1
|
Gutiérrez IL, Martín-Hernández D, MacDowell KS, García-Bueno B, Caso JR, Leza JC, Madrigal JLM. CX3CL1 Regulation of Gliosis in Neuroinflammatory and Neuroprotective Processes. Int J Mol Sci 2025; 26:959. [PMID: 39940727 PMCID: PMC11817243 DOI: 10.3390/ijms26030959] [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: 12/19/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Among the different chemokines, C-X3-C motif chemokine ligand 1 or CX3CL1, also named fractalkine, is one of the most interesting due to its characteristics, including its unique structure, not shared by any other chemokine, and its ability to function both in a membrane-bound form and in a soluble form, among others. However, undoubtedly, its most relevant characteristic from the neuroscientific point of view is its role as a messenger used by neurons to communicate with microglia. The study of the interaction between both cell types and the key role that CX3CL1 seems to play has facilitated the identification of CX3CL1 as a crucial modulator of microglial activation and a promising target in the fight against neuroinflammation. As a result, numerous studies have contributed to elucidate the involvement of CX3CL1 and its specific receptor CCX3CR1 in the progression of different neuroinflammatory and neurodegenerative processes, with Alzheimer's and Parkinson's diseases being the most studied ones. However, the different animal and cellular models used to reproduce the pathological conditions to be analyzed, as well as the difficulties inherent to studies performed on human samples, have hindered the collection of compatible results in many cases. In this review, we summarize some of the most relevant data describing the alterations found for the CX3CL1/CX3CR1 signaling axis in different neurodegenerative conditions in which neuroinflammation is known to play a relevant role.
Collapse
Affiliation(s)
| | | | | | | | | | | | - José L. M. Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Av. Complutense s/n, 28040, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Neuroquímica (IUINQ-UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), 28040 Madrid, Spain
| |
Collapse
|
2
|
Zhan L, Qiu M, Zheng J, Lai M, Lin K, Dai J, Sun W, Xu E. Fractalkine/CX3CR1 axis is critical for neuroprotection induced by hypoxic postconditioning against cerebral ischemic injury. Cell Commun Signal 2024; 22:457. [PMID: 39327578 PMCID: PMC11426015 DOI: 10.1186/s12964-024-01830-4] [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: 04/29/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Microglial activation-mediated neuroinflammation is a major contributor to neuronal damage after cerebral ischemia. The Fractalkine (FKN)/CX3C chemokine receptor 1 (CX3CR1) axis plays a critical role in regulating microglial activation and neuroinflammation. The aim of this study is to ascertain the role and mechanism of FKN/CX3CR1 axis in hypoxic postconditioning (HPC)-induced anti-inflammatory and neuroprotective effects on transient global cerebral ischemia (tGCI). We found that HPC suppressed microglial activation and alleviated neuroinflammation in hippocampal CA1 after tGCI. Meanwhile, HPC upregulated the expression of FKN and CX3CR1 in neurons, but it downregulated the expression of CX3CR1 in glial cells after tGCI. In addition, the overexpression of FKN induced by the administration of FKN-carried lentivirus reduced microglial activation and inhibited neuroinflammation in CA1 after tGCI. Furthermore, silencing CX3CR1 with CX3CRi-carried lentivirus in CA1 after tGCI suppressed microglial activation and neuroinflammation and exerted neuroprotective effects. Finally, the overexpression of FKN caused a marked increase of neuronal CX3CR1 receptors, upregulated the phosphorylation of Akt, and reduced neuronal loss of rats in CA1 after tGCI. These findings demonstrated that HPC protected against neuronal damage in CA1 of tGCI rats through inhibiting microglial activation and activating Akt signaling pathway via FKN/CX3CR1 axis.
Collapse
Affiliation(s)
- Lixuan Zhan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Meiqian Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Jianhua Zheng
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Meijing Lai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Kunqin Lin
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Jiahua Dai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - Weiwen Sun
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China
| | - En Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong RD, Guangzhou, 510260, P. R. China.
| |
Collapse
|
3
|
Kalayasiri R, Dadwat K, Thika S, Sirivichayakul S, Maes M. Methamphetamine (MA) use and MA-induced psychosis are associated with increasing aberrations in the compensatory immunoregulatory system, interleukin-1α, and CCL5 levels. Transl Psychiatry 2023; 13:361. [PMID: 37996407 PMCID: PMC10667231 DOI: 10.1038/s41398-023-02645-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/25/2023] Open
Abstract
There are only a few studies reporting on the immunological profiles of methamphetamine (MA) use, MA dependency, or MA-induced psychosis (MAP). This study measured M1 macrophage, T helper (Th)-1, Th-2, growth factor, and chemokine profiles, as well as the immune inflammatory response system (IRS) and compensatory immunoregulatory system (CIRS) in peripheral blood samples from patients with MA use (n = 51), MA dependence (n = 47), and MAP (n = 43) in comparison with controls (n = 32). We discovered that persistent MA use had a robust immunosuppressive impact on all immunological profiles. The most reliable biomarker profile of MA use is the combination of substantial CIRS suppression and a rise in selected pro-inflammatory cytokines, namely CCL27 (CTACK), CCL11 (eotaxin), and interleukin (IL)-1α. In addition, MA dependency is associated with increased immunosuppression, as demonstrated by lower stem cell factor levels and higher IL-10 levels. MAP is related to a significant decrease in all immunological profiles, particularly CIRS, and an increase in CCL5 (RANTES), IL-1α, and IL-12p70 signaling. In conclusion, long-term MA use and dependency severely undermine immune homeostasis, whereas MAP may be the consequence of increased IL-1α - CCL5 signaling superimposed on strongly depleted CIRS and Th-1 functions. The widespread immunosuppression established in longstanding MA use may increase the likelihood of infectious and immune illness or exacerbate disorders such as hepatitis and AIDS. Furthermore, elevated levels of CCL5, CCL11, CCL27, IL-1α, and/or IL-12p70 may play a role in the peripheral (atherosclerosis, cutaneous inflammation, immune aberrations, hypospermatogenesis) and central (neuroinflammation, neurotoxic, neurodegenerative, depression, anxiety, and psychosis) side effects of MA use.
Collapse
Affiliation(s)
- Rasmon Kalayasiri
- Department of Psychiatry, Epidemiology of Psychiatric Disorders and Mental Health Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Kanokwan Dadwat
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Supaksorn Thika
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Sunee Sirivichayakul
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Maes
- Department of Psychiatry, Epidemiology of Psychiatric Disorders and Mental Health Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.
- Cognitive Fitness and Technology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research Institute, Medical University Plovdiv, Plovdiv, Bulgaria.
- Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China.
| |
Collapse
|
4
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
5
|
Wang S, Bai J, Zhang YL, Lin QY, Han X, Qu WK, Zhang PF, Ge YS, Zhao Q, Li HH. CXCL1-CXCR2 signalling mediates hypertensive retinopathy by inducing macrophage infiltration. Redox Biol 2022; 56:102438. [PMID: 35981418 PMCID: PMC9418605 DOI: 10.1016/j.redox.2022.102438] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/04/2022] Open
Abstract
Inflammation plays an important role in hypertensive retinal vascular injury and subsequent retinopathy. Monocyte chemotaxis via CXCL1-CXCR2 binding has been implicated in various cardiovascular diseases, but the function of CXCL1-CXCR2 signalling involved in retinopathy, which was investigated as angiotensin II (Ang II)-induced retinopathy, is unclear. In our study, we established a hypertensive retinopathy (HR) model by Ang II infusion (3000 ng/min/kg) for 3 weeks. To determine the involvement of CXCR2 signalling, we used CXCR2 knockout (KO) mice or C57BL/6J wild-type (WT) mice as experimental subjects. The mice were treated with a CXCL1 neutralizing antibody or SB225002 (the specific CXCR2 inhibitor). Our results showed that after Ang II treatment, the mRNA levels of CXCL1 and CXCR2 and the number of CXCR2+ inflammatory cells were significantly elevated. Conversely, unlike in the IgG control group, the CXCL1 neutralizing antibody greatly reduced the increase in central retinal thickness induced by Ang II infusion, arteriolar remodelling, superoxide production, and retinal dysfunction in WT mice. Furthermore, Ang II infusion induced arteriolar remodelling, infiltration of Iba1+ macrophages, the production of oxidative stress, and retinal dysfunction, but the symptoms were ameliorated in CXCR2 KO mice and SB225002-treated mice. These protective effects were related to the reduction in the number of CXCR2+ immune cells, particularly macrophages, and the decrease in proinflammatory cytokine (IL-1β, IL-6, TNF-ɑ, and MCP-1) expression in Ang II-treated retinas. Notably, serum CXCL1 levels and the number of CXCR2+ monocytes/neutrophils were higher in HR patients than in healthy controls. In conclusion, this study provides new evidence that the CXCL1-CXCR2 axis plays a vital role in the pathogenesis of hypertensive retinopathy, and selective blockade of CXCL1-CXCR2 activation may be a potential treatment for HR.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Ophthalmology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Jie Bai
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, 116004, China
| | - Yun-Long Zhang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Qiu-Yue Lin
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Xiao Han
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Wei-Kun Qu
- Department of Ophthalmology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Peng-Fei Zhang
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, 116024, China
| | - Yu-Song Ge
- Department of Ophthalmology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qi Zhao
- Department of Ophthalmology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
6
|
A CCR5 antagonist, maraviroc, alleviates neural circuit dysfunction and behavioral disorders induced by prenatal valproate exposure. J Neuroinflammation 2022; 19:195. [PMID: 35906621 PMCID: PMC9335995 DOI: 10.1186/s12974-022-02559-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/22/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Valproic acid (VPA) is a clinically used antiepileptic drug, but it is associated with a significant risk of a low verbal intelligence quotient (IQ) score, attention-deficit hyperactivity disorder and autism spectrum disorder in children when it is administered during pregnancy. Prenatal VPA exposure has been reported to affect neurogenesis and neuronal migration and differentiation. In addition, growing evidence has shown that microglia and brain immune cells are activated by VPA treatment. However, the role of VPA-activated microglia remains unclear. METHODS Pregnant female mice received sodium valproate on E11.5. A microglial activation inhibitor, minocycline or a CCR5 antagonist, maraviroc was dissolved in drinking water and administered to dams from P1 to P21. Measurement of microglial activity, evaluation of neural circuit function and expression analysis were performed on P10. Behavioral tests were performed in the order of open field test, Y-maze test, social affiliation test and marble burying test from the age of 6 weeks. RESULTS Prenatal exposure of mice to VPA induced microglial activation and neural circuit dysfunction in the CA1 region of the hippocampus during the early postnatal periods and post-developmental defects in working memory and social interaction and repetitive behaviors. Minocycline, a microglial activation inhibitor, clearly suppressed the above effects, suggesting that microglia elicit neural dysfunction and behavioral disorders. Next-generation sequencing analysis revealed that the expression of a chemokine, C-C motif chemokine ligand 3 (CCL3), was upregulated in the hippocampi of VPA-treated mice. CCL3 expression increased in microglia during the early postnatal periods via an epigenetic mechanism. The CCR5 antagonist maraviroc significantly suppressed neural circuit dysfunction and post-developmental behavioral disorders induced by prenatal VPA exposure. CONCLUSION These findings suggest that microglial CCL3 might act during development to contribute to VPA-induced post-developmental behavioral abnormalities. CCR5-targeting compounds such as maraviroc might alleviate behavioral disorders when administered early.
