101
|
Davoodvandi A, Sahebnasagh R, Mardanshah O, Asemi Z, Nejati M, Shahrzad MK, Mirzaei HR, Mirzaei H. Medicinal Plants As Natural Polarizers of Macrophages: Phytochemicals and Pharmacological Effects. Curr Pharm Des 2019; 25:3225-3238. [DOI: 10.2174/1381612825666190829154934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
Abstract
Macrophages are one of the crucial mediators of the immune response in different physiological and
pathological conditions. These cells have critical functions in the inflammation mechanisms that are involved in
the inhibition or progression of a wide range of diseases including cancer, autoimmune diseases, etc. It has been
shown that macrophages are generally divided into two subtypes, M1 and M2, which are distinguished on the
basis of their different gene expression patterns and phenotype. M1 macrophages are known as pro-inflammatory
cells and are involved in inflammatory mechanisms, whereas M2 macrophages are known as anti-inflammatory
cells that are involved in the inhibition of the inflammatory pathways. M2 macrophages help in tissue healing via
producing anti-inflammatory cytokines. Increasing evidence indicated that the appearance of different macrophage
subtypes is associated with the fate of diseases (progression versus suppression). Hence, polarization of
macrophages can be introduced as an important venue in finding, designing and developing novel therapeutic
approaches. Albeit, there are different pharmacological agents that are used for the treatment of various disorders,
it has been shown that several natural compounds have the potential to regulate M1 to M2 macrophage polarization
and vice versa. Herein, for the first time, we summarized new insights into the pharmacological effects of
natural compounds on macrophage polarization.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Mardanshah
- Department of Laboratory Sciences, Sirjan Faculty of Medical Sciences, Sirjan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Majid Nejati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad K. Shahrzad
- Department of Internal Medicine and Endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid R. Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
102
|
Aurelian L, Balan I. GABA AR α2-activated neuroimmune signal controls binge drinking and impulsivity through regulation of the CCL2/CX3CL1 balance. Psychopharmacology (Berl) 2019; 236:3023-3043. [PMID: 31030249 DOI: 10.1007/s00213-019-05220-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Toll-like receptors (TLRs) are a family of innate immune system receptors that respond to pathogen-derived and tissue damage-related ligands and are increasingly recognized for their impact on homeostasis and its dysregulation in the nervous system. TLR signaling participates in brain injury and addiction, but its role in the alcohol-seeking behavior, which initiates alcohol drinking, is still poorly understood. In this review, we discuss our findings designed to elucidate the potential contribution of the activated TLR4 signal located in neurons, on impulsivity and the predisposition to initiate alcohol drinking (binge drinking). RESULTS Our findings indicate that the TLR4 signal is innately activated in neurons from alcohol-preferring subjects, identifying a genetic contribution to the regulation of impulsivity and the alcohol-seeking propensity. Signal activation is through the non-canonical, previously unknown, binding of TLR4 to the α2 subunit of the γ-aminobutyric 2 acid A receptor (GABAAR α2). Activation is sustained by the stress hormone corticotrophin-releasing factor (CRF) and additional still poorly recognized ligand/scaffold proteins. Focus is on the effect of TLR4 signal activation on the balance between pro- and anti-inflammatory chemokines [chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-X3-C motif) ligand 1 (CX3CL1)] and its effect on binge drinking. CONCLUSION The results are discussed within the context of current findings on the distinct activation and functions of TLR signals located in neurons, as opposed to immune cells. They indicate that the balance between pro- and anti-inflammatory TLR4 signaling plays a major role in binge drinking. These findings have major impact on future basic and translational research, including the development of potential therapeutic and preventative strategies.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Stanford University School of Medicine OFDD, Stanford, CA, 94305, USA.
| | - Irina Balan
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
103
|
Kahanovitch U, Patterson KC, Hernandez R, Olsen ML. Glial Dysfunction in MeCP2 Deficiency Models: Implications for Rett Syndrome. Int J Mol Sci 2019; 20:ijms20153813. [PMID: 31387202 PMCID: PMC6696322 DOI: 10.3390/ijms20153813] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome (RTT) is a rare, X-linked neurodevelopmental disorder typically affecting females, resulting in a range of symptoms including autistic features, intellectual impairment, motor deterioration, and autonomic abnormalities. RTT is primarily caused by the genetic mutation of the Mecp2 gene. Initially considered a neuronal disease, recent research shows that glial dysfunction contributes to the RTT disease phenotype. In the following manuscript, we review the evidence regarding glial dysfunction and its effects on disease etiology.
Collapse
Affiliation(s)
- Uri Kahanovitch
- School of Neuroscience, Virginia Polytechnic and State University, Life Sciences I Building Room 212, 970 Washington St. SW, Blacksburg, VA 24061, USA
| | - Kelsey C Patterson
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd., Birmingham, AL 35294, USA
| | - Raymundo Hernandez
- School of Neuroscience, Virginia Polytechnic and State University, Life Sciences I Building Room 212, 970 Washington St. SW, Blacksburg, VA 24061, USA
- Graduate Program in Translational Biology Medicine and Health, Virginia Tech, Roanoke, VL 24014, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Polytechnic and State University, Life Sciences I Building Room 212, 970 Washington St. SW, Blacksburg, VA 24061, USA.
| |
Collapse
|
104
|
Wasko NJ, Kulak MH, Paul D, Nicaise AM, Yeung ST, Nichols FC, Khanna KM, Crocker S, Pachter JS, Clark RB. Systemic TLR2 tolerance enhances central nervous system remyelination. J Neuroinflammation 2019; 16:158. [PMID: 31351476 PMCID: PMC6660683 DOI: 10.1186/s12974-019-1540-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022] Open
Abstract
Background Multiple sclerosis (MS) is a central nervous system (CNS) autoimmune disease characterized by both inflammatory demyelination and impaired remyelination. Studies indicate that Toll-like receptor 2 (TLR2) signaling contributes to both the inflammatory component and the defective remyelination in MS. While most MS therapeutics target adaptive immunity, we recently reported that reducing TLR2 signaling in innate immune cells by inducing TLR2 tolerance attenuates adoptively transferred experimental autoimmune encephalomyelitis. Given that previous reports suggest TLR2 signaling also inhibits myelin repair, the objective of this study was to assess how reducing TLR2 signaling through TLR2 tolerance induction affects CNS myelin repair. Methods Chow containing 0.2% cuprizone was fed to male and female wild-type (WT) C57BL/6 mice or TLR2-deficient (TLR2−/−) mice for 5 weeks to induce demyelination. During a 2-week remyelination period following discontinuation of cuprizone, WT mice received either low dose TLR2 ligands to induce systemic TLR2 tolerance or vehicle control (VC). Remyelination was evaluated via electron microscopy and immunohistochemical analysis of microglia and oligodendrocytes in the corpus callosum. Statistical tests included 2-way ANOVA and Mann-Whitney U analyses. Results Inducing TLR2 tolerance in WT mice during remyelination significantly enhanced myelin recovery, restoring unmyelinated axon frequency and myelin thickness to baseline levels compared to VC-treated mice. Mechanistically, enhanced remyelination in TLR2 tolerized mice was associated with a shift in corpus callosum microglia from a pro-inflammatory iNOS+ phenotype to a non-inflammatory/pro-repair Arg1+ phenotype. This result was confirmed in vitro by inducing TLR2 tolerance in WT microglia cultures. TLR2−/− mice, without TLR2 tolerance induction, also significantly enhanced myelin recovery compared to WT mice, adding confirmation that reduced TLR2 signaling is associated with enhanced remyelination. Discussion Our results suggest that reducing TLR2 signaling in vivo by inducing TLR2 tolerance significantly enhances myelin repair. Furthermore, the enhanced remyelination resulting from TLR2 tolerance induction is associated with a shift in corpus callosum microglia from a pro-inflammatory iNOS+ phenotype to a non-inflammatory/pro-repair Arg1+ phenotype. While deletion of TLR2 would be an impractical approach in vivo, reducing innate immune signaling through TLR2 tolerance induction may represent a novel, two-pronged approach for treating both inflammatory and myelin repair components of MS. Electronic supplementary material The online version of this article (10.1186/s12974-019-1540-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicholas J Wasko
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
| | | | - Debayon Paul
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
| | | | - Stephen T Yeung
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA.,Present Address: Department of Microbiology, Perlmutter Cancer Center at NYU Langone School of Medicine, New York, NY, 10016, USA
| | - Frank C Nichols
- Department of Periodontology, UConn Health, Farmington, CT, 06030, USA
| | - Kamal M Khanna
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA.,Present Address: Department of Microbiology, Perlmutter Cancer Center at NYU Langone School of Medicine, New York, NY, 10016, USA
| | - Stephen Crocker
- Department of Neuroscience, UConn Health, Farmington, CT, 06030, USA
| | - Joel S Pachter
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
| | - Robert B Clark
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA. .,Department of Medicine, UConn Health, Farmington, CT, 06030, USA.
| |
Collapse
|
105
|
The Relationship Between Neuroimmunity and Bipolar Disorder: Mechanism and Translational Application. Neurosci Bull 2019; 35:595-607. [PMID: 31214924 DOI: 10.1007/s12264-019-00403-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022] Open
Abstract
Neuroimmune system may be involved in the pathological process of bipolar disorder (BD), but the essential association is not fully understood. Accumulating evidence has shown that BD involves the activation of immune cells and the release of inflammatory substances in the central nerve system (CNS). Meanwhile, neuroimmune responses also interact with other hypothesis of the etiology of BD that are widely recognized, such as neurotransmitter systems, neuroendocrine systems, neurotrophic factors, and oxidative stress. Simultaneously, related genes and immune changes in peripheral blood vary with it. Overall, neuroimmunity may play an important role in the pathogenesis of BD, and the inflammatory cytokines, especially interleukin-6 and tumor necrosis factor-alpha, have potential value for the clinical diagnosis and prognosis of BD, as well as predicting the therapeutic effects of drugs. Large-scale studies are needed to extend the evidence on neuroimmunity in BD, and to examine its clinical value for applications such as early prediction and treatment.
Collapse
|
106
|
Ascoli BM, Parisi MM, Bristot G, Antqueviezc B, Géa LP, Colombo R, Kapczinski F, Guma FTCR, Brietzke E, Barbé-Tuana FM, Rosa AR. Attenuated inflammatory response of monocyte-derived macrophage from patients with BD: a preliminary report. Int J Bipolar Disord 2019; 7:13. [PMID: 31152269 PMCID: PMC6544740 DOI: 10.1186/s40345-019-0148-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Innate immune system dysfunction has been recognized as an important element in the pathophysiology of bipolar disorder (BD). We aimed to investigate whether there are differences in the response of macrophages derived from patients in the early stages and late stages of BD and healthy subjects. METHODS Human monocytes purified from peripheral blood mononuclear cells (PBMCs) of patients with BD type I (n = 18)-further classified into early- and late stage BD patients according to their functioning- and from healthy individuals (n = 10) were differentiated into macrophages in vitro. Monocyte-derived macrophages (M) were exposed to IFNγ plus LPS-M(IFNγ + LPS)- or IL-4-M(IL-4)-to induce their polarization into the classical (also called M1) or alternative (also called M2) activation phenotypes, respectively; or either Mψ were not exposed to any stimuli characterizing the resting state (denominated M0). In vitro secretion of cytokines, such as IL-1β, IL-6, IL-10, and TNF-α, was used as an index of macrophage activity. RESULTS M(IFNγ + LPS) from late-stage BD patients produced less amount of IL-1β, IL-6, and IL-10 when compared to early-stage BD patients and healthy controls. Following alternative activation, M(IL-4) derived from late-stage patients secreted less IL-6 compared to the other groups. TNFα was less secreted by all macrophage phenotypes derived from late-stage patients when compared to healthy controls only (p < 0.005). Mψ from late-stage patients exhibited lower production of IL-1β and IL-10 compared to macrophages from healthy subjects and early-stage patients respectively. Interestingly, cytokines secretion from M(IFNγ + LPS), M(IL-4) and Mψ were similar between early-stage patients and healthy controls. CONCLUSION Our results suggest a progressive dysfunction in the response of peripheral innate immune cells of BD patients in the late stages of the illness. This failure in the regulation of the immune system function may be implicated in the multisystemic progression of BD.
