1
|
Celorrio M, Shumilov K, Ni A, Self WK, Vitorino FNL, Rodgers R, Schriefer LA, Garcia B, Layden BT, Egervari G, Baldridge MT, Friess SH. Short-chain fatty acids are a key mediator of gut microbial regulation of T cell trafficking and differentiation after traumatic brain injury. RESEARCH SQUARE 2024:rs.3.rs-5397327. [PMID: 39606443 PMCID: PMC11601855 DOI: 10.21203/rs.3.rs-5397327/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The gut microbiota has emerged as a pivotal regulator of host inflammatory processes after traumatic brain injury (TBI). However, the mechanisms by which the gut microbiota communicates to the brain in TBI are still under investigation. We previously reported that gut microbiota depletion (GMD) using antibiotics after TBI resulted in increased microglial activation, reduced neurogenesis, and reduced T cell infiltration. In the present study, we have demonstrated that intestinal T cells contribute to the pool of cells infiltrating the brain after TBI. Depletion or genetic deletion of T cells before injury reversed GMD induced reductions in post-TBI neurogenesis. Short-chain fatty acid supplementation increased T regulatory and T helper1 cell infiltration to the brain along with restoring neurogenesis and microglia activation after TBI with GMD. These data suggest that T cell subsets are essential cellular mediators by which the gut microbiota modulates TBI pathogenesis, a finding with important therapeutic implications.
Collapse
Affiliation(s)
| | | | - Allen Ni
- Washington University in St. Louis School of Medicine
| | | | | | | | | | - Ben Garcia
- Washington University in St. Louis School of Medicine
| | | | | | | | | |
Collapse
|
2
|
Wang P, Hu J, Chen C, Jiang Z, Zhang Y, Lin K, Liao L, Wang X. The immune regulatory mechanism of ketamine-induced psychiatric disorders: A new perspective on drug-induced psychiatric symptoms. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111194. [PMID: 39542202 DOI: 10.1016/j.pnpbp.2024.111194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Ketamine, a psychoactive substance strictly regulated by international drug conventions, is classified as a "new type drug" due to its excitatory, hallucinogenic, or inhibitory effects. The etiology of ketamine-induced psychiatric symptoms is multifaceted, with the immune regulatory mechanism being the most prominent among several explanatory theories. In recent years, the interaction between the immune system and nervous system have garnered significant attention in neuropsychiatric disorder research. Notably, the infiltration of peripheral lymphocytes into the central nervous system has emerged as an early hallmark of certain neuropsychiatric disorders. However, a notable gap exists in the current literature, regarding the immune regulatory mechanisms, specifically the peripheral immune alterations, associated with ketamine-induced psychiatric symptoms. To address this void, this article endeavors to provide a comprehensive overview of the pathophysiological processes implicated in psychiatric disorders or symptoms, encompassing those elicited by ketamine. This analysis delves into aspects such as nerve damage, alterations within the central immune system, and the regulation of the peripheral immune system. By emphasizing the intricate crosstalk between the peripheral immune system and the central nervous system, this study sheds light on their collaborative role in the onset and progression of psychiatric diseases or symptoms. This insight offers fresh perspectives on the underlying mechanisms, diagnosis and therapeutic strategies for mental disorders stemming from drug abuse.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Kexin Lin
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Wang P, Jiang L, Hu J, Jiang Z, Zhang Y, Chen C, Lin Y, Su M, Wang X, Liao L. The amino acid metabolism pathway of peripheral T lymphocytes and ketamine-induced schizophrenia-like phenotype. J Psychiatry Neurosci 2024; 49:E413-E426. [PMID: 39626901 PMCID: PMC11633891 DOI: 10.1503/jpn-240038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND The intricate interplay between peripheral adaptive immune cells and the central nervous system (CNS) has garnered increasing recognition. Given that alterations in cell quantities often translate into modifications in metabolite profiles and that these metabolic changes can potentially traverse the bloodstream and enter the CNS, thereby modulating the progression of mental illnesses, we sought to explore the metabolic profiles of peripheral immune cells in a ketamine-treated mouse model of schizophrenia. METHODS We used flow cytometry to scrutinize the alterations in peripheral adaptive immune cells in a ketamine-induced schizophrenia mouse model. Subsequently, we implemented an untargeted metabolomic approach with ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) to detect the metabolite profiles of peripheral abnormal lymphocytes and identify differential metabolites present in plasma. We then employed targeted metabolomics using UPLC-MS/MS to quantify the common differential metabolites detected in mouse plasma. RESULTS Flow cytometry analysis detected a notable increase in the count of peripheral CD3+ T cells in a ketamine-induced schizophrenia mouse model. Subsequent untargeted metabolomics analysis revealed that the amino acid metabolism pathway underwent substantial alterations. A detailed quantification of 22 amino acid profiles in the peripheral plasma indicated significant elevation in the levels of glycine, alanine, asparagine, and aspartic acid. LIMITATIONS Our ongoing research has yet to conclusively identify the precise amino acid metabolism pathway that serves as the pivotal factor in the manifestation of the schizophrenia-like phenotype induced by ketamine. CONCLUSION The peripheral amino acid metabolism pathway is involved in the ketamine-induced schizophrenia-like phenotype. The metabolic profile of peripheral immune cells could provide accurate biomarkers for the diagnosis and treatment of psychiatric diseases.
Collapse
Affiliation(s)
- Peipei Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Linzhi Jiang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yanchen Lin
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Mi Su
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xia Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Shabat Y, Ilan Y. Correlations between components of the immune system. F1000Res 2024; 10:1174. [PMID: 38628268 PMCID: PMC11019305 DOI: 10.12688/f1000research.54487.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/19/2024] Open
Abstract
Background No evidence of the possibility of altering a constituent of the immune system without directly affecting one of its associated components has yet been shown. Methods A schematic model was developed in which two triggers, fasting and splenectomy, were studied for their ability to affect the expression of cell membrane epitopes and the cytokine secretion of out-of-body autogeneic and syngeneic lymphocytes. Results The effect of fasting and/or splenectomy on promoting correlations between immune systems was studied by determining the alterations in expressions of cell membrane epitopes and in cytokine secretion by out-of-body autogeneic and syngeneic lymphocytes. The effect of fasting as a trigger decreased expression of CD8 and CD25 and increased TNFα levels. The effect of splenectomy as a trigger was investigated in non-fasting mice by comparing splenectomized and non-splenectomized mice. An increase in the CD8 expression and in TNFα, IFNg, and IL10 secretion was noted. The effect of splenectomy as a trigger in fasting mice was determined by comparing splenectomized and non-splenectomized mice. Splenectomy significantly affected the expression of CD25 and CD4 CD25 and on secretion of TNFα, IFNg, and IL10. To determine the effect of keeping the cells in an out-of-body location on the expression of lymphocyte epitopes, tubes kept on top of the cages of the fasting mice were compared with tubes kept on top of empty cages. The results showed a significant change in the CD8 expression was noted. To determine the effect of keeping cells in an out-of-body location on cytokine secretion, tubes kept on cages were tested for cytokine levels significant decrease was noted in the secretion of TNFα and IFNg. Conclusions The study showed that a mouse could affect cells at a distance and alter the expression of surface markers and cytokine secretion following two types of triggers: fasting and/or splenectomy. The data characterized a system for the induction of correlations between two's immune system components without a transfer of mediators. It suggests that an out-of-body correlation can be induced between two components of the immune system.
Collapse
Affiliation(s)
- Yehudit Shabat
- Hadassah University Hospital, Jerusalem, Jerusalem, Israel, Israel
| | - Yaron Ilan
- Hadassah University Hospital, Jerusalem, Jerusalem, Israel, Israel
| |
Collapse
|
5
|
Wu C, Pan Y, Wang L, Liu M, Tu P, Chen S, Shi L, Yan D, Ma Y, Guo Y. Inhibition of HDAC6 promotes microvascular endothelial cells to phagocytize myelin debris and reduces inflammatory response to accelerate the repair of spinal cord injury. CNS Neurosci Ther 2024; 30:e14439. [PMID: 37641882 PMCID: PMC10916453 DOI: 10.1111/cns.14439] [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: 01/12/2023] [Revised: 05/31/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
AIMS To identify an effective strategy for promoting microvascular endothelial cells (MECs) to phagocytize myelin debris and reduce secretion of inflammatory factors following spinal cord injury (SCI). METHODS We established a coculture model of myelin debris and vascular-like structures. The efficiency with which MECs phagocytize myelin debris under different conditions was examined via ELISA, flow cytometry, and immunofluorescence. Tubastatin-A was used to interfere with the coculture model. The anti-inflammatory effects of Tubastatin-A were observed by HE staining, flow cytometry, immunofluorescence, and ELISA. RESULTS MECs phagocytized myelin debris via IgM opsonization, and phagocytosis promoted the secretion of inflammatory factors, whereas IgG-opsonized myelin debris had no effect on inflammatory factors. Application of the HDAC6 inhibitor Tubastatin-A increased the IgG levels and decreased the IgM levels by regulating the proliferation and differentiation of B cells. Tubastatin-A exerted a regulatory effect on the HDAC6-mediated autophagy-lysosome pathway, promoting MECs to phagocytize myelin debris, reducing the secretion of inflammatory factors, and accelerating the repair of SCI. CONCLUSIONS Inhibition of HDAC6 to regulate the immune-inflammatory response and promote MECs to phagocytize myelin debris may represent a novel strategy in the treatment of SCI.