Collapse
|
7
|
He T, Yang GY, Zhang Z. Crosstalk of Astrocytes and Other Cells during Ischemic Stroke. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060910. [PMID: 35743941 PMCID: PMC9228674 DOI: 10.3390/life12060910] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Astrocytes structurally compose tripartite synapses, blood–brain barrier, and the neurovascular unit and perform multiple functions through cell-to-cell signaling of neurons, glial cells, and vasculature. The crosstalk of astrocytes and other cells is complicated and incompletely understood. Here we review the role of astrocytes in response to ischemic stroke, both beneficial and detrimental, from a cell–cell interaction perspective. Reactive astrocytes provide neuroprotection through antioxidation and antiexcitatory effects and metabolic support; they also contribute to neurorestoration involving neurogenesis, synaptogenesis, angiogenesis, and oligodendrogenesis by crosstalk with stem cells and cell lineage. In the meantime, reactive astrocytes also play a vital role in neuroinflammation and brain edema. Glial scar formation in the chronic phase hinders functional recovery. We further discuss astrocyte enriched microRNAs and exosomes in the regulation of ischemic stroke. In addition, the latest notion of reactive astrocyte subsets and astrocytic activity revealed by optogenetics is mentioned. This review discusses the current understanding of the intimate molecular conversation between astrocytes and other cells and outlines its potential implications after ischemic stroke. “Neurocentric” strategies may not be sufficient for neurological protection and recovery; future therapeutic strategies could target reactive astrocytes.
Collapse
Affiliation(s)
- Tingting He
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China;
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| |
Collapse
|
8
|
Déry L, Charest G, Guérin B, Akbari M, Fortin D. Chemoattraction of Neoplastic Glial Cells with CXCL10, CCL2 and CCL11 as a Paradigm for a Promising Therapeutic Approach for Primary Brain Tumors. Int J Mol Sci 2021; 22:ijms222212150. [PMID: 34830041 PMCID: PMC8626037 DOI: 10.3390/ijms222212150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022] Open
Abstract
Chemoattraction is a normal and essential process, but it can also be involved in tumorigenesis. This phenomenon plays a key role in glioblastoma (GBM). The GBM tumor cells are extremely difficult to eradicate, due to their strong capacity to migrate into the brain parenchyma. Consequently, a complete resection of the tumor is rarely a possibility, and recurrence is inevitable. To overcome this problem, we proposed to exploit this behavior by using three chemoattractants: CXCL10, CCL2 and CCL11, released by a biodegradable hydrogel (GlioGel) to produce a migration of tumor cells toward a therapeutic trap. To investigate this hypothesis, the agarose drop assay was used to test the chemoattraction capacity of these three chemokines on murine F98 and human U87MG cell lines. We then studied the potency of this approach in vivo in the well-established syngeneic F98-Fischer glioma-bearing rat model using GlioGel containing different mixtures of the chemoattractants. In vitro assays resulted in an invasive cell rate 2-fold higher when chemokines were present in the environment. In vivo experiments demonstrated the capacity of these specific chemoattractants to strongly attract neoplastic glioblastoma cells. The use of this strong locomotion ability to our end is a promising avenue in the establishment of a new therapeutic approach in the treatment of primary brain tumors.
Collapse
Affiliation(s)
- Laurence Déry
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Correspondence:
| | - Gabriel Charest
- Department of Surgery, Division of Neurosurgery, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (G.C.); (D.F.)
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Mohsen Akbari
- Laboratory for Innovation in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
| | - David Fortin
- Department of Surgery, Division of Neurosurgery, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (G.C.); (D.F.)
| |
Collapse
|
9
|
Mayhew JA, Cummins MJ, Cresswell ET, Callister RJ, Smith DW, Graham BA. Age-related gene expression changes in lumbar spinal cord: Implications for neuropathic pain. Mol Pain 2021; 16:1744806920971914. [PMID: 33241748 DOI: 10.1177/1744806920971914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Clinically, pain has an uneven incidence throughout lifespan and impacts more on the elderly. In contrast, preclinical models of pathological pain have typically used juvenile or young adult animals to highlight the involvement of glial populations, proinflammatory cytokines, and chemokines in the onset and maintenance of pathological signalling in the spinal dorsal horn. The potential impact of this mismatch is also complicated by the growing appreciation that the aged central nervous system exists in a state of chronic inflammation because of enhanced proinflammatory cytokine/chemokine signalling and glial activation. To address this issue, we investigated the impact of aging on the expression of genes that have been associated with neuropathic pain, glial signalling, neurotransmission and neuroinflammation. We used qRT-PCR to quantify gene expression and focussed on the dorsal horn of the spinal cord as this is an important perturbation site in neuropathic pain. To control for global vs region-specific age-related changes in gene expression, the ventral half of the spinal cord was examined. Our results show that expression of proinflammatory chemokines, pattern recognition receptors, and neurotransmitter system components was significantly altered in aged (24-32 months) versus young mice (2-4 months). Notably, the magnitude and direction of these changes were spinal-cord region dependent. For example, expression of the chemokine, Cxcl13, increased 119-fold in dorsal spinal cord, but only 2-fold in the ventral spinal cord of old versus young mice. Therefore, we propose the dorsal spinal cord of old animals is subject to region-specific alterations that prime circuits for the development of pathological pain, potentially in the absence of the peripheral triggers normally associated with these conditions.
Collapse
Affiliation(s)
- Jack A Mayhew
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Mitchell J Cummins
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Ethan T Cresswell
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Robert J Callister
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Doug W Smith
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Brett A Graham
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, and Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
10
|
Salemi M, Cannarella R, Marchese G, Salluzzo MG, Ravo M, Barone C, Cordella A, Caniglia S, Castiglione R, Ragalmuto A, Calogero AE, Romano C. CCR3 gene overexpression in patients with Down syndrome. Mol Biol Rep 2021; 48:5335-5338. [PMID: 34184200 DOI: 10.1007/s11033-021-06503-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/17/2021] [Indexed: 11/25/2022]
Abstract
Chromosome 21 trisomy or Down syndrome (DS) is the most common genetic cause of intellectual disability (ID). DS is also associated with hypotonia, muscle weakness, autoimmune diseases, and congenital heart disease. C-C chemokine receptor type 3 (CCR3) plays a role in inflammatory, autoimmune, and neuronal migration mechanisms. The present study aimed to evaluate the expression of the CCR3 gene by NGS and qRT-PCR in patients with DS and normal controls (NC). The CCR3 gene was over-expressed in DS patients compared to NC. These data suggest that an over-expression of the CCR3 gene is associated with the phenotype of patients with DS.
Collapse
Affiliation(s)
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | | | | | | | | | | | - Roberto Castiglione
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | |
Collapse
|
11
|
Cantalupo S, Lasorsa VA, Russo R, Andolfo I, D’Alterio G, Rosato BE, Frisso G, Abete P, Cassese GM, Servillo G, Gentile I, Piscopo C, Della Monica M, Fiorentino G, Russo G, Cerino P, Buonerba C, Pierri B, Zollo M, Iolascon A, Capasso M. Regulatory Noncoding and Predicted Pathogenic Coding Variants of CCR5 Predispose to Severe COVID-19. Int J Mol Sci 2021; 22:5372. [PMID: 34065289 PMCID: PMC8161088 DOI: 10.3390/ijms22105372] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Genome-wide association studies (GWAS) found locus 3p21.31 associated with severe COVID-19. CCR5 resides at the same locus and, given its known biological role in other infection diseases, we investigated if common noncoding and rare coding variants, affecting CCR5, can predispose to severe COVID-19. We combined single nucleotide polymorphisms (SNPs) that met the suggestive significance level (P ≤ 1 × 10-5) at the 3p21.31 locus in public GWAS datasets (6406 COVID-19 hospitalized patients and 902,088 controls) with gene expression data from 208 lung tissues, Hi-C, and Chip-seq data. Through whole exome sequencing (WES), we explored rare coding variants in 147 severe COVID-19 patients. We identified three SNPs (rs9845542, rs12639314, and rs35951367) associated with severe COVID-19 whose risk alleles correlated with low CCR5 expression in lung tissues. The rs35951367 resided in a CTFC binding site that interacts with CCR5 gene in lung tissues and was confirmed to be associated with severe COVID-19 in two independent datasets. We also identified a rare coding variant (rs34418657) associated with the risk of developing severe COVID-19. Our results suggest a biological role of CCR5 in the progression of COVID-19 as common and rare genetic variants can increase the risk of developing severe COVID-19 by affecting the functions of CCR5.
Collapse
Affiliation(s)
- Sueva Cantalupo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Vito Alessandro Lasorsa
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Roberta Russo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | | | - Barbara Eleni Rosato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Giulia Frisso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Pasquale Abete
- COVID Hospital, P.O.S. Anna e SS. Madonna della Neve di Boscotrecase, Ospedali Riuniti Area Vesuviana, 80042 Boscotrecase, Italy; (P.A.); (G.M.C.)
| | - Gian Marco Cassese
- COVID Hospital, P.O.S. Anna e SS. Madonna della Neve di Boscotrecase, Ospedali Riuniti Area Vesuviana, 80042 Boscotrecase, Italy; (P.A.); (G.M.C.)
| | - Giuseppe Servillo
- Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Ivan Gentile
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, A.O.R.N. ‘Antonio Cardarelli’, 80131 Napoli, Italy; (C.P.); (M.D.M.)
| | - Matteo Della Monica
- Medical and Laboratory Genetics Unit, A.O.R.N. ‘Antonio Cardarelli’, 80131 Napoli, Italy; (C.P.); (M.D.M.)
| | | | - Giuseppe Russo
- Unità di Radiologia, Casa di Cura Villa dei Fiori, 80011 Acerra, Italy;
| | - Pellegrino Cerino
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (P.C.); (C.B.); (B.P.)
| | - Carlo Buonerba
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (P.C.); (C.B.); (B.P.)
| | - Biancamaria Pierri
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (P.C.); (C.B.); (B.P.)
