s)
- Joana R Guedes
- Doctoral Programme in Experimental Biology and Biomedicine, CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.,CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Taotao Lao
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ana L Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Joseph El Khoury
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
22
|
Vérité J, Janet T, Chassaing D, Fauconneau B, Rabeony H, Page G. Longitudinal chemokine profile expression in a blood-brain barrier model from Alzheimer transgenic versus wild-type mice. J Neuroinflammation 2018; 15:182. [PMID: 29898739 PMCID: PMC6001165 DOI: 10.1186/s12974-018-1220-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease is widely described since the discovery of histopathological lesions in Mrs. Auguste Deter in 1906. However to date, there is no effective treatment to deal with the many cellular and molecular alterations. The complexity is even higher with the growing evidence of involvement of the peripheral blood mononuclear cells (PBMCs). Indeed, monocytes and T cells are shown in the cerebral parenchyma of AD patients, and these cells grafted to the periphery are able to go through the blood-brain barrier (BBB) in transgenic mouse models. It is known that BBB is disrupted at a late stage of AD. Chemokines represent major regulators of the transmigration of PBMCs, but many data were obtained on AD animal models. No data are available on the role of AD BBB in a healthy brain parenchyma. Therefore, the purpose of this study was to analyze the longitudinal chemokine profile expression in a BBB model from AD transgenic mice versus wild-type (WT) mice. METHODS A primary mouse BBB model was used with a luminal compartment either AD or WT and an abluminal compartment WT consisting of astrocytes and microglia. PBMCs were extracted by a ficoll gradient and incubated in the transwell with a direct contact with the luminal side, including the endothelial cells and pericytes. Then, the complete BBB model was incubated during 48 h, before supernatants and cell lysates were collected. Chemokines were quantified by X-MAP® luminex technology. RESULTS Abluminal CX3CL1 production increased in 12-month-old AD BBB while CX3CL1 levels decreased in luminal lysates. CCL3 in luminal compartment increased with aging and was significantly different compared to AD BBB at 12 months. In addition, abluminal CCL2 in 12-month-old AD BBB greatly decreased compared to levels in WT BBB. On the contrary, no modification was observed for CCL4, CCL5, and CXCL10. CONCLUSION These first findings highlighted the impact of AD luminal compartment on chemokine signature in a healthy brain parenchyma, suggesting new therapeutic or diagnostic approaches.
Collapse
Affiliation(s)
- J. Vérité
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, 86073 Poitiers, France
| | - T. Janet
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, 86073 Poitiers, France
| | - D. Chassaing
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, 86073 Poitiers, France
| | - B. Fauconneau
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, 86073 Poitiers, France
| | - H. Rabeony
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, 86073 Poitiers, France
- SATT Grand Centre- Société d’Accélération du Transfert de Technologie, 8, rue Pablo Picasso, 63000 Clermont-Ferrand, France
| | - G. Page
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, 86073 Poitiers, France
| |
Collapse
|
23
|
Netrin-1 improves the amyloid-β-mediated suppression of memory and synaptic plasticity. Brain Res Bull 2017; 131:107-116. [DOI: 10.1016/j.brainresbull.2017.03.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 03/14/2017] [Accepted: 03/29/2017] [Indexed: 11/24/2022]
|
24
|
Domingues C, Cruz e Silva OA, Henriques AG. Impact of Cytokines and Chemokines on Alzheimer's Disease Neuropathological Hallmarks. Curr Alzheimer Res 2017; 14:870-882. [PMID: 28317487 PMCID: PMC5543563 DOI: 10.2174/1567205014666170317113606] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/08/2017] [Accepted: 03/13/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disorder, neuropathologically characterized by aggregates of β-amyloid peptides, which deposit as senile plaques, and of TAU protein, which forms neurofibrillary tangles. It is now widely accepted that neuroinflammation is implicated in AD pathogenesis. METHOD Indeed, inflammatory mediators, such as cytokines and chemokines (chemotactic cytokines) can impact on the Alzheimer´s amyloid precursor protein by affecting its expression levels and amyloidogenic processing and/or β -amyloid aggregation. Additionally, cytokines and chemokines can influence kinases' activities, leading to abnormal TAU phosphorylation. To date there is no cure for AD, but several therapeutic strategies have been directed to prevent neuroinflammation. Anti-inflammatory, but also anti-amyloidogenic compounds, such as flavonoids were shown to favourably modulate some pathological events associated with neurodegeneration. CONCLUSION This review focuses on the role of cytokines and chemokines in AD-associated pathologies, and summarizes the potential anti-inflammatory therapeutic approaches aimed at preventing or slowing down disease progression.