Collapse
Affiliation(s)
- Chengjie Wu
- Department of Traumatology and OrthopedicsAffiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & OrthopedicsNanjing University of Chinese MedicineNanjingChina
| | - Yalan Pan
- Laboratory of Chinese Medicine Nursing Intervention for Chronic DiseasesNanjing University of Chinese MedicineNanjingChina
| | - Lining Wang
- School of Chinese Medicine, School of Integrated Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Mengmin Liu
- School of Chinese Medicine, School of Integrated Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Pengcheng Tu
- Department of Traumatology and OrthopedicsAffiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & OrthopedicsNanjing University of Chinese MedicineNanjingChina
| | - Sixian Chen
- School of Chinese Medicine, School of Integrated Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Lei Shi
- School of Chinese Medicine, School of Integrated Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Danqing Yan
- Department of Traumatology and OrthopedicsAffiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & OrthopedicsNanjing University of Chinese MedicineNanjingChina
| | - Yong Ma
- Department of Traumatology and OrthopedicsAffiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & OrthopedicsNanjing University of Chinese MedicineNanjingChina
- School of Chinese Medicine, School of Integrated Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Yang Guo
- Department of Traumatology and OrthopedicsAffiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
- Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & OrthopedicsNanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
6
|
Wang Y, Jiang R, Li M, Wang Z, Yang Y, Sun L. Characteristics of T Cells in Single-Cell Datasets of Peripheral Blood and Cerebrospinal Fluid in Alzheimer's Disease Patients. J Alzheimers Dis 2024; 99:S265-S280. [PMID: 38043012 PMCID: PMC11091562 DOI: 10.3233/jad-230784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 12/04/2023]
Abstract
Background Alzheimer's disease (AD) is the most common type of dementia, causing a huge socioeconomic burden. In parallel with the widespread uptake of single-cell RNA sequencing (scRNA-seq) technology, there has been a rapid accumulation of data produced by researching AD at single-cell resolution, which is more conductive to explore the neuroimmune-related mechanism of AD. Objective To explore the potential features of T cells in the peripheral blood and cerebrospinal fluid of AD patients. Methods Two datasets, GSE181279 and GSE134578, were integrated from GEO database. Seurat, Monocle, CellChat, scRepertoire, and singleR packages were mainly employed for data analysis. Results Our analysis demonstrated that in peripheral blood, T cells were significantly expanded, and these expanded T cells were possessed effector function, such as CD8+TEMRA, CD4+TEMRA, and CD8+TEM. Interestingly, CD8+TEMRA and CD4+TEMRA cells positioned adjacently after dimensions reduction and clustering. Notably, we identified that the expanded T cells were developed from Naïve T cells and TCM cells, and TEM cells was in the intermediate state of this developing process. Additionally, in cerebrospinal fluid of AD patients, the amplified T cells were mainly CD8+TEMRA cells, and the number and strength of communication between CD4+TEM, CD8+TEM, and CD8+TEMRA were decreased in AD patients. Conclusions Our comprehensive analyses identified the cells in cerebrospinal fluid from AD patients are expanded TEMRA or TEM cells and the TEMRA cells communicating with other immune cells is weakened, which may be an important immune feature that leads to AD.
Collapse
Affiliation(s)
- Yongchun Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Richeng Jiang
- Department of Otolaryngology Head and Neck Surgery, The First Hospital of Jilin University, Changchun, China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zicheng Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yu Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
7
|
Sun Y, Ho CT, Liu Y, Zhan S, Wu Z, Zheng X, Zhang X. The Modulatory Effect of Cyclocarya paliurus Flavonoids on Intestinal Microbiota and Hypothalamus Clock Genes in a Circadian Rhythm Disorder Mouse Model. Nutrients 2022; 14:nu14112308. [PMID: 35684108 PMCID: PMC9182649 DOI: 10.3390/nu14112308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 01/27/2023] Open
Abstract
Circadian rhythm disruption is detrimental and results in adverse health consequences. We used a multi-omics profiling approach to investigate the effects of Cyclocarya paliurus flavonoid (CPF)-enriched diets on gut microbiota, metabolites, and hypothalamus clock genes in mice with induced circadian rhythm disruption. It was observed that CPF supplementation altered the specific composition and function of gut microbiota and metabolites induced by circadian rhythm disruption. Analysis showed that the abundance of Akkermansia increased, while the abundance of Clostridiales and Ruminiclostridium displayed a significant downward trend after the CPF intervention. Correlation analysis also revealed that these gut microbes had certain correlations with the metabolites, suggesting that CPFs help the intestinal microbiota to repair the intestinal environment and modulate the release of some beneficial metabolites. Notably, single-cell RNA-seq revealed that CPF supplementation significantly regulated the expression of genes associated with circadian rhythm, myelination, and neurodegenerative diseases. Altogether, these findings highlight that CPFs may represent a promising dietary therapeutic strategy for treating circadian rhythm disruption.
Collapse
Affiliation(s)
- Ying Sun
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Y.S.); (Y.L.); (S.Z.); (Z.W.)
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
- Correspondence: (C.-T.H.); (X.Z.); (X.Z.)
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Y.S.); (Y.L.); (S.Z.); (Z.W.)
| | - Shennan Zhan
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Y.S.); (Y.L.); (S.Z.); (Z.W.)
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Y.S.); (Y.L.); (S.Z.); (Z.W.)
| | - Xiaojie Zheng
- Department of Agriculture and Biotechnology, Wenzhou Vocational College of Science and Technology, Wenzhou 325006, China
- Correspondence: (C.-T.H.); (X.Z.); (X.Z.)
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (Y.S.); (Y.L.); (S.Z.); (Z.W.)
- Correspondence: (C.-T.H.); (X.Z.); (X.Z.)
| |
Collapse
|
8
|
Wu L, Li S, Li C, He B, Lv L, Wang J, Wang J, Wang W, Zhang Y. The role of regulatory T cells on the activation of astrocytes in the brain of high-fat diet mice following lead exposure. Chem Biol Interact 2022; 351:109740. [PMID: 34742682 DOI: 10.1016/j.cbi.2021.109740] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 10/11/2021] [Accepted: 11/01/2021] [Indexed: 01/02/2023]
Abstract
Lead (Pb) exposure can cause damage to the central nervous system (CNS)*. Pb can accumulate in the hippocampus, leading to learning and memory impairments. Recent studies have shown that high-fat diet (HFD) is also associated with cognitive impairment. However, there are few reports on CNS damage due to HFD and Pb exposure. We aimed to investigate the effect of Pb on cognitive functions of HFD-fed mice, focusing on the role of regulatory T (Treg) cells in astrocyte activation. C57BL/6J mice were randomly divided into control, HFD, Pb, and HFD + Pb groups. TGF-β and IL-10 secreted by Treg cells and the intracellular transcription factor Foxp3 were evaluated as a measure of Treg cell function; astrocyte activation was assessed by evaluating glial fibrillary acidic protein (GFAP) expression. The learning and memory ability was significantly lower in the HFD + Pb group than in other groups. The brain Treg cell ratio was significantly decreased and the protein levels of TGF-β, IL-10, and Foxp3 were significantly lower, whereas the protein level of GFAP was higher in the HFD + Pb group. The hippocampus of the HFD + Pb group mice showed significantly higher levels of neurotoxic reactive astrocyte markers and astrogliosis was also much higher compared to HFD and Pb groups. Furthermore, all-trans retinoic acid treatment increased the brain Treg cell ratio, reversed cognitive decline, and suppressed astrocyte activation in the HFD + Pb group mice. We concluded that HFD along with Pb exposure could aggravate the activation of astrocytes in the brain, and the brain Treg cells may be involved in inhibiting astrocyte activation in HFD-fed mice exposed to Pb.