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana”, Università di Salerno, 84081 Baronissi, Italy
| | - Massimo Zollo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80136 Napoli, Italy; (S.C.); (V.A.L.); (R.R.); (I.A.); (B.E.R.); (G.F.); (M.Z.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy;
| |
Collapse
|
12
|
Lana D, Ugolini F, Nosi D, Wenk GL, Giovannini MG. The Emerging Role of the Interplay Among Astrocytes, Microglia, and Neurons in the Hippocampus in Health and Disease. Front Aging Neurosci 2021; 13:651973. [PMID: 33889084 PMCID: PMC8055856 DOI: 10.3389/fnagi.2021.651973] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
For over a century, neurons have been considered the basic functional units of the brain while glia only elements of support. Activation of glia has been long regarded detrimental for survival of neurons but more it appears that this is not the case in all circumstances. In this review, we report and discuss the recent literature on the alterations of astrocytes and microglia during inflammaging, the low-grade, slow, chronic inflammatory response that characterizes normal brain aging, and in acute inflammation. Becoming reactive, astrocytes and microglia undergo transcriptional, functional, and morphological changes that transform them into cells with different properties and functions, such as A1 and A2 astrocytes, and M1 and M2 microglia. This classification of microglia and astrocytes in two different, all-or-none states seems too simplistic, and does not correspond to the diverse variety of phenotypes so far found in the brain. Different interactions occur among the many cell populations of the central nervous system in health and disease conditions. Such interactions give rise to networks of morphological and functional reciprocal reliance and dependency. Alterations affecting one cell population reverberate to the others, favoring or dysregulating their activities. In the last part of this review, we present the modifications of the interplay between neurons and glia in rat models of brain aging and acute inflammation, focusing on the differences between CA1 and CA3 areas of the hippocampus, one of the brain regions most susceptible to different insults. With triple labeling fluorescent immunohistochemistry and confocal microscopy (TIC), it is possible to evaluate and compare quantitatively the morphological and functional alterations of the components of the neuron-astrocyte-microglia triad. In the contiguous and interconnected regions of rat hippocampus, CA1 and CA3 Stratum Radiatum, astrocytes and microglia show a different, finely regulated, and region-specific reactivity, demonstrating that glia responses vary in a significant manner from area to area. It will be of great interest to verify whether these differential reactivities of glia explain the diverse vulnerability of the hippocampal areas to aging or to different damaging insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli.
Collapse
Affiliation(s)
- Daniele Lana
- Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Section of Anatomopatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gary L Wenk
- Department of Psychology, The Ohio State University, Columbus, OH, United States
| | - Maria Grazia Giovannini
- Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
13
|
Upregulation of interleukin (IL)-31, a cytokine producing CXCR1 peripheral immune cells, contributes to the immune abnormalities of autism spectrum disorder. J Neuroimmunol 2020; 349:577430. [PMID: 33130460 DOI: 10.1016/j.jneuroim.2020.577430] [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: 04/23/2020] [Revised: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders characterized by communication deficits, impaired social interactions, and restricted stereotypical behaviors. Several immune cells are associated with immune dysfunction in ASD; however, IL-31 has not been explored in ASD. This study aims to investigate the role of inflammatory cytokines and transcription factors of the CXCR1 cells in children with ASD. In the current study, we investigated the cytokines and transcription factors produced by CXCR1+ cells (IL-31, IL-9, IL-21R, IL-21, NF-κB p65, RORγT, STAT1, and FoxP3) in peripheral blood mononuclear cells (PBMCs), from children with ASD and typically developing (TD) control children, using flow cytometric analysis. In addition, we measured mRNA and protein expression levels of IL-31 using quantitative real-time PCR and western blot analyses in PBMCs. In our study, children with ASD had increased CXCR1+IL-31+, CXCR1+IL-9+, CXCR1+IL-21R+, CXCR1+IL-21+, CXCR1+NF-κB+ p65, CXCR1+RORγT+, and CXCR1+STAT1+, and decreased CXCR1+FoxP3+ cells as compared with cells from the TD control samples. Similarly, children with ASD showed increased IL-31 mRNA and protein expression levels as compared to those of TD control samples. Our results suggest that upregulated production of inflammatory cytokines and transcription factors in CXCR1+ cells cause immunological imbalance in children with ASD. Therefore, attenuation of inflammatory cytokines/mediators and transcription factors could have a therapeutic potential in the treatment of ASD.
Collapse
|
14
|
Butler MJ, Cole RM, Deems NP, Belury MA, Barrientos RM. Fatty food, fatty acids, and microglial priming in the adult and aged hippocampus and amygdala. Brain Behav Immun 2020; 89:145-158. [PMID: 32544595 PMCID: PMC7572563 DOI: 10.1016/j.bbi.2020.06.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/20/2020] [Accepted: 06/06/2020] [Indexed: 02/08/2023] Open
Abstract
Short-term (3-day) consumption of a high fat diet (HFD) rich in saturated fats is associated with a neuroinflammatory response and subsequent cognitive impairment in aged, but not young adult, male rats. This exaggerated effect in aged rats could be due to a "primed" microglial phenotype observed in the normal aging process in rodents in which aged microglia display a potentiated response to immune challenge. Here, we investigated the impact of HFD on microglial priming and lipid composition in the hippocampus and amygdala of young and aged rats. Furthermore, we investigated the microglial response to palmitate, the main saturated fatty acid (SFA) found in HFD that is proinflammatory. Our results indicate that HFD increased gene expression of microglial markers of activation indicative of microglial priming, including CD11b, MHCII, CX3CR1, and NLRP3, as well as the pro-inflammatory marker IL-1β in both hippocampus and amygdala-derived microglia. Furthermore, HFD increased the concentration of SFAs and decreased the concentration of polyunsaturated fatty acids (PUFAs) in the hippocampus. We also observed a specific decrease in the anti-inflammatory PUFA docosahexaenoic acid (DHA) in the hippocampus and amygdala of aged rats. In a separate cohort of young and aged animals, isolated microglia from the hippocampus and amygdala exposed to palmitate in vitro induced an inflammatory gene expression profile mimicking the effects of HFD in vivo. These data suggest that palmitate may be a critical nutritional signal from the HFD that is directly involved in hippocampal and amygdalar inflammation. Interestingly, microglial activation markers were increased in response to HFD or palmitate in an age-independent manner, suggesting that HFD sensitivity of microglia, under these experimental conditions, is not the sole mediator of the exaggerated inflammatory response observed in whole tissue extracts from aged HFD-fed rats.
Collapse
Affiliation(s)
- Michael J. Butler
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Rachel M. Cole
- Department of Human Sciences, Ohio State University, Columbus, OH, USA
| | - Nicholas P. Deems
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Martha A. Belury
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA,Department of Human Sciences, Ohio State University, Columbus, OH, USA
| | - Ruth M. Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA,Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA,Corresponding author: Dr. Ruth M. Barrientos, Institute for Behavioral Medicine Research and Department of Psychiatry and Behavioral Health, Ohio State University, 460 Medical Center Drive, Columbus, OH 43210, Tel.: 614-293-6591,
| |
Collapse
|
15
|
Neuroinflammatory Responses and Parkinson' Disease: Pathogenic Mechanisms and Therapeutic Targets. J Neuroimmune Pharmacol 2020; 15:830-837. [PMID: 32529463 DOI: 10.1007/s11481-020-09926-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorders of the central nervous system, which mainly impairs the motor system. However, the pathogenic mechanisms are still unclear. Gene-environment complex interaction leads to selective dopaminergic neuron death in PD. Growing evidences supports that neuroinflammatory responses are involved in the pathogenesis of PD. This review critically discusses current studies on the inflammatory response of the pathological process of PD. The mechanisms and strategies of modifying inflammatory responses would be potential treatments for neurodegenerative diseases. Graphical abstract Activated microglia canpromote the damage ofdopaminergic neurons, which inturn aggravates the activation ofmicroglia in the process of PD. Atthe same time, microglia canactivate astrocytes throughproliferation and secretion ofinflammatory factors. The role ofastrocytes on the loss ofdopaminergic neurons is stillcontroversial in PD. (Nonsteroidalanti-inflammatory drugs,NSAIDs. adiposed-derived stemcells, ADSCs.nicotinamideadenine dinucleotide phosphate,NADPH. signal transducers andactivators of transcription,STAT.DJ-1,Aliases forPARK7.mesencephalic astrocytederivedneurotrophic factor,MANF.Ciliary neurotrophicfactor,CNTF.glial cell linederivedneurotrophic factor,GDNF.Wnt Family Member1,Wnt1). Graphical abstract Mitochondrial dysfunction causes neuroinflammation throughDAMPs and a series of factors such as oxidative stress andinflammatory bodies in PD. (Damage-associated molecular patterns,DAMPs. reactive oxygen species, ROS). Graphical abstract Various mechanismsparticipate in NLRP3 activation,causing microglia activation inPD. ( -synuclein, -syn.) TolllikeReceptor 2, TLR2. Toll-likeReceptor 4, TLR4. TumorNecrosis Factor, TNF.Apoptosisassociated speck like proteincontaining a CARD, ASC).
Collapse
|
16
|
Ellwanger JH, Kaminski VDL, Rodrigues AG, Kulmann-Leal B, Chies JAB. CCR5 and CCR5Δ32 in bacterial and parasitic infections: Thinking chemokine receptors outside the HIV box. Int J Immunogenet 2020; 47:261-285. [PMID: 32212259 DOI: 10.1111/iji.12485] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
The CCR5 molecule was reported in 1996 as the main HIV-1 co-receptor. In that same year, the CCR5Δ32 genetic variant was described as a strong protective factor against HIV-1 infection. These findings led to extensive research regarding the CCR5, culminating in critical scientific advances, such as the development of CCR5 inhibitors for the treatment of HIV infection. Recently, the research landscape surrounding CCR5 has begun to change. Different research groups have realized that, since CCR5 has such important effects in the chemokine system, it could also affect other different physiological systems. Therefore, the effect of reduced CCR5 expression due to the presence of the CCR5Δ32 variant began to be further studied. Several studies have investigated the role of CCR5 and the impacts of CCR5Δ32 on autoimmune and inflammatory diseases, various types of cancer, and viral diseases. However, the role of CCR5 in diseases caused by bacteria and parasites is still poorly understood. Therefore, the aim of this article is to review the role of CCR5 and the effects of CCR5Δ32 on bacterial (brucellosis, osteomyelitis, pneumonia, tuberculosis and infection by Chlamydia trachomatis) and parasitic infections (toxoplasmosis, leishmaniasis, Chagas disease and schistosomiasis). Basic information about each of these infections was also addressed. The neglected role of CCR5 in fungal disease and emerging studies regarding the action of CCR5 on regulatory T cells are briefly covered in this review. Considering the "renaissance of CCR5 research," this article is useful for updating researchers who develop studies involving CCR5 and CCR5Δ32 in different infectious diseases.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| |
Collapse
|
17
|
Sjöberg E, Meyrath M, Chevigné A, Östman A, Augsten M, Szpakowska M. The diverse and complex roles of atypical chemokine receptors in cancer: From molecular biology to clinical relevance and therapy. Adv Cancer Res 2020; 145:99-138. [PMID: 32089166 DOI: 10.1016/bs.acr.2019.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemokines regulate directed cell migration, proliferation and survival and are key components in cancer biology. They exert their functions by interacting with seven-transmembrane domain receptors that signal through G proteins (GPCRs). A subgroup of four chemokine receptors known as the atypical chemokine receptors (ACKRs) has emerged as essential regulators of the chemokine functions. ACKRs play diverse and complex roles in tumor biology from tumor initiation to metastasis, including cancer cell proliferation, adherence to endothelium, epithelial-mesenchymal transition (EMT), extravasation from blood vessels, tumor-associated angiogenesis or protection from immunological responses. This chapter gives an overview on the established and emerging roles that the atypical chemokine receptors ACKR1, ACKR2, ACKR3 and ACKR4 play in the different phases of cancer development and dissemination, their clinical relevance, as well as on the hurdles to overcome in ACKRs targeting as cancer therapy.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Max Meyrath
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
18
|
Treatment with CCR2 antagonist is neuroprotective but does not alter epileptogenesis in the pilocarpine rat model of epilepsy. Epilepsy Behav 2020; 102:106695. [PMID: 31785482 DOI: 10.1016/j.yebeh.2019.106695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 11/22/2022]
Abstract
Neuroinflammation role on epileptogenesis has been the subject of increasing interest. Many studies showed elevation in cytokines and chemokines expression following seizures, such as, CCL2 protein (C-C motif ligand 2 chemokine) and its specific receptor, CCR2. In addition, recent studies manipulating the CCL2/CCR2 complex verified improved seizure outcome in different seizure models. In the present study, the effects of CCR2 antagonist was investigated using the pilocarpine rat model of epilepsy. Status epilepticus (SE) was induced by pilocarpine i.p. injection in adult rats. Daily oral treatment with CCR2 antagonist or vehicle was initiated 5 h following SE and lasted 5 or 10 days. Rats were euthanized 5 days after SE to evaluate neuronal damage and glial density or 30 days after SE to investigate spontaneous seizures development and seizure susceptibility to a second hit pentylenetetrazol (PTZ) test. Rats that received CCR2 antagonist presented less degenerating cells at hippocampal CA1 region. There was also a significant decrease in CA1 volume after SE that was not observed in treated rats. On the other hand, microglia cell density increased after SE regardless of CCR2 antagonist use. Treatment with CCR2 antagonist did not alter spontaneous seizure occurrence or later seizure susceptibility to PTZ in chronic rats. Additional rats were pretreated with CCR2 antagonist prior to SE induction, but this did not change SE progression. The data show that oral treatment with CCR2 antagonist is neuroprotective, but does not alter other epileptogenic factors, such as, neuroinflammation, or seizure development, after pilocarpine-induced SE in rats.