Collapse
Affiliation(s)
- Catarina Domingues
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193Aveiro, Portugal
| | - Odete A.B. Cruz e Silva
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193Aveiro, Portugal
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Laboratory, Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193Aveiro, Portugal
| |
Collapse
|
25
|
Zhou M, Greenhill S, Huang S, Silva TK, Sano Y, Wu S, Cai Y, Nagaoka Y, Sehgal M, Cai DJ, Lee YS, Fox K, Silva AJ. CCR5 is a suppressor for cortical plasticity and hippocampal learning and memory. eLife 2016; 5. [PMID: 27996938 PMCID: PMC5213777 DOI: 10.7554/elife.20985] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 12/19/2016] [Indexed: 11/13/2022] Open
Abstract
Although the role of CCR5 in immunity and in HIV infection has been studied widely, its role in neuronal plasticity, learning and memory is not understood. Here, we report that decreasing the function of CCR5 increases MAPK/CREB signaling, long-term potentiation (LTP), and hippocampus-dependent memory in mice, while neuronal CCR5 overexpression caused memory deficits. Decreasing CCR5 function in mouse barrel cortex also resulted in enhanced spike timing dependent plasticity and consequently, dramatically accelerated experience-dependent plasticity. These results suggest that CCR5 is a powerful suppressor for plasticity and memory, and CCR5 over-activation by viral proteins may contribute to HIV-associated cognitive deficits. Consistent with this hypothesis, the HIV V3 peptide caused LTP, signaling and memory deficits that were prevented by Ccr5 knockout or knockdown. Overall, our results demonstrate that CCR5 plays an important role in neuroplasticity, learning and memory, and indicate that CCR5 has a role in the cognitive deficits caused by HIV.
Collapse
Affiliation(s)
- Miou Zhou
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Stuart Greenhill
- Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom.,School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Shan Huang
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Tawnie K Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yoshitake Sano
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Shumin Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, United States
| | - Ying Cai
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yoshiko Nagaoka
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, United States
| | - Megha Sehgal
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Denise J Cai
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yong-Seok Lee
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kevin Fox
- Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Alcino J Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
26
|
DHA-PC and DHA-PS improved Aβ1–40 induced cognitive deficiency uncoupled with an increase in brain DHA in rats. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.02.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
27
|
Hwang CJ, Park MH, Hwang JY, Kim JH, Yun NY, Oh SY, Song JK, Seo HO, Kim YB, Hwang DY, Oh KW, Han SB, Hong JT. CCR5 deficiency accelerates lipopolysaccharide-induced astrogliosis, amyloid-beta deposit and impaired memory function. Oncotarget 2016; 7:11984-11999. [PMID: 26910914 PMCID: PMC4914263 DOI: 10.18632/oncotarget.7453] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 02/05/2016] [Indexed: 11/25/2022] Open
Abstract
Chemokine receptors are implicated in inflammation and immune responses. Neuro-inflammation is associated with activation of astrocyte and amyloid-beta (Aβ) generations that lead to pathogenesis of Alzheimer disease (AD). Previous our study showed that deficiency of CC chemokine receptor 5 (CCR5) results in activation of astrocytes and Aβ deposit, and thus memory dysfunction through increase of CC chemokine receptor 2 (CCR2) expression. CCR5 knockout mice were used as an animal model with memory dysfunction. For the purpose LPS was injected i.p. daily (0.25 mg/kg/day). The memory dysfunctions were much higher in LPS-injected CCR5 knockout mice compared to CCR5 wild type mice as well as non-injected CCR5 knockout mice. Associated with severe memory dysfuction in LPS injected CCR5 knockout mice, LPS injection significant increase expression of inflammatory proteins, astrocyte activation, expressions of β-secretase as well as Aβ deposition in the brain of CCR5 knockout mice as compared with that of CCR5 wild type mice. In CCR5 knockout mice, CCR2 expressions were high and co-localized with GFAP which was significantly elevated by LPS. Expression of monocyte chemoattractant protein-1 (MCP-1) which ligands of CCR2 also increased by LPS injection, and increment of MCP-1 expression is much higher in CCR5 knockout mice. BV-2 cells treated with CCR5 antagonist, D-ala-peptide T-amide (DAPTA) and cultured astrocytes isolated from CCR5 knockout mice treated with LPS (1 μg/ml) and CCR2 antagonist, decreased the NF-ĸB activation and Aβ level. These findings suggest that the deficiency of CCR5 enhances response of LPS, which accelerates to neuro-inflammation and memory impairment.
