1
|
Shin J, Kim DU, Bae GS, Han JY, Lim DW, Lee YM, Kim E, Kwon E, Han D, Kim S. Antidepressant-like Effects of Cannabis sativa L. Extract in an Lipopolysaccharide Model: Modulation of Mast Cell Activation in Deep Cervical Lymph Nodes and Dura Mater. Pharmaceuticals (Basel) 2024; 17:1409. [PMID: 39459047 PMCID: PMC11510560 DOI: 10.3390/ph17101409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS)-induced neuroinflammation is a well-established model for studying depression-like behavior, driven by pro-inflammatory cytokines such as TNF-α and IL-1β. Mast cells (MCs) contribute to neuroinflammation by releasing mediators that exacerbate depressive-like symptoms. This study evaluates the antidepressant-like and anti-inflammatory effects of Cannabis sativa L. inflorescence extract (CSL) in an LPS-induced neuroinflammation model. METHODS Male C57BL/6 mice were intraperitoneally injected with CSL at doses of 10, 20, and 30 mg/kg, 30 min prior to LPS (0.83 mg/kg) administration. Depressive behaviors were assessed using the sucrose preference test (SPT), tail suspension test (TST), and forced swimming test (FST). The neutrophil-to-lymphocyte ratio (NLR) was measured to assess systemic inflammation. Cytokine levels in the prefrontal cortex (PFC) were measured, and mast cell degranulation in the lymph nodes and dura mater was analyzed histologically (approval number: WKU24-64). RESULTS CSL significantly improved depressive-like behaviors and decreased the NLR, indicating reduced systemic inflammation. CSL also significantly reduced TNF-α and IL-1β levels in the PFC. Furthermore, CSL inhibited MC degranulation in the deep cervical lymph nodes and dura mater, with the strongest effects observed at 30 mg/kg. CONCLUSIONS CSL demonstrated antidepressant-like and anti-inflammatory effects in an LPS-induced neuroinflammation model, likely through the modulation of cytokine expression and mast cell activity. These results suggest the potential of CSL as a therapeutic option for treating inflammation-related depression.
Collapse
Affiliation(s)
- Joonyoung Shin
- Institute for Global Rare Disease Network, Professional Graduate School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (J.S.); (D.H.)
| | - Dong-Uk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (G.-S.B.)
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (D.-U.K.); (G.-S.B.)
| | - Ji-Ye Han
- Department of Oriental Pharmacy, Wonkwang-Oriental Medicines Research Institute, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea; (J.-Y.H.); (D.-W.L.); (Y.-M.L.)
| | - Do-Won Lim
- Department of Oriental Pharmacy, Wonkwang-Oriental Medicines Research Institute, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea; (J.-Y.H.); (D.-W.L.); (Y.-M.L.)
| | - Young-Mi Lee
- Department of Oriental Pharmacy, Wonkwang-Oriental Medicines Research Institute, College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea; (J.-Y.H.); (D.-W.L.); (Y.-M.L.)
| | - Eunjae Kim
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (E.K.); (E.K.)
| | - Eunjeong Kwon
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (E.K.); (E.K.)
| | - Dongwoon Han
- Institute for Global Rare Disease Network, Professional Graduate School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (J.S.); (D.H.)
- Department of Global Health and Development, Hanyang University, Seoul 04763, Republic of Korea
| | - Sungchul Kim
- Institute for Global Rare Disease Network, Professional Graduate School of Korean Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (J.S.); (D.H.)