Collapse
|
19
|
Murphy A, Barbaro J, Martínez-Aguado P, Chilunda V, Jaureguiberry-Bravo M, Berman JW. The Effects of Opioids on HIV Neuropathogenesis. Front Immunol 2019; 10:2445. [PMID: 31681322 PMCID: PMC6813247 DOI: 10.3389/fimmu.2019.02445] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
HIV associated neurocognitive disorders (HAND) are a group of neurological deficits that affect approximately half of people living with HIV (PLWH) despite effective antiretroviral therapy (ART). There are currently no reliable molecular biomarkers or treatments for HAND. Given the national opioid epidemic, as well as illegal and prescription use of opioid drugs among PLWH, it is critical to characterize the molecular interactions between HIV and opioids in cells of the CNS. It is also important to study the role of opioid substitution therapies in the context of HIV and CNS damage in vitro and in vivo. A major mechanism contributing to HIV neuropathogenesis is chronic, low-level inflammation in the CNS. HIV enters the brain within 4–8 days after peripheral infection and establishes CNS reservoirs, even in the context of ART, that are difficult to identify and eliminate. Infected cells, including monocytes, macrophages, and microglia, produce chemokines, cytokines, neurotoxic mediators, and viral proteins that contribute to chronic inflammation and ongoing neuronal damage. Opioids have been shown to impact these immune cells through a variety of molecular mechanisms, including opioid receptor binding and cross desensitization with chemokine receptors. The effects of opioid use on cognitive outcomes in individuals with HAND in clinical studies is variable, and thus multiple biological mechanisms are likely to contribute to the complex relationship between opioids and HIV in the CNS. In this review, we will examine what is known about both HIV and opioid mediated neuropathogenesis, and discuss key molecular processes that may be impacted by HIV and opioids in the context of neuroinflammation and CNS damage. We will also assess what is known about the effects of ART on these processes, and highlight areas of study that should be addressed in the context of ART.
Collapse
Affiliation(s)
- Aniella Murphy
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - John Barbaro
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pablo Martínez-Aguado
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Vanessa Chilunda
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Matias Jaureguiberry-Bravo
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Joan W Berman
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States.,Laboratory of Dr. Joan W. Berman, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
20
|
Choi JY, Kim JH, Hossain FMA, Uyangaa E, Park SO, Kim B, Kim K, Eo SK. Indispensable Role of CX 3CR1 + Dendritic Cells in Regulation of Virus-Induced Neuroinflammation Through Rapid Development of Antiviral Immunity in Peripheral Lymphoid Tissues. Front Immunol 2019; 10:1467. [PMID: 31316515 PMCID: PMC6610490 DOI: 10.3389/fimmu.2019.01467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
A coordinated host immune response mediated via chemokine network plays a crucial role in boosting defense mechanisms against pathogenic infections. The speed of Ag presentation and delivery by CD11c+ dendritic cells (DCs) to cognate T cells in lymphoid tissues may decide the pathological severity of the infection. Here, we investigated the role of CX3CR1 in the neuroinflammation induced by infection with Japanese encephalitis virus (JEV), a neurotrophic virus. Interestingly, CX3CR1 deficiency strongly enhanced susceptibility to JEV only after peripheral inoculation via footpad. By contrast, both CX3CR1+/+ and CX3CR1-/- mice showed comparable susceptibility to JEV following inoculation via intranasal and intraperitoneal routes. CX3CR1-/- mice exhibited lethal neuroinflammation after JEV inoculation via footpad route, showing high mortality, morbidity, pro-inflammatory cytokine expression, and uncontrolled CNS-infiltration of peripheral leukocytes including Ly-6Chi monocytes and Ly-6Ghi granulocytes. Furthermore, the absence of CX3CR1+CD11c+ DCs appeared to enhance susceptibility of CX3CR1-/- mice to JE after peripheral JEV inoculation. CX3CR1 ablation impaired the migration of CX3CR1+CD11c+ DCs from JEV-inoculated sites to draining lymph nodes (dLNs), resulting in decreased NK cell activation and JEV-specific CD4+/CD8+ T-cell responses. However, CX3CR1-competent mice showed rapid temporal expression of viral Ags in dLNs. Subsequently, JEV was rapidly cleared, with concomitant generation of antiviral NK cell activation and T-cell responses mediated by rapid migration of JEV Ag+CX3CR1+CD11c+ DCs. Using biallelic functional CX3CR1 expression system, the functional expression of CX3CR1 on CD11chi DCs appeared to be essentially required for inducing rapid and effective responses of NK cell activation and Ag-specific CD4+ T cells in dLNs. Strikingly, adoptive transfer of CX3CR1+CD11c+ DCs was found to completely restore the resistance of CX3CR1-/- recipients to JEV, as corroborated by the rapid delivery of JEV Ags in dLNs and attenuation of neuroinflammation in the CNS. Collectively, these results indicate that CX3CR1+CD11c+ DCs play an important role in generating rapid and effective responses of antiviral NK cell activation and Ag-specific T cells after peripheral inoculation with the virus, thereby resulting in conferring resistance to viral infection by reducing the peripheral viral burden.
Collapse
Affiliation(s)
- Jin Young Choi
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Jin Hyoung Kim
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Ferdaus Mohd Altaf Hossain
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea.,Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Erdenebelig Uyangaa
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Seong Ok Park
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Bumseok Kim
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan-si, South Korea
| | - Seong Kug Eo
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, South Korea
| |
Collapse
|
21
|
Du SH, Zhang W, Yue X, Luo XQ, Tan XH, Liu C, Qiao DF, Wang H. Role of CXCR1 and Interleukin-8 in Methamphetamine-Induced Neuronal Apoptosis. Front Cell Neurosci 2018; 12:230. [PMID: 30123110 PMCID: PMC6085841 DOI: 10.3389/fncel.2018.00230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine (METH), an extremely and widely abused illicit drug, can cause serious nervous system damage and social problems. Previous research has shown that METH use causes dopaminergic neuron apoptosis and astrocyte-related neuroinflammation. However, the relationship of astrocytes and neurons in METH-induced neurotoxicity remains unclear. We hypothesized that chemokine interleukin (IL) eight released by astrocytes and C-X-C motif chemokine receptor 1 (CXCR1) in neurons are involved in METH-induced neuronal apoptosis. We tested our hypothesis by examining the changes of CXCR1 in SH-SY5Y cells and in the brain of C57BL/6 mice exposed to METH by western blotting and immunolabeling. We also determined the effects of knocking down CXCR1 expression with small interfering ribonucleic acid (siRNA) on METH-exposed SH-SY5Y cells. Furthermore, we detected the expression levels of IL-8 and the nuclear factor-kappa B (NF-κB) pathway in U87MG cells and then co-cultured the two cell types to determine the role of CXCR1 and IL-8 in neuronal apoptosis. Our results indicated that METH exposure increased CXCR1 expression both in vitro and in vivo, with the effects obtained in vitro being dose-dependent. Silencing of CXCR1 expression with siRNAs reduced the expression of cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), and other related proteins. In addition, IL-8 expression and release were increased in METH-exposed U87MG cells, which is regulated by NF-κB pathway. Neuronal apoptosis was attenuated by siCXCR1 after METH treatment in the co-cultured cells, which can be reversed after exposure to recombinant IL-8. These results demonstrate that CXCR1 plays an important role in neuronal apoptosis induced by METH and may be a potential target for METH-induced neurotoxicity therapy. Highlights -Methamphetamine exposure upregulated the expression of CXCR1.-Methamphetamine exposure increased the expression of interleukin-8 through nuclear factor-kappa B pathway.-Activation of CXCR1 by interleukin-8 induces an increase in methamphetamine-related neuronal apoptosis.
Collapse
Affiliation(s)
- Si-Hao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei Zhang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xia Yue
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiao-Qing Luo
- School of Forensic Medicine, Southern Medical University, Guangzhou, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Hui Tan
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou Public Security Bureau, Guangzhou, China
| | - Dong-Fang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Bioinformatics Genes and Pathway Analysis for Chronic Neuropathic Pain after Spinal Cord Injury. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6423021. [PMID: 29164149 PMCID: PMC5661087 DOI: 10.1155/2017/6423021] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/09/2017] [Accepted: 09/07/2017] [Indexed: 12/11/2022]
Abstract
It is well known spinal cord injury (SCI) can cause chronic neuropathic pain (NP); however its underlying molecular mechanisms remain elusive. This study aimed to disclose differentially expressed genes (DEGs) and activated signaling pathways in association with SCI induced chronic NP, in order to identify its diagnostic and therapeutic targets. Microarray dataset GSE5296 has been downloaded from Gene Expression Omnibus (GEO) database. Significant analysis of microarray (SAM), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and pathway network analysis have been used to compare changes of DEGs and signaling pathways between the SCI and sham-injury group. As a result, DEGs analysis showed there were 592 DEGs with significantly altered expression; among them Ccl3 expression showed the highest upregulation which implicated its association with SCI induced chronic NP. Moreover, KEGG analysis found 209 pathways changed significantly; among them the most significantly activated one is MAPK signaling pathway, which is in line with KEGG analysis results. Our results show Ccl3 is highly associated with SCI induced chronic NP; as the exosomes with Ccl3 can be easily and efficiently detected in peripheral blood, Ccl3 may serve as a potential prognostic target for the diagnosis and treatment of SCI induced chronic NP.