Collapse
Affiliation(s)
- Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Jae Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Ju Hwan Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Na Young Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Sang Yeon Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Ju Kyung Song
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyun Ok Seo
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Yun-Bae Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Dae Yeon Hwang
- College of Natural Resources and Life Science, Pusan National University, Pusan, Republic of Korea
| | - Ki-Wan Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
28
|
Lack of Neuronal IFN-β-IFNAR Causes Lewy Body- and Parkinson's Disease-like Dementia. Cell 2016; 163:324-39. [PMID: 26451483 PMCID: PMC4601085 DOI: 10.1016/j.cell.2015.08.069] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 05/20/2015] [Accepted: 08/13/2015] [Indexed: 11/24/2022]
Abstract
Neurodegenerative diseases have been linked to inflammation, but whether altered immunomodulation plays a causative role in neurodegeneration is not clear. We show that lack of cytokine interferon-β (IFN-β) signaling causes spontaneous neurodegeneration in the absence of neurodegenerative disease-causing mutant proteins. Mice lacking Ifnb function exhibited motor and cognitive learning impairments with accompanying α-synuclein-containing Lewy bodies in the brain, as well as a reduction in dopaminergic neurons and defective dopamine signaling in the nigrostriatal region. Lack of IFN-β signaling caused defects in neuronal autophagy prior to α-synucleinopathy, which was associated with accumulation of senescent mitochondria. Recombinant IFN-β promoted neurite growth and branching, autophagy flux, and α-synuclein degradation in neurons. In addition, lentiviral IFN-β overexpression prevented dopaminergic neuron loss in a familial Parkinson's disease model. These results indicate a protective role for IFN-β in neuronal homeostasis and validate Ifnb mutant mice as a model for sporadic Lewy body and Parkinson's disease dementia.
Collapse
|
29
|
Minter MR, Taylor JM, Crack PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer's disease. J Neurochem 2015; 136:457-74. [PMID: 26509334 DOI: 10.1111/jnc.13411] [Citation(s) in RCA: 323] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/11/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia. Deposition of amyloid-β (Aβ) remains a hallmark feature of the disease, yet the precise mechanism(s) by which this peptide induces neurotoxicity remain unknown. Neuroinflammation has long been implicated in AD pathology, yet its contribution to disease progression is still not understood. Recent evidence suggests that various Aβ complexes interact with microglial and astrocytic expressed pattern recognition receptors that initiate innate immunity. This process involves secretion of pro-inflammatory cytokines, chemokines and generation of reactive oxygen species that, in excess, drive a dysregulated immune response that contributes to neurodegeneration. The mechanisms by which a neuroinflammatory response can influence Aβ production, aggregation and eventual clearance are now becoming key areas where future therapeutic intervention may slow progression of AD. This review will focus on evidence supporting the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD, describing the key cell types, pathways and mediators involved. Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia worldwide. Deposition of intracellular plaques containing amyloid-beta (Aβ) is a hallmark proteinopathy of the disease yet the precise mechanisms by which this peptide induces neurotoxicity remains unknown. A neuroinflammatory response involving polarized microglial activity, enhanced astrocyte reactivity and elevated pro-inflammatory cytokine and chemokine load has long been implicated in AD and proposed to facilitate neurodegeneration. In this issue we discuss key receptor systems of innate immunity that detect Aβ, drive pro-inflammatory cytokine and chemokine production and influence Aβ aggregation and clearance. Evidence summarized in this review supports the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD and highlights the potential of immunomodulatory agents as potential future therapies for AD patients.
Collapse
Affiliation(s)
- Myles R Minter
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Juliet M Taylor
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Marciniak E, Faivre E, Dutar P, Alves Pires C, Demeyer D, Caillierez R, Laloux C, Buée L, Blum D, Humez S. The Chemokine MIP-1α/CCL3 impairs mouse hippocampal synaptic transmission, plasticity and memory. Sci Rep 2015; 5:15862. [PMID: 26511387 PMCID: PMC4625372 DOI: 10.1038/srep15862] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/05/2015] [Indexed: 11/20/2022] Open
Abstract
Chemokines are signaling molecules playing an important role in immune regulations. They are also thought to regulate brain development, neurogenesis and neuroendocrine functions. While chemokine upsurge has been associated with conditions characterized with cognitive impairments, their ability to modulate synaptic plasticity remains ill-defined. In the present study, we specifically evaluated the effects of MIP1-α/CCL3 towards hippocampal synaptic transmission, plasticity and spatial memory. We found that CCL3 (50 ng/ml) significantly reduced basal synaptic transmission at the Schaffer collateral-CA1 synapse without affecting NMDAR-mediated field potentials. This effect was ascribed to post-synaptic regulations, as CCL3 did not impact paired-pulse facilitation. While CCL3 did not modulate long-term depression (LTD), it significantly impaired long-term potentiation (LTP), an effect abolished by Maraviroc, a CCR5 specific antagonist. In addition, sub-chronic intracerebroventricular (icv) injections of CCL3 also impair LTP. In accordance with these electrophysiological findings, we demonstrated that the icv injection of CCL3 in mouse significantly impaired spatial memory abilities and long-term memory measured using the two-step Y-maze and passive avoidance tasks. These effects of CCL3 on memory were inhibited by Maraviroc. Altogether, these data suggest that the chemokine CCL3 is an hippocampal neuromodulator able to regulate synaptic plasticity mechanisms involved in learning and memory functions.