| |
Collapse
|
2
|
Tarkany Basting R, Henrique Napimoga M, Antônio Trindade Silva C, Ballassini Abdalla H, Campos Durso B, Henrique Barboza Martins L, de Abreu Cavalcanti H, Hammock BD, Trindade Clemente-Napimoga J. Soluble epoxide hydrolase inhibitor blockage microglial cell activation in subnucleus caudalis in a persistent model of arthritis. Int Immunopharmacol 2023; 120:110320. [PMID: 37230034 PMCID: PMC10631565 DOI: 10.1016/j.intimp.2023.110320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/30/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic condition characterized by pain and infiltration of immune cells into the joint. Immune cells can be activated, producing inflammatory cytokines, leading to continuously degenerative and inflammatory reactions and the temporomandibular joint (TMJ) can be affected by RA. In this scenario, novel targets are needed to increase treatment efficacy with minimized side effects. The epoxy-eicosatrienoic acids (EETs), are endogenous signaling molecules, playing important roles in diminishing inflammation and pain but are promptly metabolized by soluble epoxide hydrolase (sEH), generating less-bioactive acids.Therefore, sEH inhibitors is an interest therapeutic target to enhance the beneficial effect of natural EETs. TPPU is a potent sEH inhibitor that is capable of dampening EETs hydrolysis. Thus, we aimed to assess the impact of pharmacological sEH inhibition on a persistent model of albumin-induced arthritis in the TMJ, in two scenarios: first, as post-treatment, in an installed arthritic condition, and second, the protective role, in preventing the development of an arthritic condition. In addition, we investigate the influence of sEH inhibition on microglia cell activation in the trigeminal subnucleus caudalis (TSC) and in vitro experiments. Finally, we examined the astrocyte phenotype. Oral administration of TPPU, acts in multiple pathways, in a protective and reparative post-treatment, ameliorating the preservation of the TMJ morphology, reducing the hypernociception, with an immunosuppressive action reducing neutrophil and lymphocytes and pro-inflammatory cytokines in the TMJ of rats. In TSC, TPPU reduces the cytokine storm and attenuates the microglia activated P2X7/Cathepsin S/Fractalkine pathway and reduces the astrocyte activation and glutamate levels. Collectively, our findings revealed that sEH inhibition mitigates hypersensitive nociception through the regulation of microglia activation and astrocyte modulation, demonstrating the potential use of sEH inhibitors as immunoresolvents in the treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Rosanna Tarkany Basting
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Marcelo Henrique Napimoga
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Carlos Antônio Trindade Silva
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Henrique Ballassini Abdalla
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Braz Campos Durso
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | | | - Herbert de Abreu Cavalcanti
- São Leopoldo Mandic Institute and Research Center, Laboratory of Neuroimmune Interface of Pain Research, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, CA, United States of America; EicOsis LLC, Davis, CA, United States of America
| | | |
Collapse
|
3
|
The Neuroinflammatory Role of Pericytes in Epilepsy. Biomedicines 2021; 9:biomedicines9070759. [PMID: 34209145 PMCID: PMC8301485 DOI: 10.3390/biomedicines9070759] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Pericytes are a component of the blood-brain barrier (BBB) neurovascular unit, in which they play a crucial role in BBB integrity and are also implicated in neuroinflammation. The association between pericytes, BBB dysfunction, and the pathophysiology of epilepsy has been investigated, and links between epilepsy and pericytes have been identified. Here, we review current knowledge about the role of pericytes in epilepsy. Clinical evidence has shown an accumulation of pericytes with altered morphology in the cerebral vascular territories of patients with intractable epilepsy. In vitro, proinflammatory cytokines, including IL-1β, TNFα, and IL-6, cause morphological changes in human-derived pericytes, where IL-6 leads to cell damage. Experimental studies using epileptic animal models have shown that cerebrovascular pericytes undergo redistribution and remodeling, potentially contributing to BBB permeability. These series of pericyte-related modifications are promoted by proinflammatory cytokines, of which the most pronounced alterations are caused by IL-1β, a cytokine involved in the pathogenesis of epilepsy. Furthermore, the pericyte-glial scarring process in leaky capillaries was detected in the hippocampus during seizure progression. In addition, pericytes respond more sensitively to proinflammatory cytokines than microglia and can also activate microglia. Thus, pericytes may function as sensors of the inflammatory response. Finally, both in vitro and in vivo studies have highlighted the potential of pericytes as a therapeutic target for seizure disorders.