Collapse
|
23
|
Pittaluga A. CCL5-Glutamate Cross-Talk in Astrocyte-Neuron Communication in Multiple Sclerosis. Front Immunol 2017; 8:1079. [PMID: 28928746 PMCID: PMC5591427 DOI: 10.3389/fimmu.2017.01079] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022] Open
Abstract
The immune system (IS) and the central nervous system (CNS) are functionally coupled, and a large number of endogenous molecules (i.e., the chemokines for the IS and the classic neurotransmitters for the CNS) are shared in common between the two systems. These interactions are key elements for the elucidation of the pathogenesis of central inflammatory diseases. In recent years, evidence has been provided supporting the role of chemokines as modulators of central neurotransmission. It is the case of the chemokines CCL2 and CXCL12 that control pre- and/or post-synaptically the chemical transmission. This article aims to review the functional cross-talk linking another endogenous pro-inflammatory factor released by glial cells, i.e., the chemokine Regulated upon Activation Normal T-cell Expressed and Secreted (CCL5) and the principal neurotransmitter in CNS (i.e., glutamate) in physiological and pathological conditions. In particular, the review discusses preclinical data concerning the role of CCL5 as a modulator of central glutamatergic transmission in healthy and demyelinating disorders. The CCL5-mediated control of glutamate release at chemical synapses could be relevant either to the onset of psychiatric symptoms that often accompany the development of multiple sclerosis (MS), but also it might indirectly give a rationale for the progression of inflammation and demyelination. The impact of disease-modifying therapies for the cure of MS on the endogenous availability of CCL5 in CNS will be also summarized. We apologize in advance for omission in our coverage of the existing literature.
Collapse
Affiliation(s)
- Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
24
|
Khaibullin T, Ivanova V, Martynova E, Cherepnev G, Khabirov F, Granatov E, Rizvanov A, Khaiboullina S. Elevated Levels of Proinflammatory Cytokines in Cerebrospinal Fluid of Multiple Sclerosis Patients. Front Immunol 2017; 8:531. [PMID: 28572801 PMCID: PMC5435759 DOI: 10.3389/fimmu.2017.00531] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/20/2017] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disease characterized by chronic brain inflammation. Leukocyte infiltration of brain tissue causes inflammation, demyelination, and the subsequent formation of sclerotic plaques, which are a hallmark of MS. Activation of proinflammatory cytokines is essential for regulation of lymphocyte migration across the blood–brain barrier. We demonstrate increased levels of many cytokines, including IL-2RA, CCL5, CCL11, MIF, CXCL1, CXCL10, IFNγ, SCF, and TRAIL, were upregulated in cerebrospinal fluid (CSF), whereas IL-17, CCL2, CCL3, CCL4, and IL-12(p40) were activated in MS serum. Interaction analysis of cytokines in CSF demonstrated a connection between IFNγ and CCL5 as well as MIF. Many cells can contribute to production of these cytokines including CD8 and Th1 lymphocytes and astrocytes. Therefore, we suggest that IFNγ released by Th1 lymphocytes can activate astrocytes, which then produce chemoattractants, including CCL5 and MIF. These chemokines promote an inflammatory milieu and interact with multiple chemokines including CCL27 and CXCL1. Of special note, upregulation of CCL27 was found in CSF of MS cases. This observation is the first to demonstrate CCL27 as a potential contributor of brain pathology in MS. Our data suggest that CCL27 may be involved in activation and migration of autoreactive encephalitogenic immune effectors in the brain. Further, our data support the role of Th1 lymphocytes in the pathogenesis of brain inflammation in MS, with several cytokines playing a central role.
Collapse
Affiliation(s)
| | - Vilena Ivanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Georgy Cherepnev
- University Kazan Clinic, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | | | | | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Republic of Tatarstan, Russian Federation.,Nevada Center for Biomedical Research, Reno, NV, USA
| |
Collapse
|
25
|
Blatt NL, Khaiboullin TI, Lombardi VC, Rizvanov AA, Khaiboullina SF. The Skin-Brain Connection Hypothesis, Bringing Together CCL27-Mediated T-Cell Activation in the Skin and Neural Cell Damage in the Adult Brain. Front Immunol 2017; 7:683. [PMID: 28138328 PMCID: PMC5237636 DOI: 10.3389/fimmu.2016.00683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/22/2016] [Indexed: 11/21/2022] Open
Abstract
Recent discovery of an association of low serum melatonin levels with relapse in multiple sclerosis (MS) opens a new horizon in understanding the pathogenesis of this disease. Skin is the main organ for sensing seasonal changes in duration of sunlight exposure. Level of melatonin production is dependent on light exposure. The molecular mechanisms connecting peripheral (skin) sensing of the light exposure and developing brain inflammation (MS) have not been investigated. We hypothesize that there is a connection between the reaction of skin to seasonal changes in sunlight exposure and the risk of MS and that seasonal changes in light exposure cause peripheral (skin) inflammation, the production of cytokines, and the subsequent inflammation of the brain. In skin of genetically predisposed individuals, cytokines attract memory cutaneous lymphocyte-associated antigen (CLA+) T lymphocytes, which then maintain local inflammation. Once inflammation is resolved, CLA+ lymphocytes return to the circulation, some of which eventually migrate to the brain. Once in the brain these lymphocytes may initiate an inflammatory response. Our observation of increased CC chemokine ligand 27 (CCL27) in MS sera supports the involvement of skin in the pathogenesis of MS. Further, the importance of our data is that CCL27 is a chemokine released by activated keratinocytes, which is upregulated in inflamed skin. We propose that high serum levels of CCL27 in MS are the result of skin inflammation due to exposure to seasonal changes in the sunlight. Future studies will determine whether CCL27 serum level correlates with seasonal changes in sunlight exposure, MS exacerbation, and skin inflammation.
Collapse
Affiliation(s)
- Nataliya L Blatt
- Institute of Fundamental Medicine and Biology, Kazan Federal University , Kazan , Russia
| | | | - Vincent C Lombardi
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Nevada Center for Biomedical Research, Reno, NV, USA
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University , Kazan , Russia
| | - Svetlana F Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Nevada Center for Biomedical Research, Reno, NV, USA
| |
Collapse
|
26
|
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
|
27
|
Heo JC, Jung TH, Lee S, Kim HY, Choi G, Jung M, Jung D, Lee HK, Lee JO, Park JH, Hwang D, Seol HJ, Cho H. Effect of bexarotene on differentiation of glioblastoma multiforme compared with ATRA. Clin Exp Metastasis 2016; 33:417-29. [DOI: 10.1007/s10585-016-9786-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/25/2016] [Indexed: 12/27/2022]
|
28
|
Couturier J, Stancu IC, Schakman O, Pierrot N, Huaux F, Kienlen-Campard P, Dewachter I, Octave JN. Activation of phagocytic activity in astrocytes by reduced expression of the inflammasome component ASC and its implication in a mouse model of Alzheimer disease. J Neuroinflammation 2016; 13:20. [PMID: 26818951 PMCID: PMC4729126 DOI: 10.1186/s12974-016-0477-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/06/2016] [Indexed: 12/19/2022] Open
Abstract
Background The proinflammatory cytokine interleukin-1β (IL-1β) is overexpressed in Alzheimer disease (AD) as a key regulator of neuroinflammation. Amyloid-β (Aβ) peptide triggers activation of inflammasomes, protein complexes responsible for IL-1β maturation in microglial cells. Downregulation of NALP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome has been shown to decrease amyloid load and rescue cognitive deficits in a mouse model of AD. Whereas activation of inflammasome in microglial cells has been described in AD, no data are currently available concerning activation of inflammasome in astrocytes, although they are involved in inflammatory response and phagocytosis. Here, by targeting the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD domain), we investigated the influence of activation of the inflammasome on the phagocytic activity of astrocytes. Methods We used an ASC knockout mouse model, as ASC is a central protein in the inflammasome, acting as an adaptor and stabilizer of the complex and thus critical for its activation. Lipopolysaccharide (LPS)-primed primary cultures of astrocytes from newborn mice were utilized to evaluate Aβ-induced inflammasome activation by measuring IL-1β release by ECLIA (electro-chemiluminescence immunoassay). Phagocytosis efficiency was measured by incorporation of bioparticles, and the release of the chemokine CCL3 (C-C motif ligand 3) was measured by ECLIA. ASC mice were crossbred with 5xFAD (familial Alzheimer disease) mice and tested for spatial reference memory using the Morris water maze (MWM) at 7–8 months of age. Amyloid load and CCL3 were quantified by thioflavine S staining and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. Results Cultured astrocytes primed with LPS and treated with Aβ showed an ASC-dependent production of IL-1β resulting from inflammasome activation mediated by Aβ phagocytosis and cathepsin B enzymatic activity. ASC+/− astrocytes displayed a higher phagocytic activity as compared to ASC+/+ and ASC −/− cells, resulting from a higher release of the chemokine CCL3. A significant decrease in amyloid load was measured in the brain of 7–8-month-old 5xFAD mice carrying the ASC +/− genotype, correlated with an increase in CCL3 gene expression. In addition, the ASC +/− genotype rescued spatial reference memory deficits observed in 5xFAD mice. Conclusions Our results demonstrate that Aβ is able to activate astrocytic inflammasome. Downregulation of inflammasome activity increases phagocytosis in astrocytes due to the release of CCL3. This could explain why downregulation of inflammasome activity decreases amyloid load and rescues memory deficits in a mouse model of AD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0477-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julien Couturier
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Ilie-Cosmin Stancu
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Olivier Schakman
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Nathalie Pierrot
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - François Huaux
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Université catholique de Louvain, Brussels, Belgium.
| | - Pascal Kienlen-Campard
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Ilse Dewachter
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| | - Jean-Noël Octave
- Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium. .,Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate 54, B1.5410, B-1200, Brussels, Belgium.
| |
Collapse
|
29
|
Identification of neurotoxic cytokines by profiling Alzheimer's disease tissues and neuron culture viability screening. Sci Rep 2015; 5:16622. [PMID: 26564777 PMCID: PMC4643219 DOI: 10.1038/srep16622] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) therapeutics based on the amyloid hypothesis have shown minimal efficacy in patients, suggesting that the activity of amyloid beta (Aβ) represents only one aspect of AD pathogenesis. Since neuroinflammation is thought to play an important role in AD, we hypothesized that cytokines may play a direct role in promoting neuronal death. Here, we profiled cytokine expression in a small cohort of human AD and control brain tissues. We identified AD-associated cytokines using partial least squares regression to correlate cytokine expression with quantified pathologic disease state and then used neuron cultures to test whether cytokines up-regulated in AD tissues could affect neuronal viability. This analysis identified cytokines that were associated with the pathological severity. Of the top correlates, only TNF-α reduced viability in neuron culture when applied alone. VEGF also reduced viability when applied together with Aβ, which was surprising because VEGF has been viewed as a neuro-protective protein. We found that this synthetic pro-death effect of VEGF in the context of Aβ was commensurate with VEGFR-dependent changes in multiple signaling pathways that govern cell fate. Our findings suggest that profiling of tissues combined with a culture-based screening approach can successfully identify new mechanisms driving neuronal death.