Collapse
Affiliation(s)
- Lei Wu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Shuang Li
- Experiment Animal Center, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Chao Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Bin He
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Linyi Lv
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Jia Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Jierui Wang
- Rheumatology Department, Kailuan General Hospital, Tangshan, 063000, Hebei, China
| | - Weixuan Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China; Experiment Animal Center, North China University of Science and Technology, Tangshan, 063210, Hebei, China.
| |
Collapse
|
9
|
Li Z, Wang Q, Hu H, Zheng W, Gao C. Research advances of biomaterials-based microenvironment-regulation therapies for repair and regeneration of spinal cord injury. Biomed Mater 2021; 16. [PMID: 34384071 DOI: 10.1088/1748-605x/ac1d3c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022]
Abstract
Traumatic spinal cord injury (SCI) usually results in restricted behaviour recovery and even life-changing paralysis, accompanied with numerous complications. Pathologically, the initial injuries trigger a series of secondary injuries, leading to an expansion of lesion site, a mass of neuron loss, and eventual failure of endogenous axon regeneration. As the advances rapidly spring up in regenerative medicine and tissue engineering biomaterials, regulation of these secondary injuries becomes possible, shedding a light on normal functional restoration. The successful tissue regeneration lies in proper regulation of the inflammatory microenvironment, including the inflammatory immune cells and inflammatory factors that lead to oxidative stress, inhibitory glial scar and neuroexcitatory toxicity. Specifically, the approaches based on microenvironment-regulating biomaterials have shown great promise in the repair and regeneration of SCI. In this review, the pathological inflammatory microenvironments of SCI are discussed, followed by the introduction of microenvironment-regulating biomaterials in terms of their impressive therapeutic effect in attenuation of secondary inflammation and promotion of axon regrowth. With the emphasis on regulating secondary events, the biomaterials for SCI treatment will become promising for clinical applications.
Collapse
Affiliation(s)
- Ziming Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China.,Dr Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
10
|
Chagas LDS, Sandre PC, Ribeiro e Ribeiro NCA, Marcondes H, Oliveira Silva P, Savino W, Serfaty CA. Environmental Signals on Microglial Function during Brain Development, Neuroplasticity, and Disease. Int J Mol Sci 2020; 21:ijms21062111. [PMID: 32204421 PMCID: PMC7139373 DOI: 10.3390/ijms21062111] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/15/2022] Open
Abstract
Recent discoveries on the neurobiology of the immunocompetent cells of the central nervous system (CNS), microglia, have been recognized as a growing field of investigation on the interactions between the brain and the immune system. Several environmental contexts such as stress, lesions, infectious diseases, and nutritional and hormonal disorders can interfere with CNS homeostasis, directly impacting microglial physiology. Despite many encouraging discoveries in this field, there are still some controversies that raise issues to be discussed, especially regarding the relationship between the microglial phenotype assumed in distinct contexts and respective consequences in different neurobiological processes, such as disorders of brain development and neuroplasticity. Also, there is an increasing interest in discussing microglial–immune system cross-talk in health and in pathological conditions. In this review, we discuss recent literature concerning microglial function during development and homeostasis. In addition, we explore the contribution of microglia to synaptic disorders mediated by different neuroinflammatory outcomes during pre- and postnatal development, with long-term consequences impacting on the risk and vulnerability to the emergence of neurodevelopmental, neurodegenerative, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
| | - Poliana Capucho Sandre
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Natalia Cristina Aparecida Ribeiro e Ribeiro
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
| | - Henrique Marcondes
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
| | - Priscilla Oliveira Silva
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
- National Institute of Science and Technology on Neuroimmunomodulation –INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation –INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Correspondence: (W.S.); (C.A.S.)
| | - Claudio A. Serfaty
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
- National Institute of Science and Technology on Neuroimmunomodulation –INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Correspondence: (W.S.); (C.A.S.)
| |
Collapse
|
11
|
Abstract
Regulatory T cells (Tregs) are important for limiting inflammation-dependent damage in neural tissue. However, Tregs have also been shown to inhibit neural repair associated with type 2 (anti-inflammatory/wound healing) immune responses. Recently, it was demonstrated that Sirtuins, a family of proteins that contribute to the control of cellular responses to metabolic stimuli, influence the functions of Tregs. Specifically, SIRT4 was found to suppress the anti-neuroinflammatory activity of Tregs infiltrating the spinal cord following injury; when SIRT4 expression was genetically suppressed, Tregs made more anti-inflammatory factors, IL-10, FoxP3, and transforming growth factor beta (TGFβ). Thus, understanding how the SIRT4-Treg pathway can be manipulated could provide useful avenues to control both pathogenic and neuroprotective immune responses.
Collapse
Affiliation(s)
- Simon Milling
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Julia M Edgar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
12
|
Mittal K, Eremenko E, Berner O, Elyahu Y, Strominger I, Apelblat D, Nemirovsky A, Spiegel I, Monsonego A. CD4 T Cells Induce A Subset of MHCII-Expressing Microglia that Attenuates Alzheimer Pathology. iScience 2019; 16:298-311. [PMID: 31203186 PMCID: PMC6581663 DOI: 10.1016/j.isci.2019.05.039] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/01/2019] [Accepted: 05/28/2019] [Indexed: 01/10/2023] Open
Abstract
Microglia play a key role in innate immunity in Alzheimer disease (AD), but their role as antigen-presenting cells is as yet unclear. Here we found that amyloid β peptide (Aβ)-specific T helper 1 (Aβ-Th1 cells) T cells polarized to secrete interferon-γ and intracerebroventricularly (ICV) injected to the 5XFAD mouse model of AD induced the differentiation of major histocompatibility complex class II (MHCII)+ microglia with distinct morphology and enhanced plaque clearance capacity than MHCII- microglia. Notably, 5XFAD mice lacking MHCII exhibited an enhanced amyloid pathology in the brain along with exacerbated innate inflammation and reduced phagocytic capacity. Using a bone marrow chimera mouse model, we showed that infiltrating macrophages did not differentiate to MHCII+ cells following ICV injection of Aβ-Th1 cells and did not support T cell-mediated amyloid clearance. Overall, we demonstrate that CD4 T cells induce a P2ry12+ MHCII+ subset of microglia, which play a key role in T cell-mediated effector functions that abrogate AD-like pathology.
Collapse
Affiliation(s)
- Kritika Mittal
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ekaterina Eremenko
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Omer Berner
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Yehezqel Elyahu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Itai Strominger
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Daniella Apelblat
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Anna Nemirovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ivo Spiegel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Zlotowski Center for Neuroscience, The National Institute of Biotechnology in the Negev, Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
13
|
Kumar V. Toll-like receptors in the pathogenesis of neuroinflammation. J Neuroimmunol 2019; 332:16-30. [PMID: 30928868 DOI: 10.1016/j.jneuroim.2019.03.012] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Toll-like receptors (TLRs) are discovered as crucial pattern recognition receptors (PRRs) involved in the recognition of pathogen-associated molecular patterns (PAMPs). Later studies showed their involvement in the recognition of various damage/danger-associated molecular patterns (DAMPs) generated by host itself. Thus, TLRs are capable of recognizing wide-array of patterns/molecules derived from pathogens and host as well and initiating a proinflammatory immune response through the activation of NF-κB and other transcription factors causing synthesis of proinflammatory molecules. The process of neuroinflammation is seen under both sterile and infectious inflammatory diseases of the central nervous system (CNS) and may lead to the development of neurodegeneration. The present article is designed to highlight the importance of TLRs in the pathogenesis of neuroinflammation under diverse conditions. TLRs are expressed by various immune cells present in CNS along with neurons. However out of thirteen TLRs described in mammals, some are present and active in these cells, while some are absent and are described in detail in main text. The role of various immune cells present in the brain and their role in the pathogenesis of neuroinflammation depending on the type of TLR expressed is described. Thereafter the role of TLRs in bacterial meningitis, viral encephalitis, stroke, Alzheimer's disease (AD), Parkinson's disease (PD), and autoimmune disease including multiple sclerosis (MS) is described. The article is designed for both neuroscientists needing information regarding TLRs in neuroinflammation and TLR biologists or immunologists interested in neuroinflammation.