Collapse
|
4
|
Une H, Yamasaki R, Nagata S, Yamaguchi H, Nakamuta Y, Indiasari UC, Cui Y, Shinoda K, Masaki K, Götz M, Kira JI. Brain gray matter astroglia-specific connexin 43 ablation attenuates spinal cord inflammatory demyelination. J Neuroinflammation 2021; 18:126. [PMID: 34090477 PMCID: PMC8180177 DOI: 10.1186/s12974-021-02176-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Brain astroglia are activated preceding the onset of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We characterized the effects of brain astroglia on spinal cord inflammation, focusing on astroglial connexin (Cx)43, because we recently reported that Cx43 has a critical role in regulating neuroinflammation. Methods Because glutamate aspartate transporter (GLAST)+ astroglia are enriched in the brain gray matter, we generated Cx43fl/fl;GLAST-CreERT2/+ mice that were brain gray matter astroglia-specific Cx43 conditional knockouts (Cx43 icKO). EAE was induced by immunization with myelin oligodendroglia glycoprotein (MOG) 35–55 peptide 10 days after tamoxifen injection. Cx43fl/fl mice were used as controls. Results Acute and chronic EAE signs were significantly milder in Cx43 icKO mice than in controls whereas splenocyte MOG-specific responses were unaltered. Histologically, Cx43 icKO mice showed significantly less demyelination and fewer CD45+ infiltrating immunocytes, including F4/80+ macrophages, and Iba1+ microglia in the spinal cord than controls. Microarray analysis of the whole cerebellum revealed marked upregulation of anti-inflammatory A2-specific astroglia gene sets in the pre-immunized phase and decreased proinflammatory A1-specific and pan-reactive astroglial gene expression in the onset phase in Cx43 icKO mice compared with controls. Astroglia expressing C3, a representative A1 marker, were significantly decreased in the cerebrum, cerebellum, and spinal cord of Cx43 icKO mice compared with controls in the peak phase. Isolated Cx43 icKO spinal microglia showed more anti-inflammatory and less proinflammatory gene expression than control microglia in the pre-immunized phase. In particular, microglial expression of Ccl2, Ccl5, Ccl7, and Ccl8 in the pre-immunized phase and of Cxcl9 at the peak phase was lower in Cx43 icKO than in controls. Spinal microglia circularity was significantly lower in Cx43 icKO than in controls in the peak phase. Significantly lower interleukin (IL)-6, interferon-γ, and IL-10 levels were present in cerebrospinal fluid from Cx43 icKO mice in the onset phase compared with controls. Conclusions The ablation of Cx43 in brain gray matter astroglia attenuates EAE by promoting astroglia toward an anti-inflammatory phenotype and suppressing proinflammatory activation of spinal microglia partly through depressed cerebrospinal fluid proinflammatory cytokine/chemokine levels. Brain astroglial Cx43 might be a novel therapeutic target for MS. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02176-1.
Collapse
Affiliation(s)
- Hayato Une
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuko Nakamuta
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ulfa Camelia Indiasari
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yiwen Cui
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shinoda
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Ookawa, Fukuoka, 831-8501, Japan. .,Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 2-6-11 Yakuin, Chuo-ku, Fukuoka, 810-0022, Japan.