Collapse
Affiliation(s)
- Elodie Marciniak
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - Emilie Faivre
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - Patrick Dutar
- Centre de Psychiatrie et Neurosciences, UMR_S 894, Faculté de Médecine, Université Paris Descartes, 75014, Paris
| | - Claire Alves Pires
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - Dominique Demeyer
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - Raphaëlle Caillierez
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - Charlotte Laloux
- Université de Lille, F-59000 Lille, France
- SFR DN2M, 59000 Lille France
| | - Luc Buée
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - David Blum
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| | - Sandrine Humez
- Université de Lille, F-59000 Lille, France
- Inserm UMR_S1172, Jean-Pierre Aubert Research Centre, F-59000 Lille France
- CHU-Lille, F-59000 Lille France
| |
Collapse
|
31
|
Lee DH, Park MH, Hwang CJ, Hwang JY, Yoon HS, Yoon DY, Hong JT. CCR5 deficiency increased susceptibility to lipopolysaccharide-induced acute renal injury. Arch Toxicol 2015; 90:1151-62. [PMID: 26055553 DOI: 10.1007/s00204-015-1530-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 05/05/2015] [Indexed: 01/08/2023]
Abstract
C-C chemokine receptor 5 (CCR5) regulates leukocyte chemotaxis and activation, and its deficiency exacerbates development of nephritis. Therefore, we investigated the role of CCR5 during lipopolysaccharide (LPS)-induced acute kidney injury. CCR5-deficient (CCR5-/-) and wild-type (CCR5+/+) mice, both aged about 10 months, had acute renal injury induced by intraperitoneal injection of LPS (10 mg/kg). Compared with CCR5+/+ mice, CCR5-/- mice showed increased mortality and renal injury, including elevated creatinine and blood urea nitrogen levels, following LPS challenge. Compared to CCR5+/+ mice, CCR5-/- mice also exhibited greater increases in the serum concentrations of pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β following LPS challenge. Furthermore, infiltration of macrophages and neutrophils, expression of intracellular adhesion molecule (ICAM)-1, and the number of apoptotic cells were more greatly increased by LPS treatment in CCR5-/- mice than in CCR5+/+ mice. The concentrations of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β were also significantly increased in the kidney of CCR5-/- mice after LPS challenge. Moreover, primary kidney cells from CCR5-/- mice showed greater increases in TNF-α production and p38 MAP kinase activation following treatment with LPS compared with that observed in the cells from CCR5+/+ mice. LPS-induced TNF-α production and apoptosis in the primary kidney cells from CCR5-/- mice were inhibited by treatment with p38 MAP kinase inhibitor. These results suggest that CCR5 deficiency increased the production of TNF-α following LPS treatment through increased activation of the p38 pathway in the kidney, resulting in renal apoptosis and leukocyte infiltration and led to exacerbation of LPS-induced acute kidney injury.
Collapse
Affiliation(s)
- Dong Hun Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Jae Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Hae Suk Yoon
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea
| | - Do Young Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, 1, Hwayang-dong, Gwangjin-gu, Seoul, 143-701, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| |
Collapse
|
32
|
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: 4166] [Impact Index Per Article: 416.6] [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
|
33
|
Janssen K, Rickert M, Clarner T, Beyer C, Kipp M. Absence of CCL2 and CCL3 Ameliorates Central Nervous System Grey Matter But Not White Matter Demyelination in the Presence of an Intact Blood-Brain Barrier. Mol Neurobiol 2015; 53:1551-1564. [PMID: 25663168 DOI: 10.1007/s12035-015-9113-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/22/2015] [Indexed: 11/25/2022]
Abstract
A broad spectrum of diseases is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation, among multiple sclerosis (MS). Our knowledge regarding the factors triggering gliosis and demyelination is scanty. Chemokines are pivotal for microglia and astrocyte activation and orchestrate critical steps during the formation of central nervous system (CNS) demyelinating lesions. Redundant functions of chemokines complicate, however, the study of their functional relevance. We used the cuprizone model to study redundant functions of two chemokines, CCL2/MCP1 and CCL3/MIP1α, which are critically involved in the pathological process of cuprizone-induced demyelination. First, we generated a mutant mouse strain lacking functional genes of both chemokines and demonstrated that double-mutant animals are viable, fertile, and do not present with gross abnormalities. Astrocytes and peritoneal macrophages, cultured form tissues of these animals did neither express CCL2 nor CCL3. Exposure to cuprizone resulted in increased CCL2 and CCL3 brain levels in wild-type but not mutant animals. Cuprizone-induced demyelination, oligodendrocyte loss, and astrogliosis were significantly ameliorated in the cortex but not corpus callosum of chemokine-deficient animals. In summary, we provide a novel powerful model to study the redundant function of two important chemokines. Our study reveals that chemokine function in the CNS redounds to region-specific pathophysiological events.