Collapse
Affiliation(s)
- Bruna M Ascoli
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil.,Postgraduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2400, Porto Alegre, RS, Brazil
| | - Mariana M Parisi
- Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry, UFRGS, Rua Ramiro Barcelos, 2600, Porto Alegre, Brazil.,Postgraduate Program in Biological Sciences: Biochemistry, UFRGS, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil
| | - Giovana Bristot
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil.,Postgraduate Program in Biological Sciences: Biochemistry, UFRGS, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil
| | - Bárbara Antqueviezc
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil
| | - Luiza P Géa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil.,Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Rua Sarmento Leite 500, Porto Alegre, RS, Brazil
| | - Rafael Colombo
- Laboratory of Pharmacology and Physiology, Universidade de Caxias do Sul (UCS), Rua Francisco Getúlio Vargas, 1130, Caxias Do Sul, RS, Brazil
| | - Flávio Kapczinski
- Postgraduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2400, Porto Alegre, RS, Brazil.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, 1280 Main Street West, Hamilton, ON, Canada.,St. Joseph's Healthcare Hamilton, 100 West 5th Street, Hamilton, ON, Canada
| | - Fátima Theresinha Costa Rodrigues Guma
- Postgraduate Program in Biological Sciences: Biochemistry, UFRGS, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil.,Laboratory of Biochemistry and Cellular Biology of Lipids, Department of Biochemistry, UFRGS, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, Brazil
| | - Elisa Brietzke
- Mood Disorders Molecular and Behavioral Neurosciences Research Group, Department of Psychiatry, Universidade Federal de São Paulo (USP), Rua Sena Madureira, 1500, São Paulo, SP, Brazil
| | - Florencia M Barbé-Tuana
- Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry, UFRGS, Rua Ramiro Barcelos, 2600, Porto Alegre, Brazil.,Postgraduate Program in Cellular and Molecular Biology, School of Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, RS, Brazil
| | - Adriane R Rosa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, Brazil. .,Postgraduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2400, Porto Alegre, RS, Brazil. .,Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Rua Sarmento Leite 500, Porto Alegre, RS, Brazil.
| |
Collapse
|
107
|
Tellez-Merlo G, Morales-Medina JC, Camacho-Ábrego I, Juárez-Díaz I, Aguilar-Alonso P, de la Cruz F, Iannitti T, Flores G. Prenatal immune challenge induces behavioral deficits, neuronal remodeling, and increases brain nitric oxide and zinc levels in the male rat offspring. Neuroscience 2019; 406:594-605. [DOI: 10.1016/j.neuroscience.2019.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 12/11/2018] [Accepted: 02/12/2019] [Indexed: 12/20/2022]
|
108
|
Lee Y, Lee S, Chang SC, Lee J. Significant roles of neuroinflammation in Parkinson's disease: therapeutic targets for PD prevention. Arch Pharm Res 2019; 42:416-425. [PMID: 30830660 DOI: 10.1007/s12272-019-01133-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023]
Abstract
Glial cells outnumber neurons in the brain and play important roles in the neuroinflammation that accompanies brain damage in neurodegenerative diseases. In Parkinson's disease (PD), dopaminergic neuronal loss is accompanied by inflammatory changes in microglia, astrocytes, innate immune cells, and infiltrating peripheral immune cells. Neuroinflammation is probably a fundamental immune response to protect neurons from harm and compensate for neuronal damage, but at the same time, its neurotoxic effects exacerbate neuron damage. Furthermore, neuroinflammatory response is regulated by immune cells, such as microglia, astrocytes, and peripheral immune cells, and by cytokines and chemokines. Accordingly, it is crucial that we understand how such immune cells in the brain regulate neuroinflammatory responses in PD pathology. This review describes the roles played by glia-mediated neuroinflammation in PD, both good and bad, and the therapeutic strategies used to treat PD.
Collapse
Affiliation(s)
- Yujeong Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seulah Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seung-Cheol Chang
- Institute of BioPhysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
109
|
Meng X, Duan C, Pang H, Chen Q, Han B, Zha C, Dinislam M, Wu P, Li Z, Zhao S, Wang R, Lin L, Jiang C, Cai J. DNA damage repair alterations modulate M2 polarization of microglia to remodel the tumor microenvironment via the p53-mediated MDK expression in glioma. EBioMedicine 2019; 41:185-199. [PMID: 30773478 PMCID: PMC6442002 DOI: 10.1016/j.ebiom.2019.01.067] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/25/2022] Open
Abstract
Background DNA damage repair (DDR) alterations are important events in cancer initiation, progression, and therapeutic resistance. However, the involvement of DDR alterations in glioma malignancy needs further investigation. This study aims to characterize the clinical and molecular features of gliomas with DDR alterations and elucidate the biological process of DDR alterations that regulate the cross talk between gliomas and the tumor microenvironment. Methods Integrated transcriptomic and genomic analyses were undertaken to conduct a comprehensive investigation of the role of DDR alterations in glioma. The prognostic DDR-related cytokines were identified from multiple datasets. In vivo and in vitro experiments validated the role of p53, the key molecule of DDR, regulating M2 polarization of microglia in glioma. Findings DDR alterations are associated with clinical and molecular characteristics of glioma. Gliomas with DDR alterations exhibit distinct immune phenotypes, and immune cell types and cytokine processes. DDR-related cytokines have an unfavorable prognostic implication for GBM patients and are synergistic with DDR alterations. Overexpression of MDK mediated by p53, the key transcriptional factor in DDR pathways, remodels the GBM immunosuppressive microenvironment by promoting M2 polarization of microglia, suggesting a potential role of DDR in regulating the glioma microenvironment. Interpretation Our work suggests that DDR alterations significantly contribute to remodeling the glioma microenvironment via regulating the immune response and cytokine pathways. Fund This study was supported by: 1. The National Key Research and Development Plan (No. 2016YFC0902500); 2. National Natural Science Foundation of China (No. 81702972, No. 81874204, No. 81572701, No. 81772666); 3. China Postdoctoral Science Foundation (2018M640305); 4. Special Fund Project of Translational Medicine in the Chinese-Russian Medical Research Center (No. CR201812); 5. The Research Project of the Chinese Society of Neuro-oncology, CACA (CSNO-2016-MSD12); 6. The Research Project of the Health and Family Planning Commission of Heilongjiang Province (2017–201); and 7. Harbin Medical University Innovation Fund (2017LCZX37, 2017RWZX03). Gliomas with DNA damage repair alterations had distinct genomic variation spectrum. DDR alterations exhibit distinct immune phenotypes, cytokine processes and immune cell types in glioma. DDR-related cytokines in GME have an unfavorable prognostic implication for GBM patients. P53-mediated midkine expression derived from glioma cells promotes M2 polarization of microglia.
Collapse
Affiliation(s)
- Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Chunbin Duan
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Hengyuan Pang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Qun Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Bo Han
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Caijun Zha
- Department of Laboratory Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Magafurov Dinislam
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neurosurgical department, Bashkir State Medical University, Ufa 450008, Russia
| | - Pengfei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Ziwei Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Shihong Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ruijia Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Lin Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China.
| |
Collapse
|
110
|
Fries GR, Walss-Bass C, Bauer ME, Teixeira AL. Revisiting inflammation in bipolar disorder. Pharmacol Biochem Behav 2019; 177:12-19. [DOI: 10.1016/j.pbb.2018.12.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 12/05/2018] [Accepted: 12/20/2018] [Indexed: 01/11/2023]
|
111
|
Jin X, Zhang X, Li J, Yu W, Chen F. Activation of chicken macrophages during in vitro stimulation and expression of immune genes. Am J Vet Res 2019; 79:1306-1312. [PMID: 30457902 DOI: 10.2460/ajvr.79.12.1306] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To characterize activation and expression of immune genes of chicken macrophages after in vitro stimulation with lipopolysaccharide (LPS) and mouse erythrocytes. ANIMALS Five 15-day-old chickens and 2 BALB/c mice. PROCEDURES Macrophages were extracted from chicken bone marrow or peripheral blood and then stimulated with cytokines secreted from cell lines L929 and HD11. Stimulated chicken macrophages were further cocultured with LPS or mouse erythrocytes, and gene transcription of some distinctive cytokines was detected by use of a real-time PCR assay. RESULTS Morphological features and phagocytic function of macrophages were characterized. Activated macrophages had an elongated shape with a large cell nucleus, and they had phagocytic function. Distinctive genes encoding the surface marker gene CD11b were identified; high quantities of CD11b were transcribed. Relative transcription of chicken genes BF and BL in mature cells cocultured with both stimuli was lower than for control cells. However, the quantity of genes encoding M1- or M2-distinctive cytokines (interleukin [IL]-1β, IL-10, IL-12, inducible nitric oxide synthase, tumor necrosis factor-α, and transforming growth factor-β) that were transcribed differed significantly between stimulation with LPS and mouse erythrocytes. CONCLUSIONS AND CLINICAL RELEVANCE Chicken macrophages were differentially stimulated by LPS and mouse erythrocytes, which suggested that in vitro stimulation can distinctly influence the transcription and expression of immune genes of chicken macrophages.
Collapse
|
112
|
Scheu S, Ali S, Mann-Nüttel R, Richter L, Arolt V, Dannlowski U, Kuhlmann T, Klotz L, Alferink J. Interferon β-Mediated Protective Functions of Microglia in Central Nervous System Autoimmunity. Int J Mol Sci 2019; 20:E190. [PMID: 30621022 PMCID: PMC6337097 DOI: 10.3390/ijms20010190] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/23/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination and axonal damage. It often affects young adults and can lead to neurological disability. Interferon β (IFNβ) preparations represent widely used treatment regimens for patients with relapsing-remitting MS (RRMS) with therapeutic efficacy in reducing disease progression and frequency of acute exacerbations. In mice, IFNβ therapy has been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS while genetic deletion of IFNβ or its receptor augments clinical severity of disease. However, the complex mechanism of action of IFNβ in CNS autoimmunity has not been fully elucidated. Here, we review our current understanding of the origin, phenotype, and function of microglia and CNS immigrating macrophages in the pathogenesis of MS and EAE. In addition, we highlight the emerging roles of microglia as IFNβ-producing cells and vice versa the impact of IFNβ on microglia in CNS autoimmunity. We finally discuss recent progress in unraveling the underlying molecular mechanisms of IFNβ-mediated effects in EAE.
Collapse
Affiliation(s)
- Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Shafaqat Ali
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
- Cells in Motion, Cluster of Excellence, University of Münster, 48149 Münster, Germany.
| | - Ritu Mann-Nüttel
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Lisa Richter
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Volker Arolt
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
| | - Udo Dannlowski
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany.
| | - Luisa Klotz
- Department of Neurology, University of Münster, 48149 Münster, Germany.
| | - Judith Alferink
- Department of Psychiatry and Psychotherapy, University of Münster, 48149 Münster, Germany.
- Cells in Motion, Cluster of Excellence, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
113
|
Mohammadi A, Blesso CN, Barreto GE, Banach M, Majeed M, Sahebkar A. Macrophage plasticity, polarization and function in response to curcumin, a diet-derived polyphenol, as an immunomodulatory agent. J Nutr Biochem 2018; 66:1-16. [PMID: 30660832 DOI: 10.1016/j.jnutbio.2018.12.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/04/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022]
Abstract
Monocytes and macrophages are important cells of the innate immune system that have diverse functions, including defense against invading pathogens, removal of dead cells by phagocytosis, antigen presentation in the context of MHC class I and class II molecules, and production of various pro-inflammatory cytokines and chemokines such as IL-1β, IL-6, TNF-α and MCP-1. In addition, pro-inflammatory (M1) and anti-inflammatory (M2) macrophages clearly play important roles in the progression of several inflammatory diseases. Therefore, therapies that target macrophage polarization and function by either blocking their trafficking to sites of inflammation, or skewing M1 to M2 phenotype polarization may hold clinical promise in several inflammatory diseases. Dietary-derived polyphenols have potent natural anti-oxidative properties. Within this group of polyphenols, curcumin has been shown to suppress macrophage inflammatory responses. Curcumin significantly reduces co-stimulatory molecules and also inhibits MAPK activation and the translocation of NF-κB p65. Curcumin can also polarize/repolarize macrophages toward the M2 phenotype. Curcumin-treated macrophages have been shown to be highly efficient at antigen capture and endocytosis via the mannose receptor. These novel findings provide new perspectives for the understanding of the immunopharmacological role of curcumin, as well as its therapeutic potential for impacting macrophage polarization and function in the context of inflammation-related disease. However, the precise effects of curcumin on the migration, differentiation, polarization and immunostimulatory functions of macrophages remain unknown. Therefore, in this review, we summarized whether curcumin can influence macrophage polarization, surface molecule expression, cytokine and chemokine production and their underlying pathways in the prevention of inflammatory diseases.