| |
Collapse
|
5
|
Yegla B, Boles J, Kumar A, Foster TC. Partial microglial depletion is associated with impaired hippocampal synaptic and cognitive function in young and aged rats. Glia 2021; 69:1494-1514. [PMID: 33586813 DOI: 10.1002/glia.23975] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
The role of microglia in mediating age-related changes in cognition and hippocampal synaptic function was examined by microglial depletion and replenishment using PLX3397. We observed age-related differences in microglial number and morphology, as well as increased Iba-1 expression, indicating microglial activation. PLX3397 treatment decreased microglial number, with aged rats exhibiting the lowest density. Young rats exhibited increased expression of pro-inflammatory cytokines during depletion and repopulation and maintenance of Iba-1 levels despite reduced microglial number. For aged rats, several cytokines increased with depletion and recovered during repopulation; however, aged rats did not fully recover microglial cell number or Iba-1 expression during repopulation, with a recovery comparable to young control levels rather than aged controls. Hippocampal CA3-CA1 synaptic transmission was impaired with age, and microglial depletion was associated with decreased total synaptic transmission in young and aged rats. A robust decline in N-methyl-d-aspartate-receptor-mediated synaptic transmission arose in young depleted rats specifically. Microglial replenishment normalized depletion-induced synaptic function to control levels; however, recovery of aged animals did not mirror young. Microglial depletion was associated with decreased context-object discrimination memory in both age groups, which recovered with microglial repopulation. Aged rats displayed impaired contextual and cued fear memory, and microglial replenishment did not recover their memory to the level of young. The current study indicates that cognitive function and synaptic transmission benefit from the support of aged microglia and are hindered by removal of these cells. Replenishment of microglia in aging did not ameliorate age-related cognitive impairments or senescent synaptic function.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Jake Boles
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics Program, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
6
|
Florensa-Zanuy E, Garro-Martínez E, Adell A, Castro E, Díaz Á, Pazos Á, Mac-Dowell KS, Martín-Hernández D, Pilar-Cuéllar F. Cannabidiol antidepressant-like effect in the lipopolysaccharide model in mice: Modulation of inflammatory pathways. Biochem Pharmacol 2021; 185:114433. [PMID: 33513342 DOI: 10.1016/j.bcp.2021.114433] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 12/12/2022]
Abstract
Major Depression is a severe psychiatric condition with a still poorly understood etiology. In the last years, evidence supporting the neuroinflammatory hypothesis of depression has increased. In the current clinical scenario, in which the available treatments for depression is far from optimal, there is an urgent need to develop fast-acting drugs with fewer side effects. In this regard, recent pieces of evidence suggest that cannabidiol (CBD), the major non-psychotropic component of Cannabis sativa with anti-inflammatory properties, appears as a drug with antidepressant properties. In this work, CBD 30 mg/kg was administered systemically to mice 30 min before lipopolysaccharide (LPS; 0.83 mg/kg) administration as a neuroinflammatory model, and behavioral tests for depressive-, anhedonic- and anxious-like behavior were performed. NF-ĸB, IκBα and PPARγ levels were analyzed by western blot in nuclear and cytosolic fractions of cortical samples. IL-6 and TNFα levels were determined in plasma and prefrontal cortex using ELISA and qPCR techniques, respectively. The precursor tryptophan (TRP), and its metabolites kynurenine (KYN) and serotonin (5-HT) were measured in hippocampus and cortex by HPLC. The ratios KYN/TRP and KYN/5-HT were used to estimate indoleamine 2,3-dioxygenase (IDO) activity and the balance of both metabolic pathways, respectively. CBD reduced the immobility time in the tail suspension test and increased sucrose preference in the LPS model, without affecting locomotion and central activity in the open-field test. CBD diminished cortical NF-ĸB activation, IL-6 levels in plasma and brain, and the increased KYN/TRP and KYN/5-HT ratios in hippocampus and cortex in the LPS model. Our results demonstrate that CBD produced antidepressant-like effects in the LPS neuroinflammatory model, associated to a reduction in the kynurenine pathway activation, IL-6 levels and NF-ĸB activation. As CBD stands out as a promising antidepressant drug, more research is needed to completely understand its mechanisms of action in depression linked to inflammation.