Collapse
Affiliation(s)
- V Kumar
- Children Health Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
14
|
Noé FM, Marchi N. Central nervous system lymphatic unit, immunity, and epilepsy: Is there a link? Epilepsia Open 2019; 4:30-39. [PMID: 30868113 PMCID: PMC6398113 DOI: 10.1002/epi4.12302] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/08/2018] [Accepted: 01/06/2019] [Indexed: 12/16/2022] Open
Abstract
The recent definition of a network of lymphatic vessels in the meninges surrounding the brain and the spinal cord has advanced our knowledge on the functional anatomy of fluid movement within the central nervous system (CNS). Meningeal lymphatic vessels along dural sinuses and main nerves contribute to cerebrospinal fluid (CSF) drainage, integrating the cerebrovascular and periventricular routes, and forming a circuit that we here define as the CNS-lymphatic unit. The latter unit is important for parenchymal waste clearance, brain homeostasis, and the regulation of immune or inflammatory processes within the brain. Disruption of fluid drain mechanisms may promote or sustain CNS disease, conceivably applicable to epilepsy where extracellular accumulation of macromolecules and metabolic by-products occur in the interstitial and perivascular spaces. Herein we address an emerging concept and propose a theoretical framework on: (a) how a defect of brain clearance of macromolecules could favor neuronal hyperexcitability and seizures, and (b) whether meningeal lymphatic vessel dysfunction contributes to the neuroimmune cross-talk in epileptic pathophysiology. We propose possible molecular interventions targeting meningeal lymphatic dysfunctions, a potential target for immune-mediated epilepsy.
Collapse
Affiliation(s)
- Francesco M. Noé
- Neuro‐Lymphatic GroupA.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
- Biology of Neuro‐Immune InteractionHiLife‐Neuroscience CenterHelsinki UniversityHelsinkiFinland
| | - Nicola Marchi
- Cerebrovascular Mechanisms of Brain DisordersDepartment of NeuroscienceInstitute of Functional Genomics (UMR5203 CNRS – U1191 INSERM)University of MontpellierMontpellierFrance
| |
Collapse
|
15
|
Clinically relevant and simple immune system measure is related to symptom burden in bipolar disorder. Acta Neuropsychiatr 2018; 30:297-305. [PMID: 29212563 DOI: 10.1017/neu.2017.34] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Immunological theories, particularly the sickness syndrome theory, may explain psychopathology in mood disorders. However, no clinical trials have investigated the association between overall immune system markers with a wide range of specific symptoms including potential gender differences. METHODS We included two similar clinical trials, the lithium treatment moderate-dose use study and clinical and health outcomes initiatives in comparative effectiveness for bipolar disorder study, enrolling 765 participants with bipolar disorder. At study entry, white blood cell (WBC) count was measured and psychopathology assessed with the Montgomery and Aasberg depression rating scale (MADRS). We performed analysis of variance and linear regression analyses to investigate the relationship between the deviation from the median WBC, and multinomial regression analysis between different WBC levels. All analyses were performed gender-specific and adjusted for age, body mass index, smoking, race, and somatic diseases. RESULTS The overall MADRS score increased significantly for each 1.0×109/l deviation from the median WBC among 322 men (coefficient=1.10; 95% CI=0.32-1.89; p=0.006), but not among 443 women (coefficient=0.56; 95% CI=-0.19-1.31; p=0.14). Among men, WBC deviations were associated with increased severity of sadness, inner tension, reduced sleep, reduced appetite, concentration difficulties, inability to feel, and suicidal thoughts. Among women, WBC deviations were associated with increased severity of reduced appetite, concentration difficulties, lassitude, inability to feel, and pessimistic thoughts. Both higher and lower WBC levels were associated with increased severity of several specific symptoms. CONCLUSION Immune system alterations were associated with increased severity of specific mood symptoms, particularly among men. Our results support the sickness syndrome theory, but furthermore emphasise the relevance to study immune suppression in bipolar disorder. Due to the explorative nature and cross-sectional design, future studies need to confirm these findings.
Collapse
|
16
|
Frank MG, Fonken LK, Dolzani SD, Annis JL, Siebler PH, Schmidt D, Watkins LR, Maier SF, Lowry CA. Immunization with Mycobacterium vaccae induces an anti-inflammatory milieu in the CNS: Attenuation of stress-induced microglial priming, alarmins and anxiety-like behavior. Brain Behav Immun 2018; 73:352-363. [PMID: 29807129 PMCID: PMC6129419 DOI: 10.1016/j.bbi.2018.05.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/17/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
Exposure to stressors induces anxiety- and depressive-like behaviors, which are mediated, in part, by neuroinflammatory processes. Recent findings demonstrate that treatment with the immunoregulatory and anti-inflammatory bacterium, Mycobacterium vaccae (M. vaccae), attenuates stress-induced exaggeration of peripheral inflammation and stress-induced anxiety-like behavioral responses. However, the effects of M. vaccae on neuroimmune processes have largely been unexplored. In the present study, we examined the effect of M. vaccae NCTC11659 on neuroimmune regulation, stress-induced neuroinflammatory processes and anxiety-like behavior. Adult male rats were immunized 3× with a heat-killed preparation of M. vaccae (0.1 mg, s.c.) or vehicle. M. vaccae induced an anti-inflammatory immunophenotype in hippocampus (increased interleukin (Il)4, Cd200r1, and Mrc1 mRNA expression) and increased IL4 protein 8 d after the last immunization. Central administration of recombinant IL4 recapitulated the effects of M. vaccae on Cd200r1 and Mrc1 mRNA expression. M. vaccae reduced basal levels of genes (Nlrp3 and Nfkbia) involved in microglial priming; thus, we explored the effects of M. vaccae on stress-induced hippocampal microglial priming and HMGB1, which mediates priming. We found that M. vaccae blocked stress-induced decreases in Cd200r1, increases in the alarmin HMGB1, and priming of the microglial response to immune challenge. Furthermore, M. vaccae prevented stress-induced increases in anxiety-like behavior. The present findings suggest that M. vaccae enhances immunomodulation in the CNS and mitigates the neuroinflammatory and behavioral effects of stress, which may underpin its capacity to impart a stress resilient phenotype.
Collapse
Affiliation(s)
- Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
| | - Samuel D Dolzani
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jessica L Annis
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Philip H Siebler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Dominic Schmidt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Christopher A Lowry
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80220, USA
| |
Collapse
|
17
|
Köhler-Forsberg O, He W, Chang Y, Atlas SJ, Meigs JB, Nierenberg AA. White blood cell count at first depression diagnosis as predictor for risk of subsequent hospitalization with depression. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.npbr.2017.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
18
|
Köhler O, Sylvia LG, Bowden CL, Calabrese JR, Thase M, Shelton RC, McInnis M, Tohen M, Kocsis JH, Ketter TA, Friedman ES, Deckersbach T, Ostacher MJ, Iosifescu DV, McElroy S, Nierenberg AA. White blood cell count correlates with mood symptom severity and specific mood symptoms in bipolar disorder. Aust N Z J Psychiatry 2017; 51:355-365. [PMID: 27126391 DOI: 10.1177/0004867416644508] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Immune alterations may play a role in bipolar disorder etiology; however, the relationship between overall immune system functioning and mood symptom severity is unknown. METHODS The two comparative effectiveness trials, the Clinical and Health Outcomes Initiatives in Comparative Effectiveness for Bipolar Disorder Study (Bipolar CHOICE) and the Lithium Treatment Moderate-Dose Use Study (LiTMUS), were similar trials among patients with bipolar disorder. At study entry, white blood cell count and bipolar mood symptom severity (via Montgomery-Aasberg Depression Rating Scale and Bipolar Inventory of Symptoms Scale) were assessed. We performed analysis of variance and linear regression analyses to investigate relationships between deviations from median white blood cell and multinomial regression analysis between higher and lower white blood cell levels. All analyses were adjusted for age, gender, body mass index, smoking, diabetes, hypertension and hyperlipidemia. RESULTS Among 482 Bipolar CHOICE participants, for each 1.0 × 109/L white blood cell deviation, the overall Bipolar Inventory of Symptoms Scale severity increased significantly among men (coefficient = 2.13; 95% confidence interval = [0.46, -3.79]; p = 0.013), but not among women (coefficient = 0.87; 95% confidence interval = [-0.87, -2.61]; p = 0.33). Interaction analyses showed a trend toward greater Bipolar Inventory of Symptoms Scale symptom severity among men (coefficient = 1.51; 95% confidence interval = [-0.81, -3.82]; p = 0.2). Among 283 LiTMUS participants, higher deviation from the median white blood cell showed a trend toward higher Montgomery-Aasberg Depression Rating Scale scores among men (coefficient = 1.33; 95% confidence interval = [-0.22, -2.89]; p = 0.09), but not among women (coefficient = 0.34; 95% confidence interval = [-0.64, -1.32]; p = 0.50). When combining LiTMUS and Bipolar CHOICE, Montgomery-Aasberg Depression Rating Scale scores increased significantly among men (coefficient = 1.09; 95% confidence interval = [0.31, -1.87]; p = 0.006) for each 1.0 × 109/L white blood cell deviation, whereas we found a weak association among women (coefficient = 0.55; 95% confidence interval = [-0.20, -1.29]; p = 0.14). Lower and higher white blood cell levels correlated with greater symptom severity and specific symptoms, varying according to gender. CONCLUSION Deviations in an overall immune system marker, even within the normal white blood cell range, correlated with mood symptom severity in bipolar disorder, mostly among males. Studies are warranted investigating whether white blood cell count may predict response to mood-stabilizing treatment.