Collapse
|
30
|
Fitting S, Zou S, El-Hage N, Suzuki M, Paris JJ, Schier CJ, Rodríguez JW, Rodriguez M, Knapp PE, Hauser KF. Opiate addiction therapies and HIV-1 Tat: interactive effects on glial [Ca²⁺]i, oxyradical and neuroinflammatory chemokine production and correlative neurotoxicity. Curr HIV Res 2015; 12:424-34. [PMID: 25760046 PMCID: PMC4475822 DOI: 10.2174/1570162x1206150311161147] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/13/2014] [Accepted: 02/12/2015] [Indexed: 01/05/2023]
Abstract
Few preclinical studies have compared the relative therapeutic efficacy of medications used to treat opiate addiction in relation to neuroAIDS. Here we compare the ability of methadone and buprenorphine, and the prototypic opiate morphine, to potentiate the neurotoxic and proinflammatory ([Ca2+]i, ROS, H2O2, chemokines) effects of HIV-1 Tat in neuronal and/or mixed-glial co-cultures. Repeated observations of neurons during 48 h exposure to combinations of Tat, equimolar concentrations (500 nM) of morphine, methadone, or buprenorphine exacerbated neurotoxicity significantly above levels seen with Tat alone. Buprenorphine alone displayed marked neurotoxicity at 500 nM, prompting additional studies of its neurotoxic effects at 5 nM and 50 nM concentrations ± Tat. In combination with Tat, buprenorphine displayed paradoxical, concentration-dependent, neurotoxic and neuroprotective actions. Buprenorphine neurotoxicity coincided with marked elevations in [Ca2+]i, but not increases in glial ROS or chemokine release. Tat by itself elevated the production of CCL5/RANTES, CCL4/MIP-1β, and CCL2/MCP-1. Methadone and buprenorphine alone had no effect, but methadone interacted with Tat to further increase production of CCL5/RANTES. In combination with Tat, all drugs significantly increased glial [Ca2+]i, but ROS was only significantly increased by co-exposure with morphine. Taken together, the increases in glial [Ca2+]i, ROS, and neuroinflammatory chemokines were not especially accurate predictors of neurotoxicity. Despite similarities, opiates displayed differences in their neurotoxic and neuroinflammatory interactions with Tat. Buprenorphine, in particular, was partially neuroprotective at a low concentration, which may result from its unique pharmacological profile at multiple opioid receptors. Overall, the results reveal differences among addiction medications that may impact neuroAIDS.
Collapse
|
31
|
Khaiboullina SF, Gumerova AR, Khafizova IF, Martynova EV, Lombardi VC, Bellusci S, Rizvanov AA. CCL27: Novel Cytokine with Potential Role in Pathogenesis of Multiple Sclerosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:189638. [PMID: 26295034 PMCID: PMC4532821 DOI: 10.1155/2015/189638] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/11/2014] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune and neurodegenerative disease of unknown etiology. Leukocyte infiltration of brain tissue and the subsequent inflammation, demyelination, axonal damage, and formation of sclerotic plaques is a hallmark of MS. Upregulation of proinflammatory cytokines has been suggested to play an essential role in regulating lymphocyte migration in MS. Here we present data on serum cytokine expression in MS cases. Increased serum levels of IL-17 and IL-23 were observed, suggesting activation of the Th17 population of immune effector cells. Additionally, increased levels of IL-22 were observed in the serum of those with acute phase MS. Unexpectedly, we observed an upregulation of the serum chemokine CCL27 in newly diagnosed and acute MS cases. CCL27 is an inflammatory chemokine associated with homing of memory T cells to sites of inflammation. Therefore, its upregulation in association with MS suggests a potential role in disease pathogenesis. Our data supports previous reports showing IL-17 and -23 upregulation in association with MS and potentially identify a previously unknown involvement for CCL27.
Collapse
Affiliation(s)
- Svetlana F. Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- WP Institute, Reno, NV 89557, USA
| | - Aigul R. Gumerova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Kazan State Medical University, 49 Butlerova Street, Kazan, Tatarstan 420012, Russia
| | - Irina F. Khafizova
- Kazan State Medical University, 49 Butlerova Street, Kazan, Tatarstan 420012, Russia
| | - Ekaterina V. Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Kazan State Medical University, 49 Butlerova Street, Kazan, Tatarstan 420012, Russia
| | - Vincent C. Lombardi
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- WP Institute, Reno, NV 89557, USA
| | - Saverio Bellusci
- Excellence Cluster Cardio-Pulmonary System, Justus Liebig University, Aulweg 130, 35392 Giessen, Germany
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia
| |
Collapse
|
32
|
Abstract
Neurodegenerative diseases (NDs) collectively afflict more than 40 million people worldwide. The majority of these diseases lack therapies to slow or stop progression due in large part to the challenge of disentangling the simultaneous presentation of broad, multifaceted pathophysiologic changes. Present technologies and computational capabilities suggest an optimistic future for deconvolving these changes to identify novel mechanisms driving ND onset and progression. In particular, integration of highly multi-dimensional omic analytical techniques (e.g., microarray, mass spectrometry) with computational systems biology approaches provides a systematic methodology to elucidate new mechanisms driving NDs. In this review, we begin by summarizing the complex pathophysiology of NDs associated with protein aggregation, emphasizing the shared complex dysregulation found in all of these diseases, and discuss available experimental ND models. Next, we provide an overview of technological and computational techniques used in systems biology that are applicable to studying NDs. We conclude by reviewing prior studies that have applied these approaches to NDs and comment on the necessity of combining analysis from both human tissues and model systems to identify driving mechanisms. We envision that the integration of computational approaches with multiple omic analyses of human tissues, and mouse and in vitro models, will enable the discovery of new therapeutic strategies for these devastating diseases.
Collapse
Affiliation(s)
- Levi B Wood
- Cancer Research Institute, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
33
|
The chemokine (C-C motif) ligand 2 in neuroinflammation and neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 824:209-19. [PMID: 25039002 DOI: 10.1007/978-3-319-07320-0_15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Among all the chemokines known so far, chemokine (C-C motif) ligand 2 (CCL2) is probably the best characterized. This is mainly due to the therapeutic potential attributed to its regulation. The suppression of CCL2 function may reduce the attraction of immune cells to the sites of inflammation and therefore slow down the progression of inflammation and the tissue damage that may be associated to it. While this has proven to be right in diverse conditions, it has also been described to have deleterious consequences such as a dual effect that is also frequently observed in other endogenous defense systems. This review discusses current knowledge about CCL2 involvement in different neurodegenerative diseases as well as its anti-inflammatory and neuro-protective actions.
Collapse
|
34
|
Arisi GM. Nervous and immune systems signals and connections: cytokines in hippocampus physiology and pathology. Epilepsy Behav 2014; 38:43-7. [PMID: 24534466 DOI: 10.1016/j.yebeh.2014.01.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 01/20/2014] [Accepted: 01/22/2014] [Indexed: 02/07/2023]
Abstract
Signaling through secretion of small molecules is a hallmark of both nervous and immune systems. The scope and influence of the intense message exchange between these two complex systems are only now becoming objects of scientific inquiry. Both neurotransmitters and cytokines affect their target cells through surface receptors and also by other molecular mechanisms. Cytokine receptors are present in neurons and glial cell populations in discrete brain regions. This review firstly focuses on the role of cytokines in hippocampal physiological processes, such as memory and learning, and secondly on the pathological involvement of cytokines in diseases like depression and epilepsy. Interleukin-1β is necessary for long-term potentiation (LTP) maintenance in the hippocampus. On the other hand, interleukin-6 has a negative regulatory role in long-term memory acquisition. Astrocyte-secreted tumor necrosis factor plays a role in synaptic strength by increasing surface translocation of glutamate AMPA receptors, and the chemokine CXCL12 can silence the tonic activity of Cajal-Retzius neurons in the hippocampus. Manifold increased concentrations of interleukin-10, interferon-γ, ICAM1, CCL2, and CCL4 are observed in the hippocampi of patients with temporal lobe epilepsy. A contemporary view of the role of cytokines as neuromodulators is emerging from studies in humans and manipulations of experimental animals. Despite the accumulating evidence of the role of cytokines on nervous system physiology and pathology, it is important not to exaggerate its relevance.
Collapse
Affiliation(s)
- Gabriel Maisonnave Arisi
- Neurobiology Laboratory, Department of Physiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
35
|
Lee JC, Ahn JH, Kim IH, Park JH, Yan BC, Cho GS, Ohk TG, Park CW, Cho JH, Kim YM, Lee HY, Won MH. Transient ischemia-induced change of CCR7 immunoreactivity in neurons and its new expression in astrocytes in the gerbil hippocampus. J Neurol Sci 2014; 336:203-10. [DOI: 10.1016/j.jns.2013.10.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/10/2013] [Accepted: 10/28/2013] [Indexed: 12/25/2022]
|
36
|
Aiamkitsumrit B, Dampier W, Antell G, Rivera N, Martin-Garcia J, Pirrone V, Nonnemacher MR, Wigdahl B. Bioinformatic analysis of HIV-1 entry and pathogenesis. Curr HIV Res 2014; 12:132-61. [PMID: 24862329 PMCID: PMC4382797 DOI: 10.2174/1570162x12666140526121746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/18/2014] [Accepted: 05/06/2014] [Indexed: 02/07/2023]
Abstract
The evolution of human immunodeficiency virus type 1 (HIV-1) with respect to co-receptor utilization has been shown to be relevant to HIV-1 pathogenesis and disease. The CCR5-utilizing (R5) virus has been shown to be important in the very early stages of transmission and highly prevalent during asymptomatic infection and chronic disease. In addition, the R5 virus has been proposed to be involved in neuroinvasion and central nervous system (CNS) disease. In contrast, the CXCR4-utilizing (X4) virus is more prevalent during the course of disease progression and concurrent with the loss of CD4(+) T cells. The dual-tropic virus is able to utilize both co-receptors (CXCR4 and CCR5) and has been thought to represent an intermediate transitional virus that possesses properties of both X4 and R5 viruses that can be encountered at many stages of disease. The use of computational tools and bioinformatic approaches in the prediction of HIV-1 co-receptor usage has been growing in importance with respect to understanding HIV-1 pathogenesis and disease, developing diagnostic tools, and improving the efficacy of therapeutic strategies focused on blocking viral entry. Current strategies have enhanced the sensitivity, specificity, and reproducibility relative to the prediction of co-receptor use; however, these technologies need to be improved with respect to their efficient and accurate use across the HIV-1 subtypes. The most effective approach may center on the combined use of different algorithms involving sequences within and outside of the env-V3 loop. This review focuses on the HIV-1 entry process and on co-receptor utilization, including bioinformatic tools utilized in the prediction of co-receptor usage. It also provides novel preliminary analyses for enabling identification of linkages between amino acids in V3 with other components of the HIV-1 genome and demonstrates that these linkages are different between X4 and R5 viruses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102.