Collapse
Affiliation(s)
- Eva Florensa-Zanuy
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Emilio Garro-Martínez
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Albert Adell
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Elena Castro
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Álvaro Díaz
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Ángel Pazos
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain
| | - Karina S Mac-Dowell
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain; Departmento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid (UCM), IUIN-UCM, Imas12 Hospital 12 de Octubre, Madrid, Spain
| | - David Martín-Hernández
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain; Department of Child and Adolescent Psychiatry, Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Fuencisla Pilar-Cuéllar
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Spain.
| |
Collapse
|
7
|
Lively S, Schlichter LC. Microglia Responses to Pro-inflammatory Stimuli (LPS, IFNγ+TNFα) and Reprogramming by Resolving Cytokines (IL-4, IL-10). Front Cell Neurosci 2018; 12:215. [PMID: 30087595 PMCID: PMC6066613 DOI: 10.3389/fncel.2018.00215] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Microglia respond to CNS injuries and diseases with complex reactions, often called "activation." A pro-inflammatory phenotype (also called classical or M1 activation) lies at one extreme of the reactivity spectrum. There were several motivations for this study. First, bacterial endotoxin (lipopolysaccharide, LPS) is the most commonly used pro-inflammatory stimulus for microglia, both in vitro and in vivo; however, pro-inflammatory cytokines (e.g., IFNγ, TNFα) rather than LPS will be encountered with sterile CNS damage and disease. We lack direct comparisons of responses between LPS and such cytokines. Second, while transcriptional profiling is providing substantial data on microglial responses to LPS, these studies mainly use mouse cells and models, and there is increasing evidence that responses of rat microglia can differ. Third, the cytokine milieu is dynamic after acute CNS damage, and an important question in microglial biology is: How malleable are their responses? There are very few studies of effects of resolving cytokines, particularly for rat microglia, and much of the work has focused on pro-inflammatory outcomes. Here, we first exposed primary rat microglia to LPS or to IFNγ+TNFα (I+T) and compared hallmark functional (nitric oxide production, migration) and molecular responses (almost 100 genes), including surface receptors that can be considered part of the sensome. Protein changes for exemplary molecules were also quantified: ARG1, CD206/MRC1, COX-2, iNOS, and PYK2. Despite some similarities, there were notable differences in responses to LPS and I+T. For instance, LPS often evoked higher pro-inflammatory gene expression and also increased several anti-inflammatory genes. Second, we compared the ability of two anti-inflammatory, resolving cytokines (IL-4, IL-10), to counteract responses to LPS and I+T. IL-4 was more effective after I+T than after LPS, and IL-10 was surprisingly ineffective after either stimulus. These results should prove useful in modeling microglial reactivity in vitro; and comparing transcriptional responses to sterile CNS inflammation in vivo.
Collapse
Affiliation(s)
- Starlee Lively
- Division of Genetics & Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Lyanne C Schlichter
- Division of Genetics & Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Park J, Wetzel I, Marriott I, Dréau D, D'Avanzo C, Kim DY, Tanzi RE, Cho H. A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer's disease. Nat Neurosci 2018; 21:941-951. [PMID: 29950669 PMCID: PMC6800152 DOI: 10.1038/s41593-018-0175-4] [Citation(s) in RCA: 412] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/25/2018] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease (AD) is characterized by beta-amyloid accumulation, phosphorylated tau formation, hyperactivation of glial cells, and neuronal loss. The mechanisms of AD pathogenesis, however, remain poorly understood, partially due to the lack of relevant models that can comprehensively recapitulate multistage intercellular interactions in human AD brains. Here we present a new three-dimensional (3D) human AD triculture model using neurons, astrocytes, and microglia in a 3D microfluidic platform. Our model provided key representative AD features: beta-amyloid aggregation, phosphorylated tau accumulation, and neuroinflammatory activity. In particular, the model mirrored microglial recruitment, neurotoxic activities such as axonal cleavage, and NO release damaging AD neurons and astrocytes. Our model will serve to facilitate the development of more precise human brain models for basic mechanistic studies in neural-glial interactions and drug discovery.