Collapse
Affiliation(s)
- Katharina Janssen
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mira Rickert
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Tim Clarner
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Markus Kipp
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany.
| |
Collapse
|
34
|
Krauthausen M, Kummer MP, Zimmermann J, Reyes-Irisarri E, Terwel D, Bulic B, Heneka MT, Müller M. CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model. J Clin Invest 2014; 125:365-78. [PMID: 25500888 DOI: 10.1172/jci66771] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/10/2014] [Indexed: 01/28/2023] Open
Abstract
Chemokines are important modulators of neuroinflammation and neurodegeneration. In the brains of Alzheimer's disease (AD) patients and in AD animal models, the chemokine CXCL10 is found in high concentrations, suggesting a pathogenic role for this chemokine and its receptor, CXCR3. Recent studies aimed at addressing the role of CXCR3 in neurological diseases indicate potent, but diverse, functions for CXCR3. Here, we examined the impact of CXCR3 in the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD. We found that, compared with control APP/PSI animals, plaque burden and Aβ levels were strongly reduced in CXCR3-deficient APP/PS1 mice. Analysis of microglial phagocytosis in vitro and in vivo demonstrated that CXCR3 deficiency increased the microglial uptake of Aβ. Application of a CXCR3 antagonist increased microglial Aβ phagocytosis, which was associated with reduced TNF-α secretion. Moreover, in CXCR3-deficient APP/PS1 mice, microglia exhibited morphological activation and reduced plaque association, and brain tissue from APP/PS1 animals lacking CXCR3 had reduced concentrations of proinflammatory cytokines compared with controls. Further, loss of CXCR3 attenuated the behavioral deficits observed in APP/PS1 mice. Together, our data indicate that CXCR3 signaling mediates development of AD-like pathology in APP/PS1 mice and suggest that CXCR3 has potential as a therapeutic target for AD.
Collapse
|
35
|
Azizi G, Khannazer N, Mirshafiey A. The Potential Role of Chemokines in Alzheimer's Disease Pathogenesis. Am J Alzheimers Dis Other Demen 2014; 29:415-25. [PMID: 24408754 PMCID: PMC10852600 DOI: 10.1177/1533317513518651] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and leading cause of dementia, which begins with impaired memory. The neuropathological hallmarks of AD include destructive alterations of neurons by neurofibrillary tangles, neuritic amyloid plaques, and neuroinflammatory process in the brain. Chemokines have a major role in inflammatory cell attraction and glial cell activation and/or modulation in the central nervous system. Moreover, the clinical and immunopathological evidence could show dual key role of chemokines in their pro- and anti-inflammatory properties in AD. However, their effects in neurodegeneration and/or neuroprotection remain an area of investigation. This review article provides an overview of characteristic, cellular source and activity of chemokines, and their roles in neuronal glial cell interaction in AD.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Nikoo Khannazer
- Department of Molecular and Cellular Biology, College of Science, University of Tehran, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Goldeck D, Larbi A, Pellicanó M, Alam I, Zerr I, Schmidt C, Fulop T, Pawelec G. Enhanced Chemokine Receptor Expression on Leukocytes of Patients with Alzheimer's Disease. PLoS One 2013; 8:e66664. [PMID: 23824053 PMCID: PMC3688934 DOI: 10.1371/journal.pone.0066664] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/08/2013] [Indexed: 01/23/2023] Open
Abstract
Although primarily a neurological complaint, systemic inflammation is present in Alzheimer's Disease, with higher than normal levels of proinflammatory cytokines and chemokines in the periphery as well as the brain. A gradient of these factors may enhance recruitment of activated immune cells into the brain via chemotaxis. Here, we investigated the phenotypes of circulating immune cells in AD patients with multi-colour flow cytometry to determine whether their expression of chemokine receptors is consistent with this hypothesis. In this study, we confirmed our previously reported data on the shift of early- to late-differentiated CD4+ T-cells in AD patients. The percentage of cells expressing CD25, a marker of acute T-cell activation, was higher in patients than in age-matched controls, and percentages of CCR6+ cells were elevated. This chemokine receptor is primarily expressed on pro-inflammatory memory cells and Th17 cells. The proportion of cells expressing CCR4 (expressed on Th2 cells) and CCR5 (Th1 cells and dendritic cells) was also greater in patients, and was more pronounced on CD4+ than CD8+ T-cells. These findings allow a more detailed insight into the systemic immune status of patients with Alzheimer's disease and suggest possible novel targets for immune therapy.