Collapse
Affiliation(s)
- Ole Köhler
- 1 Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Louisa G Sylvia
- 2 Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,3 Harvard Medical School, Boston, MA, USA
| | - Charles L Bowden
- 4 Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Joseph R Calabrese
- 5 Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - Michael Thase
- 6 Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard C Shelton
- 7 Department of Psychiatry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melvin McInnis
- 8 Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Mauricio Tohen
- 9 Department of Psychiatry, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - James H Kocsis
- 10 Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| | - Terence A Ketter
- 11 Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, USA
| | - Edward S Friedman
- 12 Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Thilo Deckersbach
- 2 Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,3 Harvard Medical School, Boston, MA, USA
| | - Michael J Ostacher
- 11 Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, USA.,13 VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Dan V Iosifescu
- 14 Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan McElroy
- 15 Department of Psychiatry and Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.,16 Lindner Center of HOPE, Mason, OH, USA
| | - Andrew A Nierenberg
- 2 Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,3 Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Braun M, Vaibhav K, Saad N, Fatima S, Brann DW, Vender JR, Wang LP, Hoda MN, Baban B, Dhandapani KM. Activation of Myeloid TLR4 Mediates T Lymphocyte Polarization after Traumatic Brain Injury. THE JOURNAL OF IMMUNOLOGY 2017; 198:3615-3626. [PMID: 28341672 DOI: 10.4049/jimmunol.1601948] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/21/2017] [Indexed: 12/15/2022]
Abstract
Traumatic brain injury (TBI) is a major public health issue, producing significant patient mortality and poor long-term outcomes. Increasing evidence suggests an important, yet poorly defined, role for the immune system in the development of secondary neurologic injury over the days and weeks following a TBI. In this study, we tested the hypothesis that peripheral macrophage infiltration initiates long-lasting adaptive immune responses after TBI. Using a murine controlled cortical impact model, we used adoptive transfer, transgenic, and bone marrow chimera approaches to show increased infiltration and proinflammatory (classically activated [M1]) polarization of macrophages for up to 3 wk post-TBI. Monocytes purified from the injured brain stimulated the proliferation of naive T lymphocytes, enhanced the polarization of T effector cells (TH1/TH17), and decreased the production of regulatory T cells in an MLR. Similarly, elevated T effector cell polarization within blood and brain tissue was attenuated by myeloid cell depletion after TBI. Functionally, C3H/HeJ (TLR4 mutant) mice reversed M1 macrophage and TH1/TH17 polarization after TBI compared with C3H/OuJ (wild-type) mice. Moreover, brain monocytes isolated from C3H/HeJ mice were less potent stimulators of T lymphocyte proliferation and TH1/TH17 polarization compared with C3H/OuJ monocytes. Taken together, our data implicate TLR4-dependent, M1 macrophage trafficking/polarization into the CNS as a key mechanistic link between acute TBI and long-term, adaptive immune responses.
Collapse
Affiliation(s)
- Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA 30912
| | - Nancy Saad
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA 30912
| | - Sumbul Fatima
- Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Lei P Wang
- Department of Psychiatry, Medical College of Georgia, Augusta University, Augusta, GA 30912; and
| | - Md Nasrul Hoda
- Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912; .,Charlie Norwood VA Medical Center, Augusta, GA 30912
| |
Collapse
|
20
|
Haroon E, Miller AH, Sanacora G. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology 2017; 42:193-215. [PMID: 27629368 PMCID: PMC5143501 DOI: 10.1038/npp.2016.199] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/05/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
Increasing data indicate that inflammation and alterations in glutamate neurotransmission are two novel pathways to pathophysiology in mood disorders. The primary goal of this review is to illustrate how these two pathways may converge at the level of the glia to contribute to neuropsychiatric disease. We propose that a combination of failed clearance and exaggerated release of glutamate by glial cells during immune activation leads to glutamate increases and promotes aberrant extrasynaptic signaling through ionotropic and metabotropic glutamate receptors, ultimately resulting in synaptic dysfunction and loss. Furthermore, glutamate diffusion outside the synapse can lead to the loss of synaptic fidelity and specificity of neurotransmission, contributing to circuit dysfunction and behavioral pathology. This review examines the fundamental role of glia in the regulation of glutamate, followed by a description of the impact of inflammation on glial glutamate regulation at the cellular, molecular, and metabolic level. In addition, the role of these effects of inflammation on glia and glutamate in mood disorders will be discussed along with their translational implications.
Collapse
Affiliation(s)
- Ebrahim Haroon
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew H Miller
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Xu K, Yang Z, Shi R, Luo C, Zhang Z. Expression of aryl hydrocarbon receptor in rat brain lesions following traumatic brain injury. Diagn Pathol 2016; 11:72. [PMID: 27506546 PMCID: PMC4977631 DOI: 10.1186/s13000-016-0522-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/31/2016] [Indexed: 01/31/2023] Open
Abstract
Background Aryl Hydrocarbon Receptor (AhR) is a ligand-activated transcription factor with multiple functions operating in a variety of organs, including the brain. Recent studies have revealed that AhR played a functional role in traumatic injuries. This paper aims to study the expression of AhR during the early phase following a traumatic brain injury (TBI) in rat brains by immunohistochemistry. Methods Weight-drop induced TBI was performed in rats. The expression of AhR in brain of TBI rats were examined by immunohistochemistry. Results Neuron expression of AhR in the rat brains of experiment group had been upregulated since day 3 in lesional hemisphere compared to that of the control group and mainly located in the cytoplasm, indicating an inactivated state. Interestingly, the accumulation of AhR+ non-neuron cells became significant as early as 18 h after injury, which had kept increasing until 24 h post injury and then decreased slowly. For AhR+ non-neuron cells, the AhR mainly located in cell nucleus, indicating a reactive status. Furthermore, double staining showed that most AhR+ non-neuron cells co-localized with W3/13, a marker for T lymphocytes, but not with ED-1 (for activated microglia/macrophages) or GFAP (for activated astrocytes), suggesting that most AhR+ non-neuron cells were T lymphocytes. Conclusion This is the first study concerning AhR expression in brains following TBI, and our data demonstrated that AhR was upregulated and activated in T lymphocytes following TBI. More research is needed to make a more conclusive conclusion.
Collapse
Affiliation(s)
- Kai Xu
- Institute of Immunology, Third Military Medical University of PLA, 30 Gaotanyan Main Street, Chongqing, 400038, People's Republic of China
| | - Zicheng Yang
- Institute of Immunology, Third Military Medical University of PLA, 30 Gaotanyan Main Street, Chongqing, 400038, People's Republic of China
| | - Rongchen Shi
- Institute of Immunology, Third Military Medical University of PLA, 30 Gaotanyan Main Street, Chongqing, 400038, People's Republic of China
| | - Chunxia Luo
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Zhiren Zhang
- Institute of Immunology, Third Military Medical University of PLA, 30 Gaotanyan Main Street, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
22
|
Bombeiro AL, Santini JC, Thomé R, Ferreira ERL, Nunes SLO, Moreira BM, Bonet IJM, Sartori CR, Verinaud L, Oliveira ALR. Enhanced Immune Response in Immunodeficient Mice Improves Peripheral Nerve Regeneration Following Axotomy. Front Cell Neurosci 2016; 10:151. [PMID: 27378849 PMCID: PMC4905955 DOI: 10.3389/fncel.2016.00151] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/27/2016] [Indexed: 12/21/2022] Open
Abstract
Injuries to peripheral nerves cause loss of motor and sensory function, greatly affecting life quality. Successful repair of the lesioned nerve requires efficient cell debris removal, followed by axon regeneration and reinnervation of target organs. Such process is orchestrated by several cellular and molecular events in which glial and immune cells actively participate. It is known that tissue clearance is largely improved by macrophages, which activation is potentiated by cells and molecules of the acquired immune system, such as T helper lymphocytes and antibodies, respectively. In the present work, we evaluated the contribution of lymphocytes in the regenerative process of crushed sciatic nerves of immunocompetent (wild-type, WT) and T and B-deficient (RAG-KO) mice. In Knockout animals, we found increased amount of macrophages under basal conditions and during the initial phase of the regenerative process, that was evaluated at 2, 4, and 8 weeks after lesion (wal). That parallels with faster axonal regeneration evidenced by the quantification of neurofilament and a growth associated protein immunolabeling. The motor function, evaluated by the sciatic function index, was fully recovered in both mouse strains within 4 wal, either in a progressive fashion, as observed for RAG-KO mice, or presenting a subtle regression, as seen in WT mice between 2 and 3 wal. Interestingly, boosting the immune response by early adoptive transference of activated WT lymphocytes at 3 days after lesion improved motor recovery in WT and RAG-KO mice, which was not ameliorated when cells were transferred at 2 wal. When monitoring lymphocytes by in vivo imaging, in both mouse strains, cells migrated to the lesion site shortly after transference, remaining in the injured limb up to its complete motor recovery. Moreover, a first peak of hyperalgesia, determined by von-Frey test, was coincident with increased lymphocyte infiltration in the damaged paw. Overall, the present results suggest that a wave of immune cell infiltration takes place during subacute phase of axonal regeneration, resulting in transient set back of motor recovery following peripheral axonal injury. Moreover, modulation of the immune response can be an efficient approach to speed up nerve regeneration.