Collapse
Affiliation(s)
- Asadollah Mohammadi
- Cellular & Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, University of Western Australia, Perth, Australia.
| |
Collapse
|
114
|
Kelly ÁM. Exercise-Induced Modulation of Neuroinflammation in Models of Alzheimer's Disease. Brain Plast 2018; 4:81-94. [PMID: 30564548 PMCID: PMC6296260 DOI: 10.3233/bpl-180074] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2018] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD), a progressive, neurodegenerative condition characterised by accumulation of toxic βeta-amyloid (Aβ) plaques, is one of the leading causes of dementia globally. The cognitive impairment that is a hallmark of AD may be caused by inflammation in the brain triggered and maintained by the presence of Aβ protein, ultimately leading to neuronal dysfunction and loss. Since there is a significant inflammatory component to AD, it is postulated that anti-inflammatory strategies may be of prophylactic or therapeutic benefit in AD. One such strategy is that of regular physical activity, which has been shown in epidemiological studies to be protective against various forms of dementia including AD. Exercise induces an anti-inflammatory environment in peripheral organs and also increases expression of anti-inflammatory molecules within the brain. Here we review the evidence, mainly from animal models of AD, supporting the hypothesis that exercise can reduce or slow the cellular and cognitive impairments associated with AD by modulating neuroinflammation.
Collapse
Affiliation(s)
- Áine M. Kelly
- Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
115
|
Zhou J, Mao L, Xu P, Wang Y. Effects of (-)-Epigallocatechin Gallate (EGCG) on Energy Expenditure and Microglia-Mediated Hypothalamic Inflammation in Mice Fed a High-Fat Diet. Nutrients 2018; 10:nu10111681. [PMID: 30400620 PMCID: PMC6266769 DOI: 10.3390/nu10111681] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 12/28/2022] Open
Abstract
Obesity is an escalating global epidemic caused by an imbalance between energy intake and expenditure. (−)-Epigallocatechin-3-gallate (EGCG), the major polyphenol in green tea, has been reported to be conducive to preventing obesity and alleviating obesity-related chronic diseases. However, the role of EGCG in energy metabolism disorders and central nervous system dysfunction induced by a high-fat diet (HFD) remains to be elucidated. The aim of this study was to evaluate the effects of EGCG on brown adipose tissue (BAT) thermogenesis and neuroinflammation in HFD-induced obese C57BL/6J mice. Mice were randomly divided into four groups with different diets: normal chow diet (NCD), normal chow diet supplemented with 1% EGCG (NCD + EGCG), high-fat diet (HFD), and high-fat diet supplemented with 1% EGCG (HFD + EGCG). Investigations based on a four-week experiment were carried out including the BAT activity, energy consumption, mRNA expression of major inflammatory cytokines in the hypothalamus, nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) phosphorylation, and immunofluorescence staining of microglial marker Iba1 in hypothalamic arcuate nucleus (ARC). Experimental results demonstrated that dietary supplementation of EGCG significantly inhibited HFD-induced obesity by enhancing BAT thermogenesis, and attenuated the hypothalamic inflammation and microglia overactivation by regulating the NF-κB and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Jihong Zhou
- Tea Research Institute, Zhejiang University, Hangzhou 310058, China.
| | - Limin Mao
- Tea Research Institute, Zhejiang University, Hangzhou 310058, China.
| | - Ping Xu
- Tea Research Institute, Zhejiang University, Hangzhou 310058, China.
| | - Yuefei Wang
- Tea Research Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
116
|
Aratake T, Higashi Y, Ueba Y, Hamada T, Shimizu T, Shimizu S, Yawata T, Ueba T, Saito M. The inhibitory role of intracellular free zinc in the regulation of Arg-1 expression in interleukin-4-induced activation of M2 microglia. Metallomics 2018; 10:1501-1509. [PMID: 30206632 DOI: 10.1039/c8mt00248g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Microglia, the resident immune cells of the central nervous system, can display a pro-inflammatory M1 phenotype or an anti-inflammatory M2 phenotype. Arginase (Arg)-1 expressed in interleukin-4 (IL-4)-induced M2 microglia reduces nitric oxide (NO) production by competing with inducible NO synthase for l-arginine, which contributes to the attenuation of brain inflammation. Although previous studies have indicated that brain zinc promotes M1 activation, the effect of zinc on M2 microglial activation remains to be determined. In the present study, murine primary microglia treated with 10 ng mL-1 IL-4 exhibited increased Arg-1 mRNA expression and levels of intracellular free zinc. Chelation of this increased intracellular free zinc by the cell permeable zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) aggravated the IL-4-induced mRNA expression and enzymatic activity of Arg-1. However, the cell impermeable zinc chelator CaEDTA had no effect on Arg-1 expression or cytosolic levels of free zinc in IL-4-induced M2-polarized microglia. Furthermore, treatment with IL-4 resulted in upregulation of phagocytic activity in microglia, while administration of TPEN abolished IL-4-induced phagocytic activity. Moreover, this effect was reversed vial-arginine supplementation. These findings suggest that IL-4 induces an increase in intracellular free zinc in microglia, which may act as a negative regulator of IL-4-induced Arg-1 expression, and that such negative regulation is essential for microglial phagocytic activity.
Collapse
Affiliation(s)
- Takaaki Aratake
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Yusuke Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Tomoya Hamada
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Toshio Yawata
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Tetsuya Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| |
Collapse
|
117
|
Dello Russo C, Cappoli N, Coletta I, Mezzogori D, Paciello F, Pozzoli G, Navarra P, Battaglia A. The human microglial HMC3 cell line: where do we stand? A systematic literature review. J Neuroinflammation 2018; 15:259. [PMID: 30200996 PMCID: PMC6131758 DOI: 10.1186/s12974-018-1288-0] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/21/2018] [Indexed: 02/08/2023] Open
Abstract
Microglia, unique myeloid cells residing in the brain parenchyma, represent the first line of immune defense within the central nervous system. In addition to their immune functions, microglial cells play an important role in other cerebral processes, including the regulation of synaptic architecture and neurogenesis. Chronic microglial activation is regarded as detrimental, and it is considered a pathogenic mechanism common to several neurological disorders. Microglial activation and function have been extensively studied in rodent experimental models, whereas the characterization of human cells has been limited due to the restricted availability of primary sources of human microglia. To overcome this problem, human immortalized microglial cell lines have been developed. The human microglial clone 3 cell line, HMC3, was established in 1995, through SV40-dependent immortalization of human embryonic microglial cells. It has been recently authenticated by the American Type Culture Collection (ATCC®) and distributed under the name of HMC3 (ATCC®CRL-3304). The HMC3 cells have been used in six research studies, two of which also indicated by ATCC® as reference articles. However, a more accurate literature revision suggests that clone 3 was initially distributed under the name of CHME3. In this regard, several studies have been published, thus contributing to a more extensive characterization of this cell line. Remarkably, the same cell line has been used in different laboratories with other denominations, i.e., CHME-5 cells and C13-NJ cells. In view of the fact that "being now authenticated by ATCC®" may imply a wider distribution of the cells, we aimed at reviewing data obtained with the human microglia cell line clone 3, making the readers aware of this complicated nomenclature. In addition, we also included original data, generated in our laboratory with the HMC3 (ATCC®CRL-3304) cells, providing information on the current state of the culture together with supplementary details on the culturing procedures to obtain and maintain viable cells.
Collapse
Affiliation(s)
- Cinzia Dello Russo
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy. .,Pharmacology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Natalia Cappoli
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy
| | - Isabella Coletta
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A., Rome, Italy
| | - Daniele Mezzogori
- Institute of Human Physiology, Università Cattolica del S. Cuore, Rome, Italy
| | - Fabiola Paciello
- Institute of Otolaryngology, Università Cattolica del S. Cuore, Rome, Italy
| | - Giacomo Pozzoli
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy.,Pharmacology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Pierluigi Navarra
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy.,Pharmacology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandra Battaglia
- Immunology Laboratory, Department of Oncological Gynecology, Università Cattolica del S. Cuore, Rome, Italy
| |
Collapse
|
118
|
Alvariño R, Alonso E, Lacret R, Oves-Costales D, Genilloud O, Reyes F, Alfonso A, Botana LM. Streptocyclinones A and B ameliorate Alzheimer's disease pathological processes in vitro. Neuropharmacology 2018; 141:283-295. [PMID: 30205103 DOI: 10.1016/j.neuropharm.2018.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/14/2018] [Accepted: 09/07/2018] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is a pathology characterized by the abnormal accumulation of amyloid-beta (Aβ) and hyperphosphorylated tau. Oxidative stress and neuroinflammation are also strongly related to this disease. The ability of two new glycosylated angucyclinones, streptocyclinones A and B (1 and 2), isolated from Streptomyces sp to improve AD hallmarks was evaluated. Compounds were able to protect SH-SY5Y neuroblastoma cells from H2O2-induced oxidative injury by activating the nuclear factor E2-related factor (Nrf2). Their capacity to modulate neuroinflammation was tested in lipopolysaccharide-activated BV2 microglial cells. Compounds reduced the release of pro-inflammatory factors, inhibited the activation of NFκB and mitogen activated kinases (MAPK), and induced the translocation of Nrf2 to the nucleus of microglial cells. A trans-well co-culture was established to determine the effect of microglia treated with streptocyclinones on the survival of SH-SY5Y cells. The cell viability of neuroblastoma cells increased when the compounds were added to BV2 cells. SH-SY5Y-TMHT441 cells were used to determine the effect of compounds on tau phosphorylation. Both compounds reduced tau hyperphophorylation by targeting MAPK kinases. Moreover, streptocyclinone B (2) was able to inhibit the activity of β-secretase 1 and decrease the release of reactive oxygen species in BV2 cells stimulated with Aβ. With the same co-culture trans-well system, the treatment of Aβ-stimulated microglia with compound 2 augmented the viability of SH-SY5Y-TMHT441 cells. The results presented in this work provide evidences of the multitarget activities displayed by these new Streptomyces compounds, making them good candidates for further studies in the treatment of AD.
Collapse
Affiliation(s)
- Rebeca Alvariño
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo 27003, Spain
| | - Eva Alonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo 27003, Spain.
| | - Rodney Lacret
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento 34, 18016, Granada, Spain
| | - Daniel Oves-Costales
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento 34, 18016, Granada, Spain
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento 34, 18016, Granada, Spain
| | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento 34, 18016, Granada, Spain
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo 27003, Spain
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo 27003, Spain.
| |
Collapse
|
119
|
Lurie DI. An Integrative Approach to Neuroinflammation in Psychiatric disorders and Neuropathic Pain. J Exp Neurosci 2018; 12:1179069518793639. [PMID: 30127639 PMCID: PMC6090491 DOI: 10.1177/1179069518793639] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is a complex process involving both the peripheral circulation
and the Central Nervous System (CNS) and is considered to underlie many CNS
disorders including depression, anxiety, schizophrenia, and pain. Stressors
including early-life adversity, psychosocial stress, and infection appear to
prime microglia toward a pro-inflammatory phenotype. Subsequent inflammatory
challenges then drive an exaggerated neuroinflammatory response involving the
upregulation of pro-inflammatory mediators that is associated with CNS
dysfunction. Several pharmacologic inhibitors of pro-inflammatory cytokines
including TNF-α and IL-1β show good clinical efficacy in terms of ameliorating
neuroinflammatory processes. Mind/body and plant-based interventions such as
yoga, breathing exercises, meditation, and herbs/spices have also been
demonstrated to reduce pro-inflammatory cytokines and have a positive impact on
depression, anxiety, cognition, and pain. As the intricate connections between
the immune system and the nervous system continue to be elucidated, successful
therapies for reducing neuroinflammation will likely involve an integrated
approach combining drug therapy with nonpharmacologic interventions.