Collapse
Affiliation(s)
- David Goldeck
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
- * E-mail:
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research, Biopolis, Singapore, Singapore
| | - Mariavaleria Pellicanó
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
| | - Iftikhar Alam
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
- Abdul Wali Khan University, Mardan, Pakistan
| | - Inga Zerr
- Clinical Dementia Center, Dept. of Neurology, University Medical School, Georg August University, Göttingen, Germany
| | - Christian Schmidt
- Clinical Dementia Center, Dept. of Neurology, University Medical School, Georg August University, Göttingen, Germany
| | - Tamas Fulop
- Immunology Program, Geriatric Division, Faculty of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Graham Pawelec
- Tübingen Ageing and Tumour Immunology Group, Center for Medical Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
37
|
Choi DY, Lee MK, Hong JT. Lack of CCR5 modifies glial phenotypes and population of the nigral dopaminergic neurons, but not MPTP-induced dopaminergic neurodegeneration. Neurobiol Dis 2012; 49:159-68. [PMID: 22922220 DOI: 10.1016/j.nbd.2012.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/12/2012] [Accepted: 08/02/2012] [Indexed: 11/17/2022] Open
Abstract
Constitutive expression of C-C chemokine receptor (CCR) 5 has been detected in astrocytes, microglia and neurons, but its physiological roles in the central nervous system are obscure. The bidirectional interactions between neuron and glial cells through CCR5 and its ligands were thought to be crucial for maintaining normal neuronal activities. No study has described function of CCR5 in the dopaminergic neurodegeneration in Parkinson's disease. In order to examine effects of CCR5 on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neurodegeneration, we employed CCR5 wild type (WT) and knockout (KO) mice. Immunostainings for tyrosine hydroxylase (TH) exhibited that CCR5 KO mice had lower number of TH-positive neurons even in the absence of MPTP. Difference in MPTP (15mg/kg×4 times, 2hr interval)-mediated loss of TH-positive neurons was subtle between CCR5 WT and KO mice, but there was larger dopamine depletion, behavioral impairments and microglial activation in CCR5 deficient mice. Intriguingly, CCR5 KO brains contained higher immunoreactivity for monoamine oxidase (MAO) B which was mainly localized within astrocytes. In agreement with upregulation of MAO B, concentration of MPP+ was higher in the substantia nigra and striatum of CCR5 KO mice after MPTP injection. We found remarkable activation of p38 MAPK in CCR5 deficient mice, which positively regulates MAO B expression. These results indicate that CCR5 deficiency modifies the nigrostriatal dopaminergic neuronal system and bidirectional interaction between neurons and glial cells via CCR5 might be important for dopaminergic neuronal survival.
Collapse
Affiliation(s)
- Dong-Young Choi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea; College of Pharmacy, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea; Medical Research Center, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Myung Koo Lee
- College of Pharmacy, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea; Medical Research Center, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea.
| |
Collapse
|
38
|
Abstract
While immune responses in neurodegeneration were regarded as little more than a curiosity a decade ago, they are now increasingly moving toward center stage. Factors driving this movement include the recognition that most of the relevant immune molecules are produced within the brain, that microglia are proficient immune cells shaping neuronal circuitry and fate, and that systemic immune responses affect brain function. We will review this complex field from the perspective of neurons, extra-neuronal brain cells, and the systemic environment and highlight the possibility that cell intrinsic innate immune molecules in neurons may function in neurodegenerative processes.