| |
Collapse
|
37
|
Brissette CA, Kees ED, Burke MM, Gaultney RA, Floden AM, Watt JA. The multifaceted responses of primary human astrocytes and brain microvascular endothelial cells to the Lyme disease spirochete, Borrelia burgdorferi. ASN Neuro 2013; 5:221-9. [PMID: 23883071 PMCID: PMC3745032 DOI: 10.1042/an20130010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/24/2013] [Accepted: 07/24/2013] [Indexed: 12/17/2022] Open
Abstract
The vector-borne pathogen, Borrelia burgdorferi, causes a multi-system disorder including neurological complications. These neurological disorders, collectively termed neuroborreliosis, can occur in up to 15% of untreated patients. The neurological symptoms are probably a result of a glial-driven, host inflammatory response to the bacterium. However, the specific contributions of individual glial and other support cell types to the pathogenesis of neuroborreliosis are relatively unexplored. The goal of this project was to characterize specific astrocyte and endothelial cell responses to B. burgdorferi. Primary human astrocytes and primary HBMEC (human brain microvascular endothelial cells) were incubated with B. burgdorferi over a 72-h period and the transcriptional responses to the bacterium were analyzed by real-time PCR arrays. There was a robust increase in several surveyed chemokine and related genes, including IL (interleukin)-8, for both primary astrocytes and HBMEC. Array results were confirmed with individual sets of PCR primers. The production of specific chemokines by both astrocytes and HBMEC in response to B. burgdorferi, including IL-8, CXCL-1, and CXCL-10, were confirmed by ELISA. These results demonstrate that primary astrocytes and HBMEC respond to virulent B. burgdorferi by producing a number of chemokines. These data suggest that infiltrating phagocytic cells, particularly neutrophils, attracted by chemokines expressed at the BBB (blood-brain barrier) may be important contributors to the early inflammatory events associated with neuroborreliosis.
Collapse
Key Words
- astrocyte
- blood–brain barrier
- borrelia burgdorferi
- chemokine
- endothelium
- neuroborreliosis.
- bbb, blood–brain barrier
- ccl, cc chemokine ligand
- cns, central nervous system
- csf, cerebrospinal fluid
- cxcr, cxc chemokine receptor
- ecm, endothelial cell medium
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- hbmec, human brain microvascular endothelial cells
- il, interleukin
- moi, multiplicity of infection
- qpcr, quantitative pcr
- tnfα, tumor necrosis factor α
Collapse
Affiliation(s)
- Catherine A Brissette
- Microbiology and Immunology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Grillo E, Lo Rizzo C, Bianciardi L, Bizzarri V, Baldassarri M, Spiga O, Furini S, De Felice C, Signorini C, Leoncini S, Pecorelli A, Ciccoli L, Mencarelli MA, Hayek J, Meloni I, Ariani F, Mari F, Renieri A. Revealing the complexity of a monogenic disease: rett syndrome exome sequencing. PLoS One 2013; 8:e56599. [PMID: 23468869 PMCID: PMC3585308 DOI: 10.1371/journal.pone.0056599] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/11/2013] [Indexed: 02/04/2023] Open
Abstract
Rett syndrome (OMIM#312750) is a monogenic disorder that may manifest as a large variety of phenotypes ranging from very severe to mild disease. Since there is a weak correlation between the mutation type in the Xq28 disease-gene MECP2/X-inactivation status and phenotypic variability, we used this disease as a model to unveil the complex nature of a monogenic disorder. Whole exome sequencing was used to analyze the functional portion of the genome of two pairs of sisters with Rett syndrome. Although each pair of sisters had the same MECP2 (OMIM*300005) mutation and balanced X-inactivation, one individual from each pair could not speak or walk, and had a profound intellectual deficit (classical Rett syndrome), while the other individual could speak and walk, and had a moderate intellectual disability (Zappella variant). In addition to the MECP2 mutation, each patient has a group of variants predicted to impair protein function. The classical Rett girls, but not their milder affected sisters, have an enrichment of variants in genes related to oxidative stress, muscle impairment and intellectual disability and/or autism. On the other hand, a subgroup of variants related to modulation of immune system, exclusive to the Zappella Rett patients are driving toward a milder phenotype. We demonstrate that genome analysis has the potential to identify genetic modifiers of Rett syndrome, providing insight into disease pathophysiology. Combinations of mutations that affect speaking, walking and intellectual capabilities may represent targets for new therapeutic approaches. Most importantly, we demonstrated that monogenic diseases may be more complex than previously thought.
Collapse
Affiliation(s)
- Elisa Grillo
- Medical Genetics, University of Siena, Siena, Italy
| | - Caterina Lo Rizzo
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Veronica Bizzarri
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Ottavia Spiga
- Biochemistry and Molecular Biology, University of Siena, Siena, Italy
| | - Simone Furini
- Department of Surgery and Bioengineering University of Siena, Siena, Italy
| | - Claudio De Felice
- Neonatal Intensive Care Unit University Hospital Azienda Ospedaliera Universitaria Senese (AOUS) of Siena, Siena, Italy
| | - Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Silvia Leoncini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Child Neuropsychiatry Unit, University Hospital, AOUS, Siena, Italy
| | - Alessandra Pecorelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Child Neuropsychiatry Unit, University Hospital, AOUS, Siena, Italy
| | - Lucia Ciccoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Maria Antonietta Mencarelli
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Joussef Hayek
- Child Neuropsychiatry Unit, University Hospital, AOUS, Siena, Italy
| | | | | | - Francesca Mari
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- * E-mail:
| |
Collapse
|
39
|
Weinger JG, Marro BS, Hosking MP, Lane TE. The chemokine receptor CXCR2 and coronavirus-induced neurologic disease. Virology 2013; 435:110-7. [PMID: 23217621 PMCID: PMC3522860 DOI: 10.1016/j.virol.2012.08.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 08/31/2012] [Indexed: 12/18/2022]
Abstract
Inoculation with the neurotropic JHM strain of mouse hepatitis virus (MHV) into the central nervous system (CNS) of susceptible strains of mice results in an acute encephalomyelitis in which virus preferentially replicates within glial cells while excluding neurons. Control of viral replication during acute disease is mediated by infiltrating virus-specific T cells via cytokine secretion and cytolytic activity, however sterile immunity is not achieved and virus persists resulting in chronic neuroinflammation associated with demyelination. CXCR2 is a chemokine receptor that upon binding to specific ligands promotes host defense through recruitment of myeloid cells to the CNS as well as protecting oligodendroglia from cytokine-mediated death in response to MHV infection. These findings highlight growing evidence of the diverse and important role of CXCR2 in regulating neuroinflammatory diseases.
Collapse
Affiliation(s)
- Jason G Weinger
- Department of Molecular Biology & Biochemistry, UC Irvine, CA 92697-3900, USA
| | | | | | | |
Collapse
|
40
|
Lipfert J, Ödemis V, Engele J. Grk2 is an essential regulator of CXCR7 signalling in astrocytes. Cell Mol Neurobiol 2013; 33:111-8. [PMID: 22940879 PMCID: PMC11497910 DOI: 10.1007/s10571-012-9876-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/17/2012] [Indexed: 10/27/2022]
Abstract
We previously demonstrated that in astrocytes, SDF-1/CXCL12 exclusively signals through CXCR7 despite the additional presence of the alternate SDF-1/CXCL12 receptor, CXCR4. In addition, we provided evidence that astrocytic CXCR7-signalling involves a G protein-dependent mechanism. This is insofar remarkable as in all other cell types studied to date, CXCR7 either acts as a scavenger chemokine receptor, a modulator of CXCR4, or a non-classical chemokine receptor, signalling through ß-arrestin. To begin to unravel the molecular framework impinging the selective function of CXCR7 on a given cell type, we have now analysed the role of G protein-coupled receptor kinases (Grks) in astrocytic CXCR7 signalling. We demonstrate that Grk2 mediates signalling of SDF-1/CXCL12-bound CXCR7 as suggested by the finding that SDF-1/CXCL12-induced activation of Erk1/2 and Akt is abrogated following RNAi-mediated inhibition of Grk2, but not of Grk3, Grk5, or Grk6. We further unravel that Grk2 additionally controls signalling of SDF-1/CXCL12-bound CXCR7 in astrocytes by mediating internalization and subsequent silencing of CXCR7. Finally, we demonstrate that Grk2 is likewise expressed by microglial cells and Schwann cells, cell types in which CXCR7 does not act as a classical chemokine receptor. In conclusion, our findings establish that Grk2 tightly controls CXCR7 signalling in astrocytes, but does not imprint the cell type-specific function of this chemokine receptor.