Collapse
Affiliation(s)
- Diana I Lurie
- Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, College of Health Professions & Biomedical Sciences, The University of Montana, Missoula, MT, USA
| |
Collapse
|
120
|
Garrison AM, Parrott JM, Tuñon A, Delgado J, Redus L, O'Connor JC. Kynurenine pathway metabolic balance influences microglia activity: Targeting kynurenine monooxygenase to dampen neuroinflammation. Psychoneuroendocrinology 2018; 94:1-10. [PMID: 29734055 PMCID: PMC5995655 DOI: 10.1016/j.psyneuen.2018.04.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/12/2018] [Accepted: 04/20/2018] [Indexed: 10/17/2022]
Abstract
Chronic stress or inflammation increases tryptophan metabolism along the kynurenine pathway (KP), and the generation of neuroactive kynurenine metabolites contributes to subsequent depressive-like behaviors. Microglia regulate KP balance by preferentially producing oxidative metabolites, including quinolinic acid. Research has focused on the interplay between cytokines and HPA axis-derived corticosteroids in regulating microglial activity and effects of KP metabolites directly on neurons; however, the potential role that KP metabolites have directly on microglial activity is unknown. Here, murine microglia were stimulated with lipopolysaccharide(LPS). After 6 h, mRNA expression of interleukin(IL)-1β, IL-6, tumor necrosis factor(TNF)-α and inducible nitric oxide synthase(iNOS) was dose-dependently increased along with the rate-limiting enzymes for oxidative KP metabolism, indoleamine-2,3-dioxygenase(IDO)-1 and kynurenine 3-monooxygenase(KMO). By 24 h post-LPS, kynurenine and quinolinic acid in the media was elevated. Inhibiting KMO with Ro 61-8048 during LPS challenge attenuated extracellular nitrite accumulation and expression of KMO and TNF-α in response to LPS. Similarly, primary microglia isolated from KMO-/- mice exhibited a significantly reduced pro-inflammatory response to LPS compared to WT controls. To determine whether the substrate (kynurenine) or end product (quinolinic acid) of KMO-dependent metabolism modulates the LPS response, microglia were treated with increasing concentrations of L-kynurenine or quinolinic acid in combination with LPS or saline. Interestingly, quinolinic acid did not impact the microglial LPS response. However, L-kynurenine had dose-dependent inhibitory effect on the LPS response. These data are the first to show an anti-inflammatory effect of KMO inhibition on microglia during immune challenge and suggest that KP metabolic balance may play a direct role in regulating microglia activity.
Collapse
Affiliation(s)
- Allison M. Garrison
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio
| | - Jennifer M. Parrott
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio
| | - Arnulfo Tuñon
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio
| | - Jennifer Delgado
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio
| | - Laney Redus
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio
| | - Jason C. O'Connor
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio,The Center for Biomedical Neuroscience, School of Medicine, The University of Texas Health Science Center at San Antonio,Audie L. Murphy VA Hospital, South Texas Veterans Health System, San Antonio, Texas
| |
Collapse
|
121
|
Wu ZS, Lo JJ, Wu SH, Wang CZ, Chen RF, Lee SS, Chai CY, Huang SH. Early Hyperbaric Oxygen Treatment Attenuates Burn-Induced Neuroinflammation by Inhibiting the Galectin-3-Dependent Toll-Like Receptor-4 Pathway in a Rat Model. Int J Mol Sci 2018; 19:ijms19082195. [PMID: 30060489 PMCID: PMC6121430 DOI: 10.3390/ijms19082195] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/22/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
Hyperbaric oxygen (HBO) treatment has been proven to decrease neuroinflammation in rats. This study aimed to determine the potential mechanism underlying the anti-inflammatory effects of HBO treatment on burn-induced neuroinflammation in rats. Thirty-six adult male Sprague-Dawley (SD) rats were randomly assigned to the following six groups (n = 6 per group): (1) sham burn with sham HBO treatment; (2) sham burn with HBO treatment; (3) burn with one-week sham HBO treatment; (4) burn with two-week sham HBO treatment; (5) burn with one-week HBO treatment; and (6) burn with two-week HBO treatment. SD rats that received third-degree burn injury were used as a full-thickness burn injury model. Subsequently, we analyzed the expression of proteins involved in the galectin-3 (Gal-3)-dependent Toll-like receptor-4 (TLR-4) pathway through enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC) analysis, and Western blotting. A behavior test was also conducted, which revealed that HBO treatment significantly suppressed mechanical hypersensitivity in the burn with HBO treatment group compared to the burn with sham HBO treatment group (p < 0.05). ELISA results showed that tumor necrosis factor α (TNF-α) and interleukin 1 beta (IL-1β) levels in the dorsal horn of the spinal cord and the skin significantly decreased in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). Western blotting results demonstrated that HBO treatment significantly reduced the expression of Gal-3 and TLR-4 in the dorsal horn of the spinal cord in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). IHC analysis showed that the expression of Gal-3, TLR-4, CD68 and CD45 in the dorsal horn of the spinal cord was significantly lower in the burn with HBO treatment group than in the burn with sham HBO treatment group (p < 0.05), and the expression of CD68 and macrophage migration inhibitory factor (MIF) in the right hind paw skin was significantly lower. The expression of vimentin and fibroblast growth factor in the right hind paw skin was significantly higher after HBO treatment (p < 0.05). This study proved that early HBO treatment relieves neuropathic pain, inhibits the Gal-3-dependent TLR-4 pathway, and suppresses microglia and macrophage activation in a rat model.
Collapse
Affiliation(s)
- Zong-Sheng Wu
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Jing-Jou Lo
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Sheng-Hua Wu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, 807 Kaohsiung, Taiwan.
- Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Chau-Zen Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
- Department of Physiology, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, 807 Kaohsiung, Taiwan.
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Rong-Fu Chen
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, 807 Kaohsiung, Taiwan.
| | - Su-Shin Lee
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, 807 Kaohsiung, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Shu-Hung Huang
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, 807 Kaohsiung, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
- Hyperbaric Oxygen Therapy Room, Kaohsiung Medical University Hospital, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| |
Collapse
|
122
|
Galectin-3 in M2 Macrophages Plays a Protective Role in Resolution of Neuropathology in Brain Parasitic Infection by Regulating Neutrophil Turnover. J Neurosci 2018; 38:6737-6750. [PMID: 29946038 DOI: 10.1523/jneurosci.3575-17.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/17/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Abstract
Macrophages/microglia with M2-activation phenotype are thought to play important anti-inflammatory and tissue reparative functions in the brain, yet the molecular bases of their functions in the CNS remain to be clearly defined. In a preclinical model of neurocysticercosis using brain infection with a parasite Mesocestoides corti, we previously reported the presence of large numbers of M2 cells in the CNS. In this study using female mice, we report that M2 macrophages in the parasite-infected brain display abundant galectin-3 expression. Disease severity was increased in Galectin-3-/- mice correlating with increased neurological defects, augmented cell death and, importantly, massive accumulation of neutrophils and M2 macrophages in the CNS of these mice. Because neutrophil clearance by efferocytosis is an important function of M2 macrophages, we investigated a possible role of galectin-3 in this process. Indeed, galectin-3-deficient M2 macrophages exhibited a defect in efferocytic clearance of neutrophils in vitro Furthermore, adoptive transfer of M2 macrophages from galectin-3-sufficient WT mice reduced neutrophilia in the CNS and ameliorated disease severity in parasite-infected Galectin-3-/- mice. Together, these results demonstrate, for the first time, a novel role of galectin-3 in M2 macrophage function in neutrophil turnover and resolution of inflammatory pathology in the CNS. This likely will have implications in neurocysticercosis and neuroinflammatory diseases.SIGNIFICANCE STATEMENT Macrophages/microglia with M1-activation phenotype are thought to promote CNS pathology, whereas M2-anti-inflammatory phenotype promote CNS repair. However, the mechanisms regulating M2 cell-protective functions in the CNS microenvironment are undefined. The current study reports that helminth infection of the brain induces an increased expression of galectin-3 in M2 macrophages accumulated in the CNS. Using multiple experimental models in vivo and in vitro, they show that galectin-3 in M2 macrophages functions to clear neutrophils accumulated in the CNS. Importantly, galectin-3 in M2 macrophages plays a central role in the containment of neuropathology and disease severity. These results provide a direct mechanistic evidence of the protective function of M2 macrophages in the CNS.
Collapse
|
123
|
Wang J, Xing H, Wan L, Jiang X, Wang C, Wu Y. Treatment targets for M2 microglia polarization in ischemic stroke. Biomed Pharmacother 2018; 105:518-525. [PMID: 29883947 DOI: 10.1016/j.biopha.2018.05.143] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
As the first line of defense in the nervous system, resident microglia are the predominant immune cells in the brain. In diseases of the central nervous system such as stroke, Alzheimer's disease, and Parkinson's disease, they often cause inflammation or phagocytosis; however, some studies have found that despite the current controversy over M1, M2 polarization could be beneficial. Ischemic stroke is the third most common cause of death in humans. Patients who survive an ischemic stroke might experience a clear decline in their quality of life, owing to conditions such as hemiplegic paralysis and aphasia. After stroke, the activated microglia become a double-edged sword, with distinct phenotypic changes to the deleterious M1 and neuroprotective M2 types. Therefore, methods for promoting the differentiation of microglia into the M2 polarized form to alleviate harmful reactions after stroke have become a topic of interest in recent years. Subsequently, the discovery of new drugs related to M2 polarization has enabled the realization of targeted therapies. In the present review, we discussed the neuroprotective effects of microglia M2 polarization and the potential mechanisms and drugs by which microglia can be transformed into the M2 polarized type after stroke.
Collapse
Affiliation(s)
- Ji Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongyi Xing
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Wan
- The Children's Hospital of Soochow, Jiangsu, Hematology and Oncology, China
| | - Xingjun Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
124
|
Chen Y, Yin M, Cao X, Hu G, Xiao M. Pro- and Anti-inflammatory Effects of High Cholesterol Diet on Aged Brain. Aging Dis 2018; 9:374-390. [PMID: 29896426 PMCID: PMC5988593 DOI: 10.14336/ad.2017.0706] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022] Open
Abstract
Both hypercholesterolemia and aging are related to cognitive decline or Alzheimer's disease. However, their interactive influence on the neurodegenerative progress remains unclear. To address this issue, 6-month-old and 16-month-old female mice were fed a 3% cholesterol diet for 8 weeks, followed by hippocampus-related functional, pathological, biochemical and molecular analyses. The high cholesterol diet did not exacerbate age-dependent cognitive decline and hippocampal neuronal death, and even greatly mitigated decreases of synaptophysin and growth associated protein 43 expression in the hippocampus of aged mice. Compared with young controls, aged mice fed normal diet showed mild activation of hippocampal microglia with increased expression of CD68, a marker of the microglial M1 phenotype, and decreased expression of CD206, a marker of the microglial M2 phenotype. More interestingly, the high cholesterol diet not only improved NLRP3 inflammasome activation and IL-1β expression, but also increased levels of anti-inflammatory cytokines IL-4 and IL-6 in the hippocampus of old mice, suggesting playing pro- and anti-neuroinflammatory effects. In addition, the cholesterol rich diet resulted in a defect of the blood-brain barrier of aged hippocampus, as revealed by increased brain albumin content. These results have revealed both harmful and protective effects of high cholesterol diet on aged brain, which helps us to understand that hypercholesterolemia in the aged population is not associated with dementia and cognitive impairment.
Collapse
Affiliation(s)
- Yali Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Mengmei Yin
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Xuejin Cao
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| |
Collapse
|
125
|
Yu Y, Shen Q, Lai Y, Park SY, Ou X, Lin D, Jin M, Zhang W. Anti-inflammatory Effects of Curcumin in Microglial Cells. Front Pharmacol 2018; 9:386. [PMID: 29731715 PMCID: PMC5922181 DOI: 10.3389/fphar.2018.00386] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 04/04/2018] [Indexed: 11/26/2022] Open
Abstract
Lipoteichoic acid (LTA) induces neuroinflammatory molecules, contributing to the pathogenesis of neurodegenerative diseases. Therefore, suppression of neuroinflammatory molecules could be developed as a therapeutic method. Although previous data supports an immune-modulating effect of curcumin, the underlying signaling pathways are largely unidentified. Here, we investigated curcumin’s anti-neuroinflammatory properties in LTA-stimulated BV-2 microglial cells. Inflammatory cytokine tumor necrosis factor-α [TNF-α, prostaglandin E2 (PGE2), and Nitric Oxide (NO] secretion in LTA-induced microglial cells were inhibited by curcumin. Curcumin also inhibited LTA-induced inducible NO synthases (iNOS) and cyclooxygenase-2 (COX-2) expression. Subsequently, our mechanistic studies revealed that curcumin inhibited LTA-induced phosphorylation of mitogen-activated protein kinase (MAPK) including ERK, p38, Akt and translocation of NF-κB. Furthermore, curcumin induced hemeoxygenase (HO)-1HO-1 and nuclear factor erythroid 2-related factor 2 (Nrf-2) expression in microglial cells. Inhibition of HO-1 reversed the inhibition effect of HO-1 on inflammatory mediators release in LTA-stimulated microglial cells. Taken together, our results suggest that curcumin could be a potential therapeutic agent for the treatment of neurodegenerative disorders via suppressing neuroinflammatory responses.