Collapse
Affiliation(s)
- Eva Czirr
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5489, USA
| | | |
Collapse
|
39
|
Genetic knockouts suggest a critical role for HIV co-receptors in models of HIV gp120-induced brain injury. J Neuroimmune Pharmacol 2011; 7:306-18. [PMID: 22124968 DOI: 10.1007/s11481-011-9328-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 11/16/2011] [Indexed: 02/06/2023]
Abstract
Infection with HIV-1 frequently affects the brain and causes NeuroAIDS prior to the development of overt AIDS. The HIV-1 envelope protein gp120 interacts with host CD4 and chemokine co-receptors to initiate infection of macrophages and lymphocytes. In addition, the virus or fragments of it, such as gp120, cause macrophages to produce neurotoxins and trigger neuronal injury and apoptosis. Moreover, the two major HIV co-receptors, the chemokine receptors CCR5 and CXCR4, serve numerous physiological functions and are widely expressed beyond immune cells, including cells in the brain. Therefore, HIV co-receptors are poised to play a direct and indirect part in the development of NeuroAIDS. Although rodents are not permissive to infection with wild type HIV-1, viral co-receptors - more than CD4 - are highly conserved between species, suggesting the animals can be suitable models for mechanistic studies addressing effects of receptor-ligand interaction other than infection. Of note, transgenic mice expressing HIV gp120 in the brain share several pathological hallmarks with NeuroAIDS brains. Against this background, we will discuss recently completed or initiated, ongoing studies that utilize HIV co-receptor knockout and viral gp120-transgenic mice as models for in vitro and in vivo experimentation in order to address the potential roles of HIV gp120 and its co-receptors in the development of NeuroAIDS.
Collapse
|
40
|
The CC chemokine receptor 5 regulates olfactory and social recognition in mice. Neuroscience 2011; 197:153-61. [PMID: 21963866 DOI: 10.1016/j.neuroscience.2011.09.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 09/19/2011] [Accepted: 09/19/2011] [Indexed: 12/12/2022]
Abstract
Chemokines are chemotactic cytokines that regulate cell migration and are thought to play an important role in a broad range of inflammatory diseases. The availability of chemokine receptor blockers makes them an important therapeutic target. In vitro, chemokines are shown to modulate neurotransmission. However, it is not very clear if chemokines play a role in behavior and cognition. Here we evaluated the role of CC chemokine receptor 5 (CCR5) in various behavioral tasks in mice using Wt (Ccr5⁺/⁺) and Ccr5-null (Ccr5⁻/⁻)mice. Ccr5⁻/⁻ mice showed enhanced social recognition. Administration of CC chemokine ligand 3 (CCL3), one of the CCR5-ligands, impaired social recognition. Since the social recognition task is dependent on the sense of olfaction, we tested olfactory recognition for social and non-social scents in these mice. Ccr5⁻/⁻ mice had enhanced olfactory recognition for both these scents indicating that enhanced performance in social recognition task could be due to enhanced olfactory recognition in these mice. Spatial memory and aversive memory were comparable in Wt and Ccr5⁻/⁻ mice. Collectively, these results suggest that chemokines/chemokine receptors might play an important role in olfactory recognition tasks in mice and to our knowledge represents the first direct demonstration of an in vivo role of CCR5 in modulating social behavior in mice. These studies are important as CCR5 blockers are undergoing clinical trials and can potentially modulate behavior.
Collapse
|
41
|
Derecki NC, Quinnies KM, Kipnis J. Alternatively activated myeloid (M2) cells enhance cognitive function in immune compromised mice. Brain Behav Immun 2011; 25:379-85. [PMID: 21093578 PMCID: PMC3039052 DOI: 10.1016/j.bbi.2010.11.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 01/23/2023] Open
Abstract
It was recently shown that adaptive immunity plays a key role in cognitive function. T cells appear to be major players in learning and memory; thus, mice devoid of functional T cells are impaired in performance of cognitive tasks such as Morris water maze (MWM), Barnes maze and others. This is a reversible phenomenon; injection of immune deficient mice with T cells from wild type counterparts improves their cognitive function. Recently we described a critical role for T cell-derived IL-4 as having beneficial effects on learning and memory through regulation of meningeal myeloid cell phenotype. In the absence of IL-4, meningeal myeloid cells acquire a pro-inflammatory skew. Thus, the presence of IL-4 in the meningeal spaces maintains a delicate balance of pro- and anti-inflammatory myeloid cell phenotype. Here we show that macrophages alternatively activated in vitro (M2 cells) can circumvent the need for 'pro-cognitive' T cells when injected intravenously into immune deficient mice. These results show for the first time that M2 myeloid cells are new and unexpected players in cognitive function, conferring beneficial effects on learning and memory without adaptive immune influence. These results might lead to development of new therapeutic approaches for cognitive pathologies associated with malfunction of adaptive immunity, such as chemo-brain, age-related dementia, HIV-dementia, and others.