Collapse
Affiliation(s)
- André L Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Júlio C Santini
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Elisângela R L Ferreira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Sérgio L O Nunes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Bárbara M Moreira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Ivan J M Bonet
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Cesar R Sartori
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| | - Alexandre L R Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas Campinas, Brazil
| |
Collapse
|
23
|
Abstract
AbstractNatural bornavirus infections and their resulting diseases are largely restricted to horses and sheep in Central Europe. The disease also occurs naturally in cats, and can be induced experimentally in laboratory rodents and numerous other mammals. Borna disease virus-1 (BoDV-1), the cause of most cases of mammalian Borna disease, is a negative-stranded RNA virus that replicates within the nucleus of target cells. It causes severe, often lethal, encephalitis in susceptible species. Recent events, especially the discovery of numerous new species of bornaviruses in birds and a report of an acute, lethal bornaviral encephalitis in humans, apparently acquired from squirrels, have revived interest in this remarkable family of viruses. The clinical manifestations of the bornaviral diseases are highly variable. Thus, in addition to acute lethal encephalitis, they can cause persistent neurologic disease associated with diverse behavioral changes. They also cause a severe retinitis resulting in blindness. In this review, we discuss both the pathological lesions observed in mammalian bornaviral disease and the complex pathogenesis of the neurologic disease. Thus infected neurons may be destroyed by T-cell-mediated cytotoxicity. They may die as a result of excessive inflammatory cytokine release from microglia. They may also die as a result of a ‘glutaminergic storm’ due to a failure of infected astrocytes to regulate brain glutamate levels.
Collapse
|
24
|
Oriá RB, Murray-Kolb LE, Scharf RJ, Pendergast LL, Lang DR, Kolling GL, Guerrant RL. Early-life enteric infections: relation between chronic systemic inflammation and poor cognition in children. Nutr Rev 2016; 74:374-86. [PMID: 27142301 DOI: 10.1093/nutrit/nuw008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The intestinal microbiota undergoes active remodeling in the first 6 to 18 months of life, during which time the characteristics of the adult microbiota are developed. This process is strongly influenced by the early diet and enteric pathogens. Enteric infections and malnutrition early in life may favor microbiota dysbiosis and small intestinal bacterial overgrowth, resulting in intestinal barrier dysfunction and translocation of intestinal bacterial products, ultimately leading to low-grade, chronic, subclinical systemic inflammation. The leaky gut-derived low-grade systemic inflammation may have profound consequences on the gut-liver-brain axis, compromising normal growth, metabolism, and cognitive development. This review examines recent data suggesting that early-life enteric infections that lead to intestinal barrier disruption may shift the intestinal microbiota toward chronic systemic inflammation and subsequent impaired cognitive development.
Collapse
Affiliation(s)
- Reinaldo B Oriá
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA.
| | - Laura E Murray-Kolb
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Rebecca J Scharf
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Laura L Pendergast
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Dennis R Lang
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Glynis L Kolling
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| | - Richard L Guerrant
- R.B. Oriá is with the Laboratory of Tissue Healing, Ontogeny and Nutrition, Institute of Biomedicine and Department of Morphology, Faculty of Medicine, Federal University of Ceará, Ceará, Fortaleza, Brazil. L.E. Murray-Kolb is with The Pennsylvania State University, University Park, Pennsylvania, USA. R.J. Scharf, G. Kolling, and R.L. Guerrant are with the Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA. L.L. Pendergast is with the School Psychology Program, Temple University, Philadelphia, Pennsylvania, USA. D.R. Lang is with the Foundation for the National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Ellwardt E, Walsh JT, Kipnis J, Zipp F. Understanding the Role of T Cells in CNS Homeostasis. Trends Immunol 2016; 37:154-165. [DOI: 10.1016/j.it.2015.12.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/14/2015] [Accepted: 12/14/2015] [Indexed: 01/16/2023]
|
26
|
Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat Rev Neurosci 2015; 16:469-86. [PMID: 26189694 DOI: 10.1038/nrn3978] [Citation(s) in RCA: 337] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increasing evidence points to a central role for immune dysregulation in autism spectrum disorder (ASD). Several ASD risk genes encode components of the immune system and many maternal immune system-related risk factors--including autoimmunity, infection and fetal reactive antibodies--are associated with ASD. In addition, there is evidence of ongoing immune dysregulation in individuals with ASD and in animal models of this disorder. Recently, several molecular signalling pathways--including pathways downstream of cytokines, the receptor MET, major histocompatibility complex class I molecules, microglia and complement factors--have been identified that link immune activation to ASD phenotypes. Together, these findings indicate that the immune system is a point of convergence for multiple ASD-related genetic and environmental risk factors.
Collapse
|
27
|
Louveau A, Harris TH, Kipnis J. Revisiting the Mechanisms of CNS Immune Privilege. Trends Immunol 2015; 36:569-577. [PMID: 26431936 PMCID: PMC4593064 DOI: 10.1016/j.it.2015.08.006] [Citation(s) in RCA: 462] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 08/13/2015] [Accepted: 08/13/2015] [Indexed: 12/25/2022]
Abstract
Whereas the study of the interactions between the immune system and the central nervous system (CNS) has often focused on pathological conditions, the importance of neuroimmune communication in CNS homeostasis and function has become clear over that last two decades. Here we discuss the progression of our understanding of the interaction between the peripheral immune system and the CNS. We examine the notion of immune privilege of the CNS in light of both earlier findings and recent studies revealing a functional meningeal lymphatic system that drains cerebrospinal fluid (CSF) to the deep cervical lymph nodes, and consider the implications of a revised perspective on the immune privilege of the CNS on the etiology and pathology of different neurological disorders.
Collapse
Affiliation(s)
- Antoine Louveau
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
28
|
Loane DJ, Kumar A. Microglia in the TBI brain: The good, the bad, and the dysregulated. Exp Neurol 2015; 275 Pt 3:316-327. [PMID: 26342753 DOI: 10.1016/j.expneurol.2015.08.018] [Citation(s) in RCA: 499] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/05/2015] [Accepted: 08/25/2015] [Indexed: 01/24/2023]
Abstract
As the major cellular component of the innate immune system in the central nervous system (CNS) and the first line of defense whenever injury or disease occurs, microglia play a critical role in neuroinflammation following a traumatic brain injury (TBI). In the injured brain microglia can produce neuroprotective factors, clear cellular debris and orchestrate neurorestorative processes that are beneficial for neurological recovery after TBI. However, microglia can also become dysregulated and can produce high levels of pro-inflammatory and cytotoxic mediators that hinder CNS repair and contribute to neuronal dysfunction and cell death. The dual role of microglial activation in promoting beneficial and detrimental effects on neurons may be accounted for by their polarization state and functional responses after injury. In this review article we discuss emerging research on microglial activation phenotypes in the context of acute brain injury, and the potential role of microglia in phenotype-specific neurorestorative processes such as neurogenesis, angiogenesis, oligodendrogenesis and regeneration. We also describe some of the known molecular mechanisms that regulate phenotype switching, and highlight new therapeutic approaches that alter microglial activation state balance to enhance long-term functional recovery after TBI. An improved understanding of the regulatory mechanisms that control microglial phenotypic shifts may advance our knowledge of post-injury recovery and repair, and provide opportunities for the development of novel therapeutic strategies for TBI.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States; Shock, Trauma, and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Alok Kumar
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States; Shock, Trauma, and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
29
|
Estes ML, McAllister AK. Alterations in immune cells and mediators in the brain: it's not always neuroinflammation! Brain Pathol 2015; 24:623-30. [PMID: 25345893 DOI: 10.1111/bpa.12198] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/02/2023] Open
Abstract
Neuroinflammation was once a clearly defined term denoting pathological immune processes within the central nervous system (CNS). Historically, this term was used to indicate the four hallmarks of peripheral inflammaton that occur following severe CNS injuries, such as stroke, injury or infection. Recently, however, the definition of neuroinflammation has relaxed to the point that it is often now assumed to be present when even only a single classical hallmark of inflammation is measured. As a result, a wide range of disorders, from psychiatric to degenerative diseases, are now assumed to have an integral inflammatory component. Ironically, at the same time, research has revealed unexpected nonclassical immune actions of immune mediators and cells in the CNS in the absence of pathology, increasing the likelihood that homeostatic and adaptive immune processes in the CNS will be mistaken for neuroinflammation. Thus, we suggest reserving the term neuroinflammation for contexts where multiple signs of inflammation are present to avoid erroneously classifying disorders as inflammatory when they may instead be caused by nonimmune etiologies or secondary immune processes that serve adaptive roles.