Collapse
Affiliation(s)
- Yangyang Yu
- Shenzhen University Health Science Center, Shenzhen, China
| | - Qian Shen
- Shenzhen University Health Science Center, Shenzhen, China
| | - Yihong Lai
- Shenzhen University Health Science Center, Shenzhen, China
| | - Sun Y Park
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan, South Korea
| | - Xingmei Ou
- Shenzhen University Health Science Center, Shenzhen, China
| | - Dongxu Lin
- Shenzhen University Health Science Center, Shenzhen, China
| | - Meiling Jin
- Shenzhen University Health Science Center, Shenzhen, China
| | - Weizhen Zhang
- Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
126
|
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 2018; 233:6425-6440. [PMID: 29319160 DOI: 10.1002/jcp.26429] [Citation(s) in RCA: 3058] [Impact Index Per Article: 436.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
Abstract
Macrophages are heterogeneous and their phenotype and functions are regulated by the surrounding micro-environment. Macrophages commonly exist in two distinct subsets: 1) Classically activated or M1 macrophages, which are pro-inflammatory and polarized by lipopolysaccharide (LPS) either alone or in association with Th1 cytokines such as IFN-γ, GM-CSF, and produce pro-inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, IL-12, IL-23, and TNF-α; and 2) Alternatively activated or M2 macrophages, which are anti-inflammatory and immunoregulatory and polarized by Th2 cytokines such as IL-4 and IL-13 and produce anti-inflammatory cytokines such as IL-10 and TGF-β. M1 and M2 macrophages have different functions and transcriptional profiles. They have unique abilities by destroying pathogens or repair the inflammation-associated injury. It is known that M1/M2 macrophage balance polarization governs the fate of an organ in inflammation or injury. When the infection or inflammation is severe enough to affect an organ, macrophages first exhibit the M1 phenotype to release TNF-α, IL-1β, IL-12, and IL-23 against the stimulus. But, if M1 phase continues, it can cause tissue damage. Therefore, M2 macrophages secrete high amounts of IL-10 and TGF-β to suppress the inflammation, contribute to tissue repair, remodeling, vasculogenesis, and retain homeostasis. In this review, we first discuss the basic biology of macrophages including origin, differentiation and activation, tissue distribution, plasticity and polarization, migration, antigen presentation capacity, cytokine and chemokine production, metabolism, and involvement of microRNAs in macrophage polarization and function. Secondly, we discuss the protective and pathogenic role of the macrophage subsets in normal and pathological pregnancy, anti-microbial defense, anti-tumor immunity, metabolic disease and obesity, asthma and allergy, atherosclerosis, fibrosis, wound healing, and autoimmunity.
Collapse
Affiliation(s)
- Abbas Shapouri-Moghaddam
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Mohammadian
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Vazini
- Nursing Department, Basic Sciences Faculty, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Mahdi Taghadosi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed-Alireza Esmaeili
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mardani
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bita Seifi
- Department of Anatomy, Islamic Azad University, Mashhad Branch, Iran
| | - Asadollah Mohammadi
- Inflammation and Inflammatory Disease Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil T Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
127
|
Ferrari P, Parisi MM, Colombo R, Becker M, Fries G, Ascoli BM, Géa LP, Anna MKS, Kapczinski F, Klamt F, Guma FTCR, Rosa AR, Barbé-Tuana FM. Depression and Mania Induce Pro-inflammatory Activation of Macrophages Following Application of Serum from Individuals with Bipolar Disorder. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2018; 16:103-108. [PMID: 29397672 PMCID: PMC5810451 DOI: 10.9758/cpn.2018.16.1.103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/21/2017] [Accepted: 04/28/2017] [Indexed: 12/30/2022]
Abstract
Objective Evidence has suggested that immune imbalance is involved with bipolar disorder (BD); however, its precise mechanism is poorly understood. This study investigated whether biochemical changes in the serum from BD patients could modulate the phenotype of cultured macrophages. Methods Eighteen subjects with BD and five healthy individuals were included in this study. The human monocyte cell line U-937 was activated with phorbol 12-myristate 13-acetate (PMA) and polarization was induced with RPMI-1640 media supplemented with 10% serum from each patient for 24 hours. Gene expression of selected M1 and M2 markers was assessed by quantitative PCR. Results Macrophages exposed to serum of manic and depressive BD patients displayed an increase of interleukin-1β (6.40±3.47 and 9.04±5.84 vs. 0.23±0.11; p<0.05) and tumor necrosis factor-α (2.23±0.91 and 2.03±0.45 vs. 0.62±0.24; p=0.002 and p=0.004, respectively) compared to euthymic group (there was no difference between euthymic and controls). In parallel, U-937 macrophages treated with serum of patients in acute episode displayed a down-regulation of CXCL9 (0.29±0.20 vs. 1.86±1.61; p=0.006) and CXCL10 expression (0.36±0.15 and 0.86±0.24 vs. 1.83±0.88; p<0.000 and p=0.04) compared to the euthymia group. Conclusion Our results are consistent with previous studies showing that changes in peripheral blood markers could modulate M1/M2 polarization in BD. The evidence of macrophages as source of inflammatory cytokines might be helpful to unravel how the mononuclear phagocyte system is involved in the etiology of BD.
Collapse
Affiliation(s)
- Pamela Ferrari
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program: Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Mariana Migliorini Parisi
- Laboratory of Molecular Biology and Bioinformatics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rafael Colombo
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Laboratory of Pharmacology and Physiology, Universidade de Caxias do Sul, Petrópolis, Brazil
| | - Matheus Becker
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Gabriel Fries
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Bruna Maria Ascoli
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program: Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luiza Paul Géa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Pharmacology and Postgraduate Program: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Márcia Kauer-Sant Anna
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program: Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Flávio Kapczinski
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program: Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Fábio Klamt
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Fátima T C R Guma
- Laboratory of Molecular Biology and Bioinformatics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Biochemistry and Cellular Biology of Lipids, Department of Biochemistry, ICBS/UFRGS, Porto Alegre, Brazil
| | - Adriane Ribeiro Rosa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Postgraduate Program: Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Pharmacology and Postgraduate Program: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Florencia M Barbé-Tuana
- Laboratory of Molecular Biology and Bioinformatics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
128
|
Lou ZY, Cheng J, Wang XR, Zhao YF, Gan J, Zhou GY, Liu ZG, Xiao BG. The inhibition of CB 1 receptor accelerates the onset and development of EAE possibly by regulating microglia/macrophages polarization. J Neuroimmunol 2018; 317:37-44. [PMID: 29501084 DOI: 10.1016/j.jneuroim.2018.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022]
Abstract
Cannabinoid 1 receptor (CB1R) regulates the neuro-inflammatory and neurodegenerative damages of experimental autoimmune encephalomyelitis (EAE) and of multiple sclerosis (MS). The mechanism by which CB1R inhibition exerts inflammatory effects is still unclear. Here, we explored the cellular and molecular mechanisms of CB1R in the treatment of EAE by using a specific and selective CB1R antagonist SR141716A. Our study demonstrated that SR141716A accelerated the clinical onset and development of EAE, accompanied by body weight loss. SR141716A significantly up-regulated the expression of toll like receptor-4 (TLR-4) and nuclear factor-kappaB/p65 (NF-κB/p65) on microglia/macrophages of EAE mice as well as levels of inflammatory factors (TNF-α, IL-1β, IL-6) and chemokines (MCP-1, CX3CL1), accompanied by the shifts of cytokines from Th2 (IL-4, IL-10) to Th1 (IFN-γ)/Th17 (IL-17) in the spinal cords of EAE mice. Similar changes happened on splenic mononuclear cells (MNCs) except chemokine CX3CL1. Consistently, SR141716A promoted BV-2 microglia to release inflammatory factors (TNF-α, IL-1β, IL-6) while inhibited the production of IL-10 and chemokines (MCP-1, CX3CL1). Furthermore, when splenic CD4+ T cells co-cultured with SR141716A-administered BV-2 microglia, the levels of IL-4 and IL-10 were decreased while production of IL-17 and IFN-γ increased significantly. Our research indicated that inhibition of CB1R induced M1 phenotype-Th17 axis changed of microglia/macrophages through TLR-4 and NF-κB/p65 which accelerated the onset and development of EAE. Therefore, CB1R may be a promising target for the treatment of MS/EAE, but its complexity remains to be carefully considered and studied in further clinical application.
Collapse
Affiliation(s)
- Zhi-Yin Lou
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jie Cheng
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao-Rong Wang
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yong-Fei Zhao
- Department of Neurology, JinShan Hospital, Fudan Univeristy, Shanghai, China
| | - Jing Gan
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guo-Yu Zhou
- Department of Geriatric, Qilu Hospital, Shandong University, Jinan, China
| | - Zhen-Guo Liu
- Department of Neurology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
129
|
Huo W, Zhang Y, Liu Y, Lei Y, Sun R, Zhang W, Huang Y, Mao Y, Wang C, Ma Z, Gu X. Dehydrocorydaline attenuates bone cancer pain by shifting microglial M1/M2 polarization toward the M2 phenotype. Mol Pain 2018; 14:1744806918781733. [PMID: 29882480 PMCID: PMC6009085 DOI: 10.1177/1744806918781733] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/13/2018] [Accepted: 05/11/2018] [Indexed: 12/29/2022] Open
Abstract
Bone cancer pain remains a major challenge in patients with primary or metastatic bone cancer due to a lack of understanding the mechanisms. Previous studies have revealed the two distinct functional polarization states of microglia (classically activated M1 and alternatively activated M2) in the spinal cord in nerve injury-induced neuropathic pain. However, whether microglia in the spinal cord polarize to M1 and M2 phenotypes and contribute to the development of bone cancer pain remains unclear. In this study, we used a mouse model with bone cancer to characterize the M1/M2 polarization of microglia in the spinal cord during the development of bone cancer pain, and investigated the antinociceptive effects of dehydrocorydaline, an alkaloidal component isolated from Rhizoma corydalis on bone cancer pain. Our results show that microglia in the spinal cord presented increased M1 polarization and decreased M2 polarization, while overproduction of IL-1β and inhibited expression of IL-10 was detected during bone cancer pain development. Intraperitoneal administration of dehydrocorydaline (10 mg/kg) had significant antinociceptive effects on day 14 after osteosarcoma cell implantation, accompanied by suppressed M1 phenotype and upregulated M2 phenotype of microglia in the spinal cord, while alleviated inflammatory response was observed then. These results suggest that the imbalanced polarization of microglia toward the M1 phenotype in the spinal cord may contribute to the development of bone cancer pain, while dehydrocorydaline helps to attenuate bone cancer pain, with microglial polarization shifting toward the M2 phenotype in the spinal cord.
Collapse
Affiliation(s)
- Wenwen Huo
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Ying Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yue Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yishan Lei
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Rao Sun
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Wei Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yulin Huang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yanting Mao
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Chenchen Wang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| |
Collapse
|
130
|
The Role of Mammalian Glial Cells in Circadian Rhythm Regulation. Neural Plast 2017; 2017:8140737. [PMID: 29435373 PMCID: PMC5757113 DOI: 10.1155/2017/8140737] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/19/2017] [Indexed: 11/23/2022] Open
Abstract
Circadian rhythms are biological oscillations with a period of about 24 hours. These rhythms are maintained by an innate genetically determined time-keeping system called the circadian clock. A large number of the proteins involved in the regulation of this clock are transcription factors controlling rhythmic transcription of so-called clock-controlled genes, which participate in a plethora of physiological functions in the organism. In the brain, several areas, besides the suprachiasmatic nucleus, harbor functional clocks characterized by a well-defined time pattern of clock gene expression. This expression rhythm is not restricted to neurons but is also present in glia, suggesting that these cells are involved in circadian rhythmicity. However, only certain glial cells fulfill the criteria to be called glial clocks, namely, to display molecular oscillators based on the canonical clock protein PERIOD, which depends on the suprachiasmatic nucleus for their synchronization. In this contribution, we summarize the current information about activity of the clock genes in glial cells, their potential role as oscillators as well as clinical implications.