Collapse
Affiliation(s)
- Noel C Derecki
- Graduate Program in Neuroscience, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
42
|
Sorce S, Myburgh R, Krause KH. The chemokine receptor CCR5 in the central nervous system. Prog Neurobiol 2010; 93:297-311. [PMID: 21163326 DOI: 10.1016/j.pneurobio.2010.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 11/29/2010] [Accepted: 12/07/2010] [Indexed: 02/04/2023]
Abstract
The expression and the role of the chemokine receptor CCR5 have been mainly studied in the context of HIV infection. However, this protein is also expressed in the brain, where it can be crucial in determining the outcome in response to different insults. CCR5 expression can be deleterious or protective in controlling the progression of certain infections in the CNS, but it is also emerging that it could play a role in non-infectious diseases. In particular, it appears that, in addition to modulating immune responses, CCR5 can influence neuronal survival. Here, we summarize the present knowledge about the expression of CCR5 in the brain and highlight recent findings suggesting its possible involvement in neuroprotective mechanisms.
Collapse
Affiliation(s)
- Silvia Sorce
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva-4, Switzerland
| | | | | |
Collapse
|
43
|
Lee HJ, Lee JK, Lee H, Shin JW, Carter JE, Sakamoto T, Jin HK, Bae JS. The therapeutic potential of human umbilical cord blood-derived mesenchymal stem cells in Alzheimer's disease. Neurosci Lett 2010; 481:30-5. [PMID: 20600610 DOI: 10.1016/j.neulet.2010.06.045] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 06/09/2010] [Accepted: 06/15/2010] [Indexed: 12/15/2022]
Abstract
The neuropathological hallmarks of Alzheimer's disease (AD) include the presence of extracellular amyloid-beta peptide (Abeta) in the form of amyloid plaques in the brain parenchyma and neuronal loss. The mechanism associated with neuronal death by amyloid plaques is unclear but oxidative stress and glial activation has been implicated. Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) are being scrutinized as a potential therapeutic tool to prevent various neurodegenerative diseases including AD. However, the therapeutic impact of hUCB-MSCs in AD has not yet been reported. Here we undertook in vitro work to examine the potential impact of hUCB-MSCs treatment on neuronal loss using a paradigm of cultured hippocampal neurons treated with Abeta. We confirmed that hUCB-MSCs co-culture reduced the hippocampal apoptosis induced by Abeta treatment. Moreover, in an acute AD mouse model to directly test the efficacy of hUCB-MSCs treatment on AD-related cognitive and neuropathological outcomes, we demonstrated that markers of glial activation, oxidative stress and apoptosis levels were decreased in AD mouse brain. Interestingly, hUCB-MSCs treated AD mice demonstrated cognitive rescue with restoration of learning/memory function. These data suggest that hUCB-MSCs warrant further investigation as a potential therapeutic agent in AD.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Kim TI, Lee YK, Park SG, Choi IS, Ban JO, Park HK, Nam SY, Yun YW, Han SB, Oh KW, Hong JT. l-Theanine, an amino acid in green tea, attenuates beta-amyloid-induced cognitive dysfunction and neurotoxicity: reduction in oxidative damage and inactivation of ERK/p38 kinase and NF-kappaB pathways. Free Radic Biol Med 2009; 47:1601-10. [PMID: 19766184 DOI: 10.1016/j.freeradbiomed.2009.09.008] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/19/2009] [Accepted: 09/06/2009] [Indexed: 11/18/2022]
Abstract
Amyloid beta (Abeta)-induced neurotoxicity is a major pathological mechanism of Alzheimer disease (AD). In this study, we investigated the inhibitory effect of l-theanine, a component of green tea (Camellia sinensis), on Abeta(1-42)-induced neuronal cell death and memory impairment. Oral treatment of l-theanine (2 and 4 mg/kg) for 5 weeks in the drinking water of mice, followed by injection of Abeta(1-42) (2 microg/mouse, icv), significantly attenuated Abeta(1-42)-induced memory impairment. Furthermore, l-theanine reduced Abeta(1-42) levels and the accompanying Abeta(1-42)-induced neuronal cell death in the cortex and hippocampus of the brain. Moreover, l-theanine inhibited Abeta(1-42)-induced extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase as well as the activity of nuclear factor kappaB (NF-kappaB). l-Theanine also significantly reduced oxidative protein and lipid damage and the elevation of glutathione levels in the brain. These data suggest that the positive effects of l-theanine on memory may be mediated by suppression of ERK/p38 and NF-kappaB as well as the reduction of macromolecular oxidative damage. Thus, l-theanine may be useful in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Tae Il Kim
- College of Pharmacy and CBITRC, Chungbuk National University, Cheongju, Chungbuk 361-763, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|