Collapse
Affiliation(s)
- Joseph Park
- Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC, USA.,Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA.,The Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte, NC, USA.,Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Isaac Wetzel
- Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC, USA.,Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA.,The Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Ian Marriott
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Didier Dréau
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA.,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Carla D'Avanzo
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hansang Cho
- Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC, USA. .,Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA. .,Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA. .,The Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
9
|
Quagliato LA, Nardi AE. Cytokine alterations in panic disorder: A systematic review. J Affect Disord 2018; 228:91-96. [PMID: 29241050 DOI: 10.1016/j.jad.2017.11.094] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/01/2017] [Accepted: 11/12/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Panic disorder (PD) occurs in 3.4-4.7% of the general population. Although accumulating evidence suggests that some inflammatory processes play a role in the pathophysiology of mental disorders, very few studies have evaluated cytokine levels in patients with PD. The aim of the present study was to systematically review the characteristic cytokine profile of PD patients and discuss some possibilities for future trials on this common and disabling disorder. METHODS A comprehensive literature search was carried out in PubMed and Web of Science databases (search terms: "panic disorder" or "panic attacks" and IL-1, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, IL-13, TNF-alpha and INF-gamma). RESULTS Eleven studies involving measurements of cytokines in PD patients were included in this review article. Increased serum levels of some inflammatory markers such as IL-6, IL-1β and IL-5 were reported in PD patients compared with control subjects. There are some conflicting results regarding IL-2, IL-12, and INF-γ in association with PD. LIMITATIONS There are discrepant findings in the existing literature regarding PD and cytokines. A significant portion of the recognized heterogeneity may be attributable to variability in assay procedures. The discrepant findings may also have been due to differences in the study populations. CONCLUSIONS Cytokines induce the production of acute-phase proteins and are linked to neurogenesis, modification of the HPA axis, microglial activation, tryptophan metabolism and an imbalance in excitatory and inhibitory neurotransmission. Investigation of inflammatory biomarkers in PD could contribute to understanding the pathophysiological mechanisms in this debilitating disorder.
Collapse
Affiliation(s)
- Laiana Azevedo Quagliato
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Antonio E Nardi
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Brain interference: Revisiting the role of IFNγ in the central nervous system. Prog Neurobiol 2017; 156:149-163. [DOI: 10.1016/j.pneurobio.2017.05.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 01/28/2023]
|
11
|
Clarkson BDS, Kahoud RJ, McCarthy CB, Howe CL. Inflammatory cytokine-induced changes in neural network activity measured by waveform analysis of high-content calcium imaging in murine cortical neurons. Sci Rep 2017; 7:9037. [PMID: 28831096 PMCID: PMC5567248 DOI: 10.1038/s41598-017-09182-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/20/2017] [Indexed: 01/07/2023] Open
Abstract
During acute neuroinflammation, increased levels of cytokines within the brain may contribute to synaptic reorganization that results in long-term changes in network hyperexcitability. Indeed, inflammatory cytokines are implicated in synaptic dysfunction in epilepsy and in an array of degenerative and autoimmune diseases of the central nervous system. Current tools for studying the impact of inflammatory factors on neural networks are either insufficiently fast and sensitive or require complicated and costly experimental rigs. Calcium imaging offers a reasonable surrogate for direct measurement of neuronal network activity, but traditional imaging paradigms are confounded by cellular heterogeneity and cannot readily distinguish between glial and neuronal calcium transients. While the establishment of pure neuron cultures is possible, the removal of glial cells ignores physiologically relevant cell-cell interactions that may be critical for circuit level disruptions induced by inflammatory factors. To overcome these issues, we provide techniques and algorithms for image processing and waveform feature extraction using automated analysis of spontaneous and evoked calcium transients in primary murine cortical neuron cultures transduced with an adeno-associated viral vector driving the GCaMP6f reporter behind a synapsin promoter. Using this system, we provide evidence of network perturbations induced by the inflammatory cytokines TNFα, IL1β, and IFNγ.