Collapse
|
131
|
Jiang T, Zhang L, Pan X, Zheng H, Chen X, Li L, Luo J, Hu X. Physical Exercise Improves Cognitive Function Together with Microglia Phenotype Modulation and Remyelination in Chronic Cerebral Hypoperfusion. Front Cell Neurosci 2017; 11:404. [PMID: 29311834 PMCID: PMC5743796 DOI: 10.3389/fncel.2017.00404] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 12/04/2017] [Indexed: 01/10/2023] Open
Abstract
Myelin is closely associated with cognitive function and is extremely vulnerable to damage in ischemic cerebrovascular diseases. The failure of remyelination is mainly due to limitations in oligodendrocyte progenitor cells (OPCs) differentiation in the damaged area. Previous studies have shown that physical exercise can improve vascular cognitive impairment, but whether it can reverse the defect in remyelination during ischemic injury and the underlying mechanism remains unclear. In this study, we observed the effects of physical exercise on chronic cerebral hypoperfusion (CCH) established by bilateral carotid artery occlusion. The cognitive function, myelin integrity, OPCs proliferation and differentiation, as well as microglia polarization were analyzed at 28 days after CCH. Besides, the expression of CX3CL1/CX3CR1 axis and activation of mitogen-activated protein kinase (MAPK) signal cascades were also evaluated. We found that physical exercise improved the cognitive function of rats with CCH, alleviated myelin injury, triggered OPCs proliferation and differentiation, facilitated microglia polarization toward M2, augmented the expression of CX3CL1/CX3CR1 axis, and reduced ERK and JNK phosphorylation. The results indicated that physical exercise improved the cognitive function of rats with CCH, possibly through microglial phenotype modulation and enhancement of oligodendrocytegenesis and remyelination. Moreover, the CX3CL1/CX3CR1 axis played an important role in this process by mediating ERK- and JNK-dependent pathways.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaona Pan
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lili Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
132
|
Lannes N, Eppler E, Etemad S, Yotovski P, Filgueira L. Microglia at center stage: a comprehensive review about the versatile and unique residential macrophages of the central nervous system. Oncotarget 2017; 8:114393-114413. [PMID: 29371994 PMCID: PMC5768411 DOI: 10.18632/oncotarget.23106] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023] Open
Abstract
Microglia cells are the unique residential macrophages of the central nervous system (CNS). They have a special origin, as they derive from the embryonic yolk sac and enter the developing CNS at a very early stage. They play an important role during CNS development and adult homeostasis. They have a major contribution to adult neurogenesis and neuroinflammation. Thus, they participate in the pathogenesis of neurodegenerative diseases and contribute to aging. They play an important role in sustaining and breaking the blood-brain barrier. As innate immune cells, they contribute substantially to the immune response against infectious agents affecting the CNS. They play also a major role in the growth of tumours of the CNS. Microglia are consequently the key cell population linking the nervous and the immune system. This review covers all different aspects of microglia biology and pathology in a comprehensive way.
Collapse
Affiliation(s)
- Nils Lannes
- Albert Gockel, Anatomy, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Elisabeth Eppler
- Pestalozzistrasse Zo, Department of BioMedicine, University of Basel, CH-4056 Basel, Switzerland
| | - Samar Etemad
- Building 71/218 RBWH Herston, Centre for Clinical Research, The University of Queensland, QLD 4029 Brisbane, Australia
| | - Peter Yotovski
- Albert Gockel, Anatomy, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Luis Filgueira
- Albert Gockel, Anatomy, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
133
|
Jin M, Park SY, Shen Q, Lai Y, Ou X, Mao Z, Lin D, Yu Y, Zhang W. Anti-neuroinflammatory effect of curcumin on Pam3CSK4-stimulated microglial cells. Int J Mol Med 2017; 41:521-530. [PMID: 29115589 DOI: 10.3892/ijmm.2017.3217] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 10/24/2017] [Indexed: 11/05/2022] Open
Abstract
Curcumin is the main curcuminoid present in Curcuma longa and it has been previously reported to exhibit a wide range of pharmacological activities. In the present study, the inhibitory effects of curcumin on the inflammatory mediators released by Pam3CSK4-stimulated BV-2 microglial cells were investigated. The production of pro-inflammatory mediators and cytokines, including tumor necrosis factor-α (TNF-α) and prostaglandin E2 (PGE2), were measured by enzyme‑linked immunosorbent assay (ELISA). The expression of inflammatory genes, including inducible nitric oxide synthase and cyclooxygenase-2, were further investigated using reverse transcription-quantitative polymerase chain reaction. The effects of curcumin on heme oxygenase-1 (HO-1), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signaling pathways were analyzed by western blotting. The results revealed that curcumin dose-dependently inhibited Pam3CSK4-induced nitric oxide, PGE2, and TNF-α secretion. Curcumin suppressed the secretion of inflammatory mediators through an increase in the expression of HO-1. Curcumin induced HO-1 transcription and translation through the Nrf2/antioxidant response element signaling pathway. Inhibitory experiments revealed that HO-1 was required for the anti-inflammatory effects of curcumin. Further mechanistic studies demonstrated that curcumin inhibited neuroinflammation by suppressing NF-κB and MAPK signaling pathways in Pam3CSK4-activated microglial cells. The results of the present study suggest that curcumin may be a novel treatment for neuroinflammation-mediated neurodegenerative disorders.
Collapse
Affiliation(s)
- Meiling Jin
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Sun Young Park
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan 609-735, Republic of Korea
| | - Qian Shen
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Yihong Lai
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Xingmei Ou
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Zhuo Mao
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Dongxu Lin
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Yangyang Yu
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| | - Weizhen Zhang
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
134
|
Ahmed A, Wang LL, Abdelmaksoud S, Aboelgheit A, Saeed S, Zhang CL. Minocycline modulates microglia polarization in ischemia-reperfusion model of retinal degeneration and induces neuroprotection. Sci Rep 2017; 7:14065. [PMID: 29070819 PMCID: PMC5656679 DOI: 10.1038/s41598-017-14450-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Retinal ischemia-reperfusion (IR) injury causes irreversible loss of neurons and ultimately leads to permanent visual impairment and blindness. The cellular response under this pathological retinal condition is less clear. Using genetically modified mice, we systematically examined the behavior of microglia/macrophages after injury. We show that IR leads to activation of microglia/macrophages indicated by migration and proliferation of resident microglia and recruitment of circulating monocytes. IR-induced microglia/macrophages associate with apoptotic retinal neurons. Very interestingly, neuron loss can be mitigated by minocycline treatment. Minocycline induces Il4 expression and M2 polarization of microglia/macrophages. IL4 neutralization dampens minocycline-induced M2 polarization and neuroprotection. Given a well-established safety profile as an antibiotic, our results provide a rationale for using minocycline as a therapeutic agent for treating ischemic retinal degeneration.
Collapse
Affiliation(s)
- Amel Ahmed
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA.,Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Lei-Lei Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Safaa Abdelmaksoud
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal Aboelgheit
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Safaa Saeed
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA. .,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA.
| |
Collapse
|
135
|
Isgren A, Sellgren C, Ekman CJ, Holmén-Larsson J, Blennow K, Zetterberg H, Jakobsson J, Landén M. Markers of neuroinflammation and neuronal injury in bipolar disorder: Relation to prospective clinical outcomes. Brain Behav Immun 2017; 65:195-201. [PMID: 28483660 DOI: 10.1016/j.bbi.2017.05.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/21/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022] Open
Abstract
Neuroimmune mechanisms have been linked to the pathophysiology of bipolar disorder based on studies of biomarkers in plasma, cerebrospinal fluid (CSF), and postmortem brain tissue. There are, however, no longitudinal studies investigating if CSF markers of neuroinflammation and neuronal injury predict clinical outcomes in patients with bipolar disorder. We have in previous studies found higher CSF concentrations of interleukin-8 (IL-8), monocyte chemoattractant protein 1 (MCP-1/CCL-2), chitinase-3-like protein 1 (CHI3L1/YKL-40), and neurofilament light chain (NF-L) in euthymic patients with bipolar disorder compared with controls. Here, we investigated the relationship of these CSF markers of neuroinflammation and neuronal injury with clinical outcomes in a prospective study. 77 patients with CSF analyzed at baseline were followed for 6-7years. Associations of baseline biomarkers with clinical outcomes (manic/hypomanic and depressive episodes, suicide attempts, psychotic symptoms, inpatient care, GAF score change) were investigated. Baseline MCP-1 concentrations were positively associated with manic/hypomanic episodes and inpatient care during follow-up. YKL-40 concentrations were negatively associated with manic/hypomanic episodes and with occurrence of psychotic symptoms. The prospective negative association between YKL-40 and manic/hypomanic episodes survived multiple testing correction. Concentrations of IL-8 and NF-L were not associated with clinical outcomes. High concentrations of these selected CSF markers of neuroinflammation and neuronal injury at baseline were not consistently associated with poor clinical outcomes in this prospective study. The assessed proteins may be involved in adaptive immune processes or reflect a state of vulnerability for bipolar disorder rather than being of predictive value for disease progression.
Collapse
Affiliation(s)
- Anniella Isgren
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg and Mölndal, Sweden.
| | - Carl Sellgren
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carl-Johan Ekman
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Holmén-Larsson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg and Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg and Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg and Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Joel Jakobsson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg and Mölndal, Sweden
| | - Mikael Landén
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg and Mölndal, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
136
|
van Buel EM, Sigrist H, Seifritz E, Fikse L, Bosker FJ, Schoevers RA, Klein HC, Pryce CR, Eisel ULM. Mouse repeated electroconvulsive seizure (ECS) does not reverse social stress effects but does induce behavioral and hippocampal changes relevant to electroconvulsive therapy (ECT) side-effects in the treatment of depression. PLoS One 2017; 12:e0184603. [PMID: 28910337 PMCID: PMC5598988 DOI: 10.1371/journal.pone.0184603] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/28/2017] [Indexed: 01/21/2023] Open
Abstract
Electroconvulsive therapy (ECT) is an effective treatment for depression, but can have negative side effects including amnesia. The mechanisms of action underlying both the antidepressant and side effects of ECT are not well understood. An equivalent manipulation that is conducted in experimental animals is electroconvulsive seizure (ECS). Rodent studies have provided valuable insights into potential mechanisms underlying the antidepressant and side effects of ECT. However, relatively few studies have investigated the effects of ECS in animal models with a depression-relevant manipulation such as chronic stress. In the present study, mice were first exposed to chronic social stress (CSS) or a control procedure for 15 days followed by ECS or a sham procedure for 10 days. Behavioral effects were investigated using an auditory fear conditioning (learning) and expression (memory) test and a treadmill-running fatigue test. Thereafter, immunohistochemistry was conducted on brain material using the microglial marker Iba-1 and the cholinergic fibre marker ChAT. CSS did not increase fear learning and memory in the present experimental design; in both the control and CSS mice ECS reduced fear learning and fear memory expression. CSS induced the expected fatigue-like effect in the treadmill-running test; ECS induced increased fatigue in CSS and control mice. In CSS and control mice ECS induced inflammation in hippocampus in terms of increased expression of Iba-1 in radiatum of CA1 and CA3. CSS and ECS both reduced acetylcholine function in hippocampus as indicated by decreased expression of ChAT in several hippocampal sub-regions. Therefore, CSS increased fatigue and reduced hippocampal ChAT activity and, rather than reversing these effects, a repeated ECS regimen resulted in impaired fear learning-memory, increased fatigue, increased hippocampal Iba-1 expression, and decreased hippocampal ChAT expression. As such, the current model does not provide insights into the mechanism of ECT antidepressant function but does provide evidence for pathophysiological mechanisms that might contribute to important ECT side-effects.