Collapse
|
30
|
Gadani SP, Walsh JT, Lukens JR, Kipnis J. Dealing with Danger in the CNS: The Response of the Immune System to Injury. Neuron 2015; 87:47-62. [PMID: 26139369 PMCID: PMC4491143 DOI: 10.1016/j.neuron.2015.05.019] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fighting pathogens and maintaining tissue homeostasis are prerequisites for survival. Both of these functions are upheld by the immune system, though the latter is often overlooked in the context of the CNS. The mere presence of immune cells in the CNS was long considered a hallmark of pathology, but this view has been recently challenged by studies demonstrating that immunological signaling can confer pivotal neuroprotective effects on the injured CNS. In this review, we describe the temporal sequence of immunological events that follow CNS injury. Beginning with immediate changes at the injury site, including death of neural cells and release of damage-associated molecular patterns (DAMPs), and progressing through innate and adaptive immune responses, we describe the cascade of inflammatory mediators and the implications of their post-injury effects. We conclude by proposing a revised interpretation of immune privilege in the brain, which takes beneficial neuro-immune communications into account.
Collapse
Affiliation(s)
- Sachin P Gadani
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - James T Walsh
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
31
|
Rathbone ATL, Tharmaradinam S, Jiang S, Rathbone MP, Kumbhare DA. A review of the neuro- and systemic inflammatory responses in post concussion symptoms: Introduction of the "post-inflammatory brain syndrome" PIBS. Brain Behav Immun 2015; 46:1-16. [PMID: 25736063 DOI: 10.1016/j.bbi.2015.02.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 12/22/2022] Open
Abstract
Post-concussion syndrome is an aggregate of symptoms that commonly present together after head injury. These symptoms, depending on definition, include headaches, dizziness, neuropsychiatric symptoms, and cognitive impairment. However, these symptoms are common, occurring frequently in non-head injured controls, leading some to question the existence of post-concussion syndrome as a unique syndrome. Therefore, some have attempted to explain post-concussion symptoms as post-traumatic stress disorder, as they share many similar symptoms and post-traumatic stress disorder does not require head injury. This explanation falls short as patients with post-concussion syndrome do not necessarily experience many key symptoms of post-traumatic stress disorder. Therefore, other explanations must be sought to explain the prevalence of post-concussion like symptoms in non-head injury patients. Many of the situations in which post-concussion syndrome like symptoms may be experienced such as infection and post-surgery are associated with systemic inflammatory responses, and even neuroinflammation. Post-concussion syndrome itself has a significant neuroinflammatory component. In this review we examine the evidence of neuroinflammation in post-concussion syndrome and the potential role systemic inflammation plays in post-concussion syndrome like symptoms. We conclude that given the overlap between these conditions and the role of inflammation in their etiologies, a new term, post-inflammatory brain syndromes (PIBS), is necessary to describe the common outcomes of many different inflammatory insults. The concept of post-concussion syndrome is in its evolution therefore, the new term post-inflammatory brain syndromes provides a better understanding of etiology of its wide-array of symptoms and the wide array of conditions they can be seen in.
Collapse
Affiliation(s)
| | - Surejini Tharmaradinam
- Division of Pediatric Neurology, Department of Pediatrics, McMaster Children's Hospital, Pediatric Neurology, MUMC 3A, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Shucui Jiang
- Division of Neurosurgery, Department of Surgery, and Hamilton Neurorestorative Group, McMaster University, HSC 4E15, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Michel P Rathbone
- Department of Medicine, Division of Neurology, McMaster University - Juravinski Hospital, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada.
| | - Dinesh A Kumbhare
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Toronto, University Health Network - Toronto Rehab - University Centre, 550 University Ave, Toronto, Ontario M5G 2A2, Canada
| |
Collapse
|
32
|
Gadani SP, Walsh JT, Smirnov I, Zheng J, Kipnis J. The glia-derived alarmin IL-33 orchestrates the immune response and promotes recovery following CNS injury. Neuron 2015; 85:703-9. [PMID: 25661185 DOI: 10.1016/j.neuron.2015.01.013] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/21/2014] [Accepted: 01/13/2015] [Indexed: 12/12/2022]
Abstract
Inflammation is a prominent feature of CNS injury that heavily influences neuronal survival, yet the signals that initiate and control it remain poorly understood. Here we identify the nuclear alarmin, interleukin (IL)-33, as an important regulator of the innate immune response after CNS injury. IL-33 is expressed widely throughout the healthy brain and is concentrated in white mater due to predominant expression in post-mitotic oligodendrocytes. IL-33 is released immediately after CNS injury from damaged oligodendrocytes, acting on local astrocytes and microglia to induce chemokines critical for monocyte recruitment. Mice lacking IL-33 have impaired recovery after CNS injury, which is associated with reduced myeloid cell infiltrates and decreased induction of M2 genes at the injury site. These results demonstrate a novel molecular mediator contributing to immune cell recruitment to the injured CNS and may lead to new therapeutic insights in CNS injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sachin P Gadani
- Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | - James T Walsh
- Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jingjing Zheng
- Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Institute of Neurosciences, the Fourth Military Medical University, Xi'an, 710032 China
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
33
|
Brod S, Rattazzi L, Piras G, D'Acquisto F. 'As above, so below' examining the interplay between emotion and the immune system. Immunology 2014; 143:311-8. [PMID: 24943894 DOI: 10.1111/imm.12341] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/12/2014] [Accepted: 06/16/2014] [Indexed: 12/13/2022] Open
Abstract
While the concept of a palpable relationship between our mental and physical well-being is certainly not new, it is only in the light of modern scientific research that we have begun to realize how deeply connected our emotional and immune states may be. We begin this review with a series of studies demonstrating how four fundamental emotional responses: anger, anxiety, mirth and relaxation are able modulate cytokine production and cellular responses to a variety of immune stimuli. These modulations are shown to be either detrimental or beneficial to a patient's health dependent on the context and duration of the emotion. We also discuss the reverse, highlighting research demonstrating how the loss of key immune cells such as T lymphocytes in clinical and animal studies can negatively impact both emotional well-being and cognition. Additionally, to give a more complete picture of the manifold pathways that link emotion and the immune system, we give a brief overview of the influence the digestive system has upon mental and immunological health. Finally, throughout this review we attempt to highlight the therapeutic potential of this burgeoning field of research in both the diagnosis and treatment of immune and disorders. As well as identifying some of the key obstacles the field must address in order to put this potential into practice.
Collapse
Affiliation(s)
- Samuel Brod
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | |
Collapse
|
34
|
Theoharides TC, Athanassiou M, Panagiotidou S, Doyle R. Dysregulated brain immunity and neurotrophin signaling in Rett syndrome and autism spectrum disorders. J Neuroimmunol 2014; 279:33-8. [PMID: 25669997 DOI: 10.1016/j.jneuroim.2014.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 11/18/2014] [Accepted: 12/02/2014] [Indexed: 12/21/2022]
Abstract
Rett syndrome is a neurodevelopmental disorder, which occurs in about 1:15,000 females and presents with neurologic and communication defects. It is transmitted as an X-linked dominant linked to mutations of the methyl-CpG-binding protein (MeCP2), a gene transcription suppressor, but its definitive pathogenesis is unknown thus hindering development of effective treatments. Almost half of children with Rett syndrome also have behavioral symptoms consistent with those of autism spectrum disorders (ASDs). PubMed was searched (2005-2014) using the terms: allergy, atopy, brain, brain-derived neurotrophic factor (BDNF), corticotropin-releasing hormone (CRH), cytokines, gene mutations, inflammation, mast cells (MCs), microglia, mitochondria, neurotensin (NT), neurotrophins, seizures, stress, and treatment. There are a number of intriguing differences and similarities between Rett syndrome and ASDs. Rett syndrome occurs in females, while ASDs more often in males, and the former has neurologic disabilities unlike ASDs. There is evidence of dysregulated immune system early in life in both conditions. Lack of microglial phagocytosis and decreased levels of BDNF appear to distinguish Rett syndrome from ASDs, in which there is instead microglia activation and/or proliferation and possibly defective BDNF signaling. Moreover, brain mast cell (MC) activation and focal inflammation may be more prominent in ASDs than Rett syndrome. The flavonoid luteolin blocks microglia and MC activation, provides BDNF-like activity, reverses Rett phenotype in mouse models, and has a significant benefit in children with ASDs. Appropriate formulations of luteolin or other natural molecules may be useful in the treatment of Rett syndrome.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, USA; Department of Internal Medicine, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, USA; Tufts Medical Center, Boston, MA, USA; Department of Psychiatry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, USA.
| | - Marianna Athanassiou
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, USA
| | - Smaro Panagiotidou
- Molecular Immunopharmacology and Drug Discovery Laboratory, Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, USA
| | - Robert Doyle
- Pediatric Psychopharmacology Unit, Massachusetts General Hospital, Boston MA, USA; Harvard Medical School, Boston MA, USA
| |
Collapse
|
35
|
Clevenger AC, Kilbaugh T, Margulies SS. Carotid artery blood flow decreases after rapid head rotation in piglets. J Neurotrauma 2014; 32:120-6. [PMID: 25133889 DOI: 10.1089/neu.2014.3570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Modification of cerebral perfusion pressure and cerebral blood flow (CBF) are crucial components of the therapies designed to reduce secondary damage after traumatic brain injury (TBI). Previously we documented a robust decrease in CBF after rapid sagittal head rotation in our well-validated animal model of diffuse TBI. Mechanisms responsible for this immediate (<10 min) and sustained (∼24 h) reduction in CBF have not been explored. Because the carotid arteries are a major source of CBF, we hypothesized that blood flow through the carotid arteries (Q) and vessel diameter (D) would decrease after rapid nonimpact head rotation without cervical spine injury. Four-week-old (toddler) female piglets underwent rapid (<20 msec) sagittal head rotation without impact, previously shown to produce diffuse TBI with reductions in CBF. Ultrasonographic images of the bilateral carotid arteries were recorded at baseline (pre-injury), as well as immediately after head rotation and 15, 30, 45, and 60 min after injury. Diameter (D) and waveform velocity (V) were used to calculate blood flow (Q) through the carotid arteries using the equation Q=(0.25)πD(2)V. D, V, and Q were normalized to the pre-injury baseline values to obtain a relative change after injury in right and left carotid arteries. Three-way analysis of variance and post-hoc Tukey-Kramer analyses were used to assess statistical significance of injury, time, and side. The relative change in carotid artery diameter and flow was significantly decreased in injured animals in comparison with uninjured sham controls (p<0.0001 and p=0.0093, respectively) and did not vary with side (p>0.39). The average carotid blood velocity did not differ between sham and injured animals (p=0.91). These data suggest that a reduction in global CBF after rapid sagittal head rotation may be partially mediated by a reduction in carotid artery flow, via vasoconstriction.
Collapse
Affiliation(s)
- Amy C Clevenger
- 1 Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia , Philadelphia, Pennsylvania
| | | | | |
Collapse
|
36
|
Anderson KM, Olson KE, Estes KA, Flanagan K, Gendelman HE, Mosley RL. Dual destructive and protective roles of adaptive immunity in neurodegenerative disorders. Transl Neurodegener 2014; 3:25. [PMID: 25671101 PMCID: PMC4323229 DOI: 10.1186/2047-9158-3-25] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/28/2014] [Indexed: 12/14/2022] Open
Abstract
Inappropriate T cell responses in the central nervous system (CNS) affect the pathogenesis of a broad range of neuroinflammatory and neurodegenerative disorders that include, but are not limited to, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s disease and Parkinson’s disease. On the one hand immune responses can exacerbate neurotoxic responses; while on the other hand, they can lead to neuroprotective outcomes. The temporal and spatial mechanisms by which these immune responses occur and are regulated in the setting of active disease have gained significant recent attention. Spatially, immune responses that affect neurodegeneration may occur within or outside the CNS. Migration of antigen-specific CD4+ T cells from the periphery to the CNS and consequent immune cell interactions with resident glial cells affect neuroinflammation and neuronal survival. The destructive or protective mechanisms of these interactions are linked to the relative numerical and functional dominance of effector or regulatory T cells. Temporally, immune responses at disease onset or during progression may exhibit a differential balance of immune responses in the periphery and within the CNS. Immune responses with predominate T cell subtypes may differentially manifest migratory, regulatory and effector functions when triggered by endogenous misfolded and aggregated proteins and cell-specific stimuli. The final result is altered glial and neuronal behaviors that influence the disease course. Thus, discovery of neurodestructive and neuroprotective immune mechanisms will permit potential new therapeutic pathways that affect neuronal survival and slow disease progression.
Collapse
Affiliation(s)
- Kristi M Anderson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine A Estes
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ken Flanagan
- Prothena Biosciences, South San Francisco, 650 Gateway Boulevard, CA 94080 USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
37
|
Schwarzmaier SM, Plesnila N. Contributions of the immune system to the pathophysiology of traumatic brain injury - evidence by intravital microscopy. Front Cell Neurosci 2014; 8:358. [PMID: 25408636 PMCID: PMC4219391 DOI: 10.3389/fncel.2014.00358] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) results in immediate brain damage that is caused by the mechanical impact and is non-reversible. This initiates a cascade of delayed processes which cause additional—secondary—brain damage. Among these secondary mechanisms, the inflammatory response is believed to play an important role, mediating actions that can have both protective and detrimental effects on the progression of secondary brain damage. Histological data generated extensive information; however, this is only a snapshot of processes that are, in fact, very dynamic. In contrast, in vivo microscopy provides detailed insight into the temporal and spatial patterns of cellular dynamics. In this review, we aim to summarize data which was generated by in vivo microscopy, specifically investigating the immune response following brain trauma, and its potential effects on secondary brain damage.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- Department of Anesthesiology, University of Munich Medical Center Munich, Germany ; Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany ; Munich Cluster of Systems Neurology Munich, Germany
| |
Collapse
|
38
|
Autoantibodies in traumatic brain injury and central nervous system trauma. Neuroscience 2014; 281:16-23. [PMID: 25220901 DOI: 10.1016/j.neuroscience.2014.08.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/14/2014] [Accepted: 08/31/2014] [Indexed: 12/31/2022]
Abstract
Despite the debilitating consequences and the widespread prevalence of brain trauma insults including spinal cord injury (SCI) and traumatic brain injury (TBI), there are currently few effective therapies for most of brain trauma sequelae. As a consequence, there has been a major quest for identifying better diagnostic tools, predictive models, and directed neurotherapeutic strategies in assessing brain trauma. Among the hallmark features of brain injury pathology is the central nervous systems' (CNS) abnormal activation of the immune response post-injury. Of interest, is the occurrence of autoantibodies which are produced following CNS trauma-induced disruption of the blood-brain barrier (BBB) and released into peripheral circulation mounted against self-brain-specific proteins acting as autoantigens. Recently, autoantibodies have been proposed as the new generation class of biomarkers due to their long-term presence in serum compared to their counterpart antigens. The diagnostic and prognostic value of several existing autoantibodies is currently being actively studied. Furthermore, the degree of direct and latent contribution of autoantibodies to CNS insult is still not fully characterized. It is being suggested that there may be an analogy of CNS autoantibodies secretion with the pathophysiology of autoimmune diseases, in which case, understanding and defining the role of autoantibodies in brain injury paradigm (SCI and TBI) may provide a realistic prospect for the development of effective neurotherapy. In this work, we will discuss the accumulating evidence about the appearance of autoantibodies following brain injury insults. Furthermore, we will provide perspectives on their potential roles as pathological components and as candidate markers for detecting and assessing CNS injury.
Collapse
|