Collapse
Affiliation(s)
| | - Robert J Kahoud
- Department of Neurology, Mayo Clinic, Rochester, MN, USA 55905, USA
- Department of Pediatrics, Mayo Clinic, Rochester, MN, USA 55905, USA
| | | | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, USA 55905, USA.
- Department of Neuroscience, Mayo Clinic, Rochester, MN, USA 55905, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA 55905, USA.
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA 55905, USA.
| |
Collapse
|
12
|
Jansson D, Scotter EL, Rustenhoven J, Coppieters N, Smyth LCD, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RLM, Dragunow M. Interferon-γ blocks signalling through PDGFRβ in human brain pericytes. J Neuroinflammation 2016; 13:249. [PMID: 27654972 PMCID: PMC5031293 DOI: 10.1186/s12974-016-0722-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Neuroinflammation and blood-brain barrier (BBB) disruption are common features of many brain disorders, including Alzheimer's disease, epilepsy, and motor neuron disease. Inflammation is thought to be a driver of BBB breakdown, but the underlying mechanisms for this are unclear. Brain pericytes are critical cells for maintaining the BBB and are immunologically active. We sought to test the hypothesis that inflammation regulates the BBB by altering pericyte biology. METHODS We exposed primary adult human brain pericytes to chronic interferon-gamma (IFNγ) for 4 days and measured associated functional aspects of pericyte biology. Specifically, we examined the influence of inflammation on platelet-derived growth factor receptor-beta (PDGFRβ) expression and signalling, as well as pericyte proliferation and migration by qRT-PCR, immunocytochemistry, flow cytometry, and western blotting. RESULTS Chronic IFNγ treatment had marked effects on pericyte biology most notably through the PDGFRβ, by enhancing agonist (PDGF-BB)-induced receptor phosphorylation, internalization, and subsequent degradation. Functionally, chronic IFNγ prevented PDGF-BB-mediated pericyte proliferation and migration. CONCLUSIONS Because PDGFRβ is critical for pericyte function and its removal leads to BBB leakage, our results pinpoint a mechanism linking chronic brain inflammation to BBB dysfunction.
Collapse
Affiliation(s)
- Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Natacha Coppieters
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | | | - Peter S Bergin
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - E Scott Graham
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand. .,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand. .,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, 1142, Auckland, New Zealand.
| |
Collapse
|
13
|
Allen Reish HE, Standaert DG. Role of α-synuclein in inducing innate and adaptive immunity in Parkinson disease. JOURNAL OF PARKINSON'S DISEASE 2015; 5:1-19. [PMID: 25588354 PMCID: PMC4405142 DOI: 10.3233/jpd-140491] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alpha-synuclein (α-syn) is central to the pathogenesis of Parkinson disease (PD). Gene duplications, triplications and point mutations in SNCA1, the gene encoding α-syn, cause autosomal dominant forms of PD. Aggregated and post-translationally modified forms of α-syn are present in Lewy bodies and Lewy neurites in both sporadic and familial PD, and recent work has emphasized the prion-like ability of aggregated α-syn to produce spreading pathology. Accumulation of abnormal forms of α-syn is a trigger for PD, but recent evidence suggests that much of the downstream neurodegeneration may result from inflammatory responses. Components of both the innate and adaptive immune systems are activated in PD, and influencing interactions between innate and adaptive immune components has been shown to modify the pathological process in animal models of PD. Understanding the relationship between α-syn and subsequent inflammation may reveal novel targets for neuroprotective interventions. In this review, we examine the role of α-syn and modified forms of this protein in the initiation of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Heather E Allen Reish
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Alabama, USA
| | - David G Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
14
|
Su Y, Fan W, Ma Z, Wen X, Wang W, Wu Q, Huang H. Taurine improves functional and histological outcomes and reduces inflammation in traumatic brain injury. Neuroscience 2014; 266:56-65. [PMID: 24530657 DOI: 10.1016/j.neuroscience.2014.02.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 01/13/2014] [Accepted: 02/05/2014] [Indexed: 01/17/2023]
Abstract
We investigated the effect of taurine on inflammatory cytokine expression, on astrocyte activity and cerebral edema and functional outcomes, following traumatic brain injury (TBI) in rats. 72 rats were randomly divided into sham, TBI and Taurine groups. Rats subjected to moderate lateral fluid percussion injury were injected intravenously with taurine (200mg/kg) or saline immediately after injury or daily for 7days. Functional outcome was evaluated using Modified Neurological Severity Score (mNSS). Glial fibrillary acidic protein (GFAP) of the brain was measured using immunofluorescence. Concentration of 23 cytokines and chemokines in the injured cortex at 1 and 7days after TBI was assessed by Luminex xMAP technology. The results showed that taurine significantly improved functional recovery except 1day, reduced accumulation of GFAP and water content in the penumbral region at 7days after TBI. Compared with the TBI group, taurine significantly suppressed growth-related oncogene (GRO/KC) and interleukin (IL)-1β levels while elevating the levels of regulated on activation, normal T cell expressed and secreted (RANTES) at 1day. And taurine markedly decreased the level of 17 cytokine: eotaxin, Granulocyte colony-stimulating factor (G-CSF), Granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon-gamma (IFN-γ), IL-1α, IL-1β, IL-4, IL-5, IL-6, IL-10, IL-12p70, IL-13, IL-17, leptin, monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), vascular endothelial growth factor (VEGF), and only increased the level of MIP-1α in a week. The results suggest that taurine effectively mitigates the severity of brain damage in TBI by attenuating the increase of astrocyte activity and edema as well as pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Y Su
- The Graduate School, Tianjin Medical University, Tianjin 300070, PR China
| | - W Fan
- Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300060, PR China
| | - Z Ma
- Baoding NO. 1 Hospital, Baoding, Hebei 071000, PR China
| | - X Wen
- The Graduate School, Tianjin Medical University, Tianjin 300070, PR China
| | - W Wang
- Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300060, PR China
| | - Q Wu
- Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300060, PR China
| | - H Huang
- Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300060, PR China.
| |
Collapse
|
15
|
Classical activation of microglia in CD200-deficient mice is a consequence of blood brain barrier permeability and infiltration of peripheral cells. Brain Behav Immun 2013; 34:86-97. [PMID: 23916893 DOI: 10.1016/j.bbi.2013.07.174] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 01/08/2023] Open
Abstract
The interaction between CD200, expressed on several cell types, and its receptor CD200R, expressed on cells of the myeloid lineage, has been shown to be an important factor in modulating inflammation in macrophage function in several conditions including colitis and arthritis. More recently its modulatory effect on microglial activation has been identified and CD200-deficiency has been associated with increased microglial activation accompanied by increased production of inflammatory cytokines. The response of glia prepared from CD200-deficient mice to stimuli like lipopolysaccharide (LPS) is markedly greater than the response of cells prepared from wildtype mice and, consistent with this, is the recent observation that expression of Toll-like receptor (TLR)4 and signalling through NFκB are increased in microglia prepared from CD200-deficient mice. Here we show that glia from CD200-deficient mice are also more responsive to interferon-γ (IFNγ) which triggers classical activation of microglia. We investigated the effects of CD200-deficiency in vivo and report that there is an increase in expression of several markers of microglial activation including tumor necrosis factor (TNF)-α, which is a hallmark of classically-activated microglia. These changes are accompanied by increased IFNγ, and the evidence suggests that this is produced by infiltrating cells including T cells and macrophages. We propose that these cells enter the brain as a consequence of increased blood brain barrier (BBB) permeability in CD200-deficient mice and that infiltration is assisted by increased expression of the chemokines, monocyte