Collapse
Affiliation(s)
- Erin M. van Buel
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Nuclear Medicine & Molecular Imaging, Groningen, Netherlands
- Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, Netherlands
| | - Hannes Sigrist
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Erich Seifritz
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Lianne Fikse
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| | - Fokko J. Bosker
- University of Groningen, University Medical Centre Groningen, Dept of Nuclear Medicine & Molecular Imaging, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
| | - Robert A. Schoevers
- Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
| | - Hans C. Klein
- University of Groningen, University Medical Centre Groningen, Dept of Nuclear Medicine & Molecular Imaging, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
| | - Christopher R. Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Ulrich LM Eisel
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, Netherlands
- University of Groningen, University Medical Centre Groningen, Dept of Psychiatry, Groningen, Netherlands
- * E-mail:
| |
Collapse
|
137
|
Zhou T, Huang Z, Sun X, Zhu X, Zhou L, Li M, Cheng B, Liu X, He C. Microglia Polarization with M1/M2 Phenotype Changes in rd1 Mouse Model of Retinal Degeneration. Front Neuroanat 2017; 11:77. [PMID: 28928639 PMCID: PMC5591873 DOI: 10.3389/fnana.2017.00077] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 08/21/2017] [Indexed: 01/28/2023] Open
Abstract
Microglia activation is recognized as the hallmark of neuroinflammation. However, the activation profile and phenotype changes of microglia during the process of retinal degeneration are poorly understood. This study aimed to elucidate the time-spatial pattern of microglia distribution and characterize the polarized phenotype of activated microglia during retinal neuroinflammation and degeneration in rd1 (Pde6βrd1/rd1) mice, the classic model of inherited retinal degeneration. Retinae of rd1 mice at different postnatal days (P7, P14, P21, P28, P56, and P180) were prepared for further analysis. We found most CD11b+ or IBA1+ microglia expressed Ki-67 and CD68 in rd1 mice and these cells migrated toward the layer of degenerative photoreceptors at the rapid rods degeneration phase from P14 to P28. These microglia exhibited typical ameboid activated shape with round bodies and scarce dendrites, while at late phase at P180, they displayed resting ramified morphology with elongated dendrites. Flow cytometry revealed that the percentage of CD86+CD206- M1 microglia increased markedly in rd1 retinae, however, no significant change was observed in CD206+CD86- M2 microglia. Interestingly, CD86+CD206+ microglia, an intermediate state between the two extremes of M1 and M2, increased markedly at the rapid rods degeneration phase. The immunofluorescence images revealed that microglia in rd1 mice highly expressed M1 markers including CD16/32, CD86, and CD40. In addition, increased expression of pro-inflammatory cytokines (TNF-α, IL-6, and CCL2) was observed in rd1 mice. Our findings unfolded a panorama for the first time that microglia conducted distinctive behaviors with the progression of retinal degeneration in rd1 mice. Microglia is activated and particularly polarized to a pro-inflammatory M1 phenotype at the rapid rods degenerative phase, suggesting that the involvement of M1 microglia in the retinal neuroinflammation and degeneration. Most microglia adopted an intermediate polarization “M1½” state in rd1, revealing that microglia orchestrated a complicated continuous spectrum in degenerative retina.
Collapse
Affiliation(s)
- Tian Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Zijing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Xiaowei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Xiaowei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Lingli Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Mei Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Bing Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Chang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
138
|
Subramaniam SR, Federoff HJ. Targeting Microglial Activation States as a Therapeutic Avenue in Parkinson's Disease. Front Aging Neurosci 2017. [PMID: 28642697 PMCID: PMC5463358 DOI: 10.3389/fnagi.2017.00176] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Parkinson’s disease (PD) is a chronic and progressive disorder characterized neuropathologically by loss of dopamine neurons in the substantia nigra, intracellular proteinaceous inclusions, reduction of dopaminergic terminals in the striatum, and increased neuroinflammatory cells. The consequent reduction of dopamine in the basal ganglia results in the classical parkinsonian motor phenotype. A growing body of evidence suggest that neuroinflammation mediated by microglia, the resident macrophage-like immune cells in the brain, play a contributory role in PD pathogenesis. Microglia participate in both physiological and pathological conditions. In the former, microglia restore the integrity of the central nervous system and, in the latter, they promote disease progression. Microglia acquire different activation states to modulate these cellular functions. Upon activation to the M1 phenotype, microglia elaborate pro-inflammatory cytokines and neurotoxic molecules promoting inflammation and cytotoxic responses. In contrast, when adopting the M2 phenotype microglia secrete anti-inflammatory gene products and trophic factors that promote repair, regeneration, and restore homeostasis. Relatively little is known about the different microglial activation states in PD and a better understanding is essential for developing putative neuroprotective agents. Targeting microglial activation states by suppressing their deleterious pro-inflammatory neurotoxicity and/or simultaneously enhancing their beneficial anti-inflammatory protective functions appear as a valid therapeutic approach for PD treatment. In this review, we summarize microglial functions and, their dual neurotoxic and neuroprotective role in PD. We also review molecules that modulate microglial activation states as a therapeutic option for PD treatment.
Collapse
Affiliation(s)
| | - Howard J Federoff
- Department of Neurology, University of California, Irvine, Irvine, CAUnited States
| |
Collapse
|
139
|
Current understanding of methamphetamine-associated dopaminergic neurodegeneration and psychotoxic behaviors. Arch Pharm Res 2017; 40:403-428. [DOI: 10.1007/s12272-017-0897-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022]
|
140
|
Nemetchek MD, Stierle AA, Stierle DB, Lurie DI. The Ayurvedic plant Bacopa monnieri inhibits inflammatory pathways in the brain. JOURNAL OF ETHNOPHARMACOLOGY 2017; 197:92-100. [PMID: 27473605 PMCID: PMC5269610 DOI: 10.1016/j.jep.2016.07.073] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 07/14/2016] [Accepted: 07/25/2016] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bacopa monnieri (L) Wettst (common name, bacopa) is a medicinal plant used in Ayurveda, the traditional system of medicine of India, as a nootropic. It is considered to be a "medhya rasayana", an herb that sharpens the mind and the intellect. Bacopa is an important ingredient in many Ayurvedic herbal formulations designed to treat conditions such as memory loss, anxiety, poor cognition and loss of concentration. It has also been used in Ayurveda to treat inflammatory conditions such as arthritis. In modern biomedical studies, bacopa has been shown in animal models to inhibit the release of the pro-inflammatory cytokines TNF-α and IL-6. However, less is known regarding the anti-inflammatory activity of Bacopa in the brain. AIM OF THE STUDY The current study examines the ability of Bacopa to inhibit the release of pro-inflammatory cytokines from microglial cells, the immune cells of the brain that participate in inflammation in the CNS. The effect of Bacopa on signaling enzymes associated with CNS inflammatory pathways was also studied. MATERIALS AND METHODS Various extracts of Bacopa were prepared and examined in the N9 microglial cell line in order to determine if they inhibited the release of the proinflammatory cytokines TNF-α and IL-6. Extracts were also tested in cell free assays as inhibitors of caspase-1 and matrix metalloproteinase-3 (enzymes associated with inflammation) and caspase-3, which has been shown to cleave protein Tau, an early event in the development of Alzheimer's disease. RESULTS The tea, infusion, and alkaloid extracts of bacopa, as well as Bacoside A significantly inhibited the release of TNF-α and IL-6 from activated N9 microglial cells in vitro. In addition, the tea, infusion, and alkaloid extracts of Bacopa effectively inhibited caspase 1 and 3, and matrix metalloproteinase-3 in the cell free assay. CONCLUSIONS Bacopa inhibits the release of inflammatory cytokines from microglial cells and inhibits enzymes associated with inflammation in the brain. Thus, Bacopa can limit inflammation in the CNS, and offers a promising source of novel therapeutics for the treatment of many CNS disorders.
Collapse
Affiliation(s)
- Michelle D Nemetchek
- Department of Biomedical and Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula MT 59812, USA
| | - Andrea A Stierle
- Department of Biomedical and Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula MT 59812, USA
| | - Donald B Stierle
- Department of Biomedical and Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula MT 59812, USA
| | - Diana I Lurie
- Department of Biomedical and Pharmaceutical Sciences, College of Health Professions and Biomedical Sciences, The University of Montana, Missoula MT 59812, USA.
| |
Collapse
|
141
|
Peña-Ortega F. Pharmacological Tools to Activate Microglia and their Possible use to Study Neural Network Patho-physiology. Curr Neuropharmacol 2017; 15:595-619. [PMID: 27697040 PMCID: PMC5543677 DOI: 10.2174/1570159x14666160928151546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/05/2016] [Accepted: 09/26/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Microglia are the resident immunocompetent cells of the CNS and also constitute a unique cell type that contributes to neural network homeostasis and function. Understanding microglia cell-signaling not only will reveal their diverse functions but also will help to identify pharmacological and non-pharmacological tools to modulate the activity of these cells. METHODS We undertook a search of bibliographic databases for peer-reviewed research literature to identify microglial activators and their cell-specificity. We also looked for their effects on neural network function and dysfunction. RESULTS We identified several pharmacological targets to modulate microglial function, which are more or less specific (with the proper control experiments). We also identified pharmacological targets that would require the development of new potent and specific modulators. We identified a wealth of evidence about the participation of microglia in neural network function and their alterations in pathological conditions. CONCLUSION The identification of specific microglia-activating signals provides experimental tools to modulate the activity of this heterogeneous cell type in order to evaluate its impact on other components of the nervous system, and it also helps to identify therapeutic approaches to ease some pathological conditions related to microglial dysfunction.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| |
Collapse
|
142
|
Nakagawa Y. Psycho-Behavioral Spiral of Disturbances in Prosocial Behavior, Stress Response, and Self-Regulation inSubstance-Related and Addictive Disorders. ACTA ACUST UNITED AC 2017. [DOI: 10.4303/jdar/236017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
143
|
Suhara T, Chaki S, Kimura H, Furusawa M, Matsumoto M, Ogura H, Negishi T, Saijo T, Higuchi M, Omura T, Watanabe R, Miyoshi S, Nakatani N, Yamamoto N, Liou SY, Takado Y, Maeda J, Okamoto Y, Okubo Y, Yamada M, Ito H, Walton NM, Yamawaki S. Strategies for Utilizing Neuroimaging Biomarkers in CNS Drug Discovery and Development: CINP/JSNP Working Group Report. Int J Neuropsychopharmacol 2016; 20:285-294. [PMID: 28031269 PMCID: PMC5604546 DOI: 10.1093/ijnp/pyw111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/15/2016] [Indexed: 01/07/2023] Open
Abstract
Despite large unmet medical needs in the field for several decades, CNS drug discovery and development has been largely unsuccessful. Biomarkers, particularly those utilizing neuroimaging, have played important roles in aiding CNS drug development, including dosing determination of investigational new drugs (INDs). A recent working group was organized jointly by CINP and Japanese Society of Neuropsychopharmacology (JSNP) to discuss the utility of biomarkers as tools to overcome issues of CNS drug development.The consensus statement from the working group aimed at creating more nuanced criteria for employing biomarkers as tools to overcome issues surrounding CNS drug development. To accomplish this, a reverse engineering approach was adopted, in which criteria for the utilization of biomarkers were created in response to current challenges in the processes of drug discovery and development for CNS disorders. Based on this analysis, we propose a new paradigm containing 5 distinct tiers to further clarify the use of biomarkers and establish new strategies for decision-making in the context of CNS drug development. Specifically, we discuss more rational ways to incorporate biomarker data to determine optimal dosing for INDs with novel mechanisms and targets, and propose additional categorization criteria to further the use of biomarkers in patient stratification and clinical efficacy prediction. Finally, we propose validation and development of new neuroimaging biomarkers through public-private partnerships to further facilitate drug discovery and development for CNS disorders.
Collapse
Affiliation(s)
- Tetsuya Suhara
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Shigeyuki Chaki
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Haruhide Kimura
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Makoto Furusawa
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Mitsuyuki Matsumoto
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Hiroo Ogura
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Takaaki Negishi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Takeaki Saijo
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Tomohiro Omura
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Rira Watanabe
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Sosuke Miyoshi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Noriaki Nakatani
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Noboru Yamamoto
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Shyh-Yuh Liou
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Yuhei Takado
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Jun Maeda
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Yasumasa Okamoto
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Yoshiaki Okubo
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Makiko Yamada
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Hiroshi Ito
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Noah M. Walton
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| | - Shigeto Yamawaki
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Suhara, Higuchi, Takado, Maeda, and Yamada); Taisho Pharmaceutical Co., Ltd., Saitama, Japan (Drs Chaki and Omura); Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan (Drs Kimura and Furusawa); Astellas Pharma Inc., Ibaraki, Japan (Drs Matsumoto and Miyoshi); Eisai Co., Ltd., Tokyo, Japan (Drs Ogura and Yamamoto); Mochida Pharmaceutical Co., Ltd., Tokyo, Japan (Dr Negishi); Mitsubishi Tanabe Pharma Co., Kanagawa, Japan (Dr Saijo); Daiichi Sankyo Co., Ltd., Tokyo, Japan (Dr Watanabe); Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan (Dr Nakatani); Ono Pharmaceutical Co., Ltd., Osaka, Japan (Dr Liou); Hiroshima University, Hiroshima, Japan (Drs Okamoto and Yamawaki); Nippon Medical School, Tokyo, Japan (Dr Okubo); Fukushima Medical University, Fukushima, Japan (Dr Ito); Astellas Research Institute of America LLC, IL, USA (Dr Walton)
| |
Collapse
|
144
|
Brown JA, Codreanu SG, Shi M, Sherrod SD, Markov DA, Neely MD, Britt CM, Hoilett OS, Reiserer RS, Samson PC, McCawley LJ, Webb DJ, Bowman AB, McLean JA, Wikswo JP. Metabolic consequences of inflammatory disruption of the blood-brain barrier in an organ-on-chip model of the human neurovascular unit. J Neuroinflammation 2016; 13:306. [PMID: 27955696 PMCID: PMC5153753 DOI: 10.1186/s12974-016-0760-y] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/07/2016] [Indexed: 11/24/2022] Open
Abstract
Background Understanding blood-brain barrier responses to inflammatory stimulation (such as lipopolysaccharide mimicking a systemic infection or a cytokine cocktail that could be the result of local or systemic inflammation) is essential to understanding the effect of inflammatory stimulation on the brain. It is through the filter of the blood-brain barrier that the brain responds to outside influences, and the blood-brain barrier is a critical point of failure in neuroinflammation. It is important to note that this interaction is not a static response, but one that evolves over time. While current models have provided invaluable information regarding the interaction between cytokine stimulation, the blood-brain barrier, and the brain, these approaches—whether in vivo or in vitro—have often been only snapshots of this complex web of interactions. Methods We utilize new advances in microfluidics, organs-on-chips, and metabolomics to examine the complex relationship of inflammation and its effects on blood-brain barrier function ex vivo and the metabolic consequences of these responses and repair mechanisms. In this study, we pair a novel dual-chamber, organ-on-chip microfluidic device, the NeuroVascular Unit, with small-volume cytokine detection and mass spectrometry analysis to investigate how the blood-brain barrier responds to two different but overlapping drivers of neuroinflammation, lipopolysaccharide and a cytokine cocktail of IL-1β, TNF-α, and MCP1,2. Results In this study, we show that (1) during initial exposure to lipopolysaccharide, the blood-brain barrier is compromised as expected, with increased diffusion and reduced presence of tight junctions, but that over time, the barrier is capable of at least partial recovery; (2) a cytokine cocktail also contributes to a loss of barrier function; (3) from this time-dependent cytokine activation, metabolic signature profiles can be obtained for both the brain and vascular sides of the blood-brain barrier model; and (4) collectively, we can use metabolite analysis to identify critical pathways in inflammatory response. Conclusions Taken together, these findings present new data that allow us to study the initial effects of inflammatory stimulation on blood-brain barrier disruption, cytokine activation, and metabolic pathway changes that drive the response and recovery of the barrier during continued inflammatory exposure. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0760-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacquelyn A Brown
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA
| | - Simona G Codreanu
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mingjian Shi
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - Stacy D Sherrod
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Dmitry A Markov
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - M Diana Neely
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37203, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Clayton M Britt
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA
| | - Orlando S Hoilett
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA
| | - Ronald S Reiserer
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA
| | - Philip C Samson
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA
| | - Lisa J McCawley
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Donna J Webb
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA.,Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - Aaron B Bowman
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37203, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - John A McLean
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.,Center for Innovative Technology, Vanderbilt University, Nashville, TN, 37235, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - John P Wikswo
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, 37235, USA. .,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, 6301 Stevenson Center, Nashville, TN, 37235, USA. .,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA. .,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
145
|
Lobo-Silva D, Carriche GM, Castro AG, Roque S, Saraiva M. Balancing the immune response in the brain: IL-10 and its regulation. J Neuroinflammation 2016; 13:297. [PMID: 27881137 PMCID: PMC5121946 DOI: 10.1186/s12974-016-0763-8] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/11/2016] [Indexed: 12/15/2022] Open
Abstract
Background The inflammatory response is critical to fight insults, such as pathogen invasion or tissue damage, but if not resolved often becomes detrimental to the host. A growing body of evidence places non-resolved inflammation at the core of various pathologies, from cancer to neurodegenerative diseases. It is therefore not surprising that the immune system has evolved several regulatory mechanisms to achieve maximum protection in the absence of pathology. Main body The production of the anti-inflammatory cytokine interleukin (IL)-10 is one of the most important mechanisms evolved by many immune cells to counteract damage driven by excessive inflammation. Innate immune cells of the central nervous system, notably microglia, are no exception and produce IL-10 downstream of pattern recognition receptors activation. However, whereas the molecular mechanisms regulating IL-10 expression by innate and acquired immune cells of the periphery have been extensively addressed, our knowledge on the modulation of IL-10 expression by central nervous cells is much scattered. This review addresses the current understanding on the molecular mechanisms regulating IL-10 expression by innate immune cells of the brain and the implications of IL-10 modulation in neurodegenerative disorders. Conclusion The regulation of IL-10 production by central nervous cells remains a challenging field. Answering the many remaining outstanding questions will contribute to the design of targeted approaches aiming at controlling deleterious inflammation in the brain.
Collapse
Affiliation(s)
- Diogo Lobo-Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, Braga, Portugal
| | - Guilhermina M Carriche
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - A Gil Castro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, Braga, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, Braga, Portugal
| | - Margarida Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
146
|
Gonzales I, Rivera JT, Garcia HH. Pathogenesis of Taenia solium taeniasis and cysticercosis. Parasite Immunol 2016; 38:136-46. [PMID: 26824681 DOI: 10.1111/pim.12307] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022]
Abstract
Taenia solium infections (taeniasis/cysticercosis) are a major scourge to most developing countries. Neurocysticercosis, the infection of the human nervous system by the cystic larvae of this parasite, has a protean array of clinical manifestations varying from entirely asymptomatic infections to aggressive, lethal courses. The diversity of clinical manifestations reflects a series of contributing factors which include the number, size and location of the invading parasites, and particularly the inflammatory response of the host. This manuscript reviews the different presentations of T. solium infections in the human host with a focus on the mechanisms or processes responsible for their clinical expression.
Collapse
Affiliation(s)
- I Gonzales
- Cysticercosis Unit, Department of Transmissible Diseases, Instituto Nacional de Ciencias Neurológicas, Lima, Peru
| | - J T Rivera
- Department of Microbiology and Center for Global Health, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - H H Garcia
- Cysticercosis Unit, Department of Transmissible Diseases, Instituto Nacional de Ciencias Neurológicas, Lima, Peru.,Department of Microbiology and Center for Global Health, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | |
Collapse
|
147
|
Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target. Mol Neurobiol 2016; 54:7567-7584. [DOI: 10.1007/s12035-016-0245-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
|
148
|
Chi-Castañeda D, Ortega A. Clock Genes in Glia Cells: A Rhythmic History. ASN Neuro 2016; 8:8/5/1759091416670766. [PMID: 27666286 PMCID: PMC5037500 DOI: 10.1177/1759091416670766] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/22/2016] [Indexed: 11/17/2022] Open
Abstract
Circadian rhythms are periodic patterns in biological processes that allow the organisms to anticipate changes in the environment. These rhythms are driven by the suprachiasmatic nucleus (SCN), the master circadian clock in vertebrates. At a molecular level, circadian rhythms are regulated by the so-called clock genes, which oscillate in a periodic manner. The protein products of clock genes are transcription factors that control their own and other genes’ transcription, collectively known as “clock-controlled genes.” Several brain regions other than the SCN express circadian rhythms of clock genes, including the amygdala, the olfactory bulb, the retina, and the cerebellum. Glia cells in these structures are expected to participate in rhythmicity. However, only certain types of glia cells may be called “glial clocks,” since they express PER-based circadian oscillators, which depend of the SCN for their synchronization. This contribution summarizes the current information about clock genes in glia cells, their plausible role as oscillators and their medical implications.
Collapse
Affiliation(s)
- Donají Chi-Castañeda
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México Soluciones para un México Verde, S.A de C.V., Santa Fé Ciudad de México, México
| | - Arturo Ortega
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
149
|
Nakagawa Y, Chiba K. Involvement of Neuroinflammation during Brain Development in Social Cognitive Deficits in Autism Spectrum Disorder and Schizophrenia. J Pharmacol Exp Ther 2016; 358:504-15. [PMID: 27384073 DOI: 10.1124/jpet.116.234476] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/05/2016] [Indexed: 03/08/2025] Open
Abstract
Development of social cognition, a unique and high-order function, depends on brain maturation from childhood to adulthood in humans. Autism spectrum disorder (ASD) and schizophrenia have similar social cognitive deficits, although age of onset in each disorder is different. Pathogenesis of these disorders is complex and contains several features, including genetic risk factors, environmental risk factors, and sites of abnormalities in the brain. Although several hypotheses have been postulated, they seem to be insufficient to explain how brain alterations associated with symptoms in these disorders develop at distinct developmental stages. Development of ASD appears to be related to cerebellar dysfunction and subsequent thalamic hyperactivation in early childhood. By contrast, schizophrenia seems to be triggered by thalamic hyperactivation in late adolescence, whereas hippocampal aberration has been possibly initiated in childhood. One of the possible culprits is metal homeostasis disturbances that can induce dysfunction of blood-cerebrospinal fluid barrier. Thalamic hyperactivation is thought to be induced by microglia-mediated neuroinflammation and abnormalities of intracerebral environment. Consequently, it is likely that the thalamic hyperactivation triggers dysregulation of the dorsolateral prefrontal cortex for lower brain regions related to social cognition. In this review, we summarize the brain aberration in ASD and schizophrenia and provide a possible mechanism underlying social cognitive deficits in these disorders based on their distinct ages of onset.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Innovative Research Division, Mitsubishi Tanabe Pharma, Yokohama, Japan
| | - Kenji Chiba
- Innovative Research Division, Mitsubishi Tanabe Pharma, Yokohama, Japan
| |
Collapse
|
150
|
Ascoli BM, Géa LP, Colombo R, Barbé-Tuana FM, Kapczinski F, Rosa AR. The role of macrophage polarization on bipolar disorder: Identifying new therapeutic targets. Aust N Z J Psychiatry 2016; 50:618-30. [PMID: 27091850 DOI: 10.1177/0004867416642846] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Bipolar disorder is a chronic, severe and disabling disease; however, its pathophysiology remains poorly understood. Recent evidence has suggested that inflammation and immune dysregulation play a significant role in the pathophysiology of bipolar disorder. This review is aimed to highlight the importance of systemic inflammation in modulating the inflammatory response of microglia and hence its potential involvement with bipolar disorder. We also discuss novel therapeutic strategies that emerge from this new research. METHOD This article presents a theoretical synthesis of the effects of systemic inflammation on the immune response of the central nervous system in bipolar disorder. The complex relationship between stress, pro-inflammatory cytokines and microglial dysfunction is summarized, emphasizing the role of the kynurenine pathway in this process and, consequently, their effects on neuronal plasticity. RESULTS Bipolar patients demonstrate increased serum levels of pro-inflammatory cytokines (interleukin-1β, interleukin-6 and tumor necrosis factor-α) and lower hypothalamic-pituitary-adrenal axis sensitivity. This imbalance in the immune system promotes a change in blood-brain barrier permeability, leading to an inflammatory signal spread in the central nervous system from the periphery, through macrophages activation (M1 polarization). Chronic microglial activation can result in neuronal apoptosis, neurogenesis inhibition, hippocampal volume reduction, lower neurotransmitters synthesis and cytotoxicity, by increasing glutamate production and kynurenine metabolism. CONCLUSIONS This review provides an overview of the mechanisms involved in the immune system imbalance and its potential involvement in the pathophysiology of bipolar disorder. Consequently, new strategies that normalize the immune-inflammatory pathways may provide a valuable therapeutic target for the treatment of these disorders.
Collapse
Affiliation(s)
- Bruna M Ascoli
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil Postgraduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luiza P Géa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil Postgraduate Program in Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rafael Colombo
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil Laboratory of Pharmacology and Physiology, Universidade de Caxias do Sul (UCS), Caxias do Sul, Brazil
| | - Florência M Barbé-Tuana
- Laboratory of Molecular Biology and Bioinformatics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil Postgraduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Flávio Kapczinski
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil Postgraduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil Department of Psychiatry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Adriane Ribeiro Rosa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil Postgraduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil Postgraduate Program in Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil Department of Pharmacology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|