Collapse
Affiliation(s)
- Jana Lipfert
- Medical Faculty, Institute of Anatomy, University of Leipzig, Liebigstr 13, 04103 Leipzig, Germany
| | - Veysel Ödemis
- Medical Faculty, Institute of Anatomy, University of Leipzig, Liebigstr 13, 04103 Leipzig, Germany
| | - Jürgen Engele
- Medical Faculty, Institute of Anatomy, University of Leipzig, Liebigstr 13, 04103 Leipzig, Germany
| |
Collapse
|
41
|
Li X, Lin WJ, Chen CY, Si Y, Zhang X, Lu L, Suswam E, Zheng L, King PH. KSRP: a checkpoint for inflammatory cytokine production in astrocytes. Glia 2012; 60:1773-84. [PMID: 22847996 DOI: 10.1002/glia.22396] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/29/2012] [Indexed: 11/08/2022]
Abstract
Chronic inflammation in the central nervous system (CNS) is a central feature of many neurodegenerative and autoimmune diseases. As an immunologically competent cell, the astrocyte plays an important role in CNS inflammation. It is capable of expressing a number of cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) that promote inflammation directly and through the recruitment of immune cells. Checkpoints are therefore in place to keep tight control over cytokine production. Adenylate/uridylate-rich elements (ARE) in the 3' untranslated region of cytokine mRNAs serve as a major checkpoint by regulating mRNA stability and translational efficiency. Here, we examined the impact of KH-type splicing regulatory protein (KSRP), an RNA binding protein which destabilizes mRNAs via the ARE, on cytokine expression and paracrine phenotypes of primary astrocytes. We identified a network of inflammatory mediators, including TNF-α and IL-1β, whose expression increased 2 to 4-fold at the RNA level in astrocytes isolated from KSRP(-/-) mice compared to littermate controls. Upon activation, KSRP(-/-) astrocytes produced TNF-α and IL-1β at levels that exceeded control cells by 15-fold or more. Conditioned media from KSRP(-/-) astrocytes induced chemotaxis and neuronal cell death in vitro. Surprisingly, we observed a prolongation of half-life in only a subset of mRNA targets and only after selective astrocyte activation. Luciferase reporter studies indicated that KSRP regulates cytokine gene expression at both transcriptional and post-transcriptional levels. Our results outline a critical role for KSRP in regulating pro-inflammatory mediators and have implications for a wide range of CNS inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Xuelin Li
- Department of Neurology, University of Alabama, Birmingham, Alabama 35233-0017, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Interactive Effects of Morphine on HIV Infection: Role in HIV-Associated Neurocognitive Disorder. AIDS Res Treat 2012; 2012:953678. [PMID: 22666564 PMCID: PMC3362817 DOI: 10.1155/2012/953678] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/13/2012] [Accepted: 03/02/2012] [Indexed: 01/06/2023] Open
Abstract
HIV epidemic continues to be a severe public health problem and concern within USA and across the globe with about 33 million people infected with HIV. The frequency of drug abuse among HIV infected patients is rapidly increasing and is another major issue since injection drug users are at a greater risk of developing HIV associated neurocognitive dysfunctions compared to non-drug users infected with HIV. Brain is a major target for many of the recreational drugs and HIV. Evidences suggest that opiate drug abuse is a risk factor in HIV infection, neural dysfunction and progression to AIDS. The information available on the role of morphine as a cofactor in the neuropathogenesis of HIV is scanty. This review summarizes the results that help in understanding the role of morphine use in HIV infection and neural dysfunction. Studies show that morphine enhances HIV-1 infection by suppressing IL-8, downregulating chemokines with reciprocal upregulation of HIV coreceptors. Morphine also activates MAPK signaling and downregulates cAMP response element-binding protein (CREB). Better understanding on the role of morphine in HIV infection and mechanisms through which morphine mediates its effects may help in devising novel therapeutic strategies against HIV-1 infection in opiate using HIV-infected population.
Collapse
|
43
|
Rehni AK, Singh N. Ammonium pyrrolidine dithiocarbamate and RS 102895 attenuate opioid withdrawal in vivo and in vitro. Psychopharmacology (Berl) 2012; 220:427-38. [PMID: 21931991 DOI: 10.1007/s00213-011-2489-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 09/05/2011] [Indexed: 01/25/2023]
Abstract
RATIONALE Recently, nuclear factor kappa B is indicated in the precipitation of opioid withdrawal syndrome. NF-κB activation is noted to control the transcription and biochemical activation of chemokines. Opioid receptor activation-linked chemokine stimulation is reported to mediate certain effects produced by prolonged opioid treatment. Ammonium pyrrolidine dithiocarbamate (APD) and RS 102895 are relatively selective inhibitors of NF-κB and C-C chemokine receptor 2, respectively. OBJECTIVES The present study investigates the effect of APD and RS 102895 on morphine withdrawal signs in vitro and in vivo. MATERIALS AND METHODS Morphine was administered twice daily for 5 days, following which a single day 6 injection of naloxone (8 mg/kg, i.p.) precipitated opioid withdrawal syndrome in mice. Withdrawal syndrome was quantitatively assessed in terms of withdrawal severity score and the frequency of jumping, rearing, fore paw licking and circling. Naloxone-induced contraction in morphine-withdrawn isolated rat ileum was employed as an in vitro model. An isobolographic study design was employed in the two models to assess potential synergistic activity between APD and RS 102895. RESULTS APD and RS 102895 dose-dependently attenuated naloxone-induced morphine withdrawal syndrome both in vivo and in vitro. APD was also observed to exert a synergistic interaction with RS 102895. CONCLUSIONS It is concluded that APD and RS 102895 attenuate morphine withdrawal signs possibly by a NF-κB and C-C chemokine receptor 2 activation pathway-linked mechanisms potentially in an interdependent manner.
Collapse
Affiliation(s)
- Ashish K Rehni
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, India
| | | |
Collapse
|
44
|
Chemokines and cytokines in neuroinflammation leading to neuropathic pain. Curr Opin Pharmacol 2012; 12:55-61. [DOI: 10.1016/j.coph.2011.10.007] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 10/05/2011] [Indexed: 12/20/2022]
|
45
|
Schwarz JM, Hutchinson MR, Bilbo SD. Early-life experience decreases drug-induced reinstatement of morphine CPP in adulthood via microglial-specific epigenetic programming of anti-inflammatory IL-10 expression. J Neurosci 2011; 31:17835-47. [PMID: 22159099 PMCID: PMC3259856 DOI: 10.1523/jneurosci.3297-11.2011] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/26/2011] [Accepted: 10/26/2011] [Indexed: 01/29/2023] Open
Abstract
A critical component of drug addiction research involves identifying novel biological mechanisms and environmental predictors of risk or resilience to drug addiction and associated relapse. Increasing evidence suggests microglia and astrocytes can profoundly affect the physiological and addictive properties of drugs of abuse, including morphine. We report that glia within the rat nucleus accumbens (NAcc) respond to morphine with an increase in cytokine/chemokine expression, which predicts future reinstatement of morphine conditioned place preference (CPP) following a priming dose of morphine. This glial response to morphine is influenced by early-life experience. A neonatal handling paradigm that increases the quantity and quality of maternal care significantly increases baseline expression of the anti-inflammatory cytokine IL-10 within the NAcc, attenuates morphine-induced glial activation, and prevents the subsequent reinstatement of morphine CPP in adulthood. IL-10 expression within the NAcc and reinstatement of CPP are negatively correlated, suggesting a protective role for this specific cytokine against morphine-induced glial reactivity and drug-induced reinstatement of morphine CPP. Neonatal handling programs the expression of IL-10 within the NAcc early in development, and this is maintained into adulthood via decreased methylation of the IL-10 gene specifically within microglia. The effect of neonatal handling is mimicked by pharmacological modulation of glia in adulthood with ibudilast, which increases IL-10 expression, inhibits morphine-induced glial activation within the NAcc, and prevents reinstatement of morphine CPP. Taken together, we have identified a novel gene × early-life environment interaction on morphine-induced glial activation and a specific role for glial activation in drug-induced reinstatement of drug-seeking behavior.
Collapse
Affiliation(s)
- Jaclyn M Schwarz
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina 27705, USA.
| | | | | |
Collapse
|
46
|
Kong LL, Hu JF, Zhang W, Yuan YH, Ma KL, Han N, Chen NH. Expression of chemokine-like factor 1 after focal cerebral ischemia in the rat. Neurosci Lett 2011; 505:14-18. [PMID: 21964493 DOI: 10.1016/j.neulet.2011.09.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 09/15/2011] [Accepted: 09/15/2011] [Indexed: 11/19/2022]
Abstract
Cerebral ischemic injury is associated with the induction of a series of pro-inflammatory mediators such as cytokines and chemokines. The chemokine-like factor 1(CKLF1), as a novel human cytokine, displays chemotactic activities in a wide spectrum of leukocytes. The present study was conducted to determine if CKLF1 was produced in the brain of rats subjected to transient middle cerebral artery occlusion (TMCAO). Therefore, RT-PCR, Western blot and immunohistochemistry were utilized to characterize the expression of CKLF1 at different times after TMCAO. The result showed that almost no expression of CKLF1 was found in the sham-operated or contralateral cerebral cortex and hippocampus. CKLF1 expression significantly increased in the ischemic cerebral cortex and hippocampus, elevating at 12 h and peaking at 2 days after reperfusion. CKLF1 positive staining was mainly present in the cerebral cortex, hippocampus, thalamus, and hypothalamus. These results demonstrate that the expression of CKLF1 increases after focal cerebral ischemia in rat brain. Thus, CKLF1 may be a potential therapeutic target for cerebral ischemia.
Collapse
Affiliation(s)
- Ling-Lei Kong
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Xuanwu District, Beijing 100050, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Grab DJ, Chakravorty SJ, van der Heyde H, Stins MF. How can microbial interactions with the blood-brain barrier modulate astroglial and neuronal function? Cell Microbiol 2011; 13:1470-8. [DOI: 10.1111/j.1462-5822.2011.01661.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
48
|
Salminen A, Ojala J, Kaarniranta K, Haapasalo A, Hiltunen M, Soininen H. Astrocytes in the aging brain express characteristics of senescence-associated secretory phenotype. Eur J Neurosci 2011; 34:3-11. [PMID: 21649759 DOI: 10.1111/j.1460-9568.2011.07738.x] [Citation(s) in RCA: 248] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cellular stress increases progressively with aging in mammalian tissues. Chronic stress triggers several signaling cascades that can induce a condition called cellular senescence. Recent studies have demonstrated that senescent cells express a senescence-associated secretory phenotype (SASP). Emerging evidence indicates that the number of cells expressing biomarkers of cellular senescence increases in tissues with aging, which implies that cellular senescence is an important player in organismal aging. In the brain, the aging process is associated with degenerative changes, e.g. synaptic loss and white matter atrophy, which lead to progressive cognitive impairment. There is substantial evidence for the presence of oxidative, proteotoxic and metabolic stresses in aging brain. A low-level, chronic inflammatory process is also present in brain during aging. Astrocytes demonstrate age-related changes that resemble those of the SASP: (i) increased level of intermediate glial fibrillary acidic protein and vimentin filaments, (ii) increased expression of several cytokines and (iii) increased accumulation of proteotoxic aggregates. In addition, in vitro stress evokes a typical senescent phenotype in cultured astrocytes and, moreover, isolated astrocytes from aged brain display the proinflammatory phenotype. All of these observations indicate that astrocytes are capable of triggering the SASP and the astrocytes in aging brain display typical characteristics of cellular senescence. Bearing in mind the many functions of astrocytes, it is evident that the age-related senescence of astrocytes enhances the decline in functional capacity of the brain. We will review the astroglial changes occurring during aging and emphasize that senescent astrocytes can have an important role in age-related neuroinflammation and neuronal degeneration.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Atypical chemokine receptors (ACRs) are cell surface receptors with seven transmembrane domains structurally homologous to chemokine G-protein coupled receptors (GPCRs). However, upon ligation by cognate chemokines, ACRs fail to induce classical signaling and downstream cellular responses characteristic for GPCRs. Despite this, by affecting chemokine availability and function, ACRs impact on a multitude of pathophysiological events and have emerged as important molecular players in health and disease. This review discusses individual characteristics of the currently known ACRs, highlights their similarities and differences and attempts to establish their group identity. It summarizes the progress made in mapping ACR expression, understanding their diverse in vitro and in vivo functions of ACRs and uncovering their contributions to disease pathogeneses.
Collapse
Affiliation(s)
| | | | - Antal Rot
- MRC Centre for Immune Regulation, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
50
|
Immunohistochemical localization of chemokine CXCL14 in rat hypothalamic neurons. Neurosci Lett 2011; 487:335-40. [DOI: 10.1016/j.neulet.2010.10.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 10/11/2010] [Accepted: 10/19/2010] [Indexed: 11/23/2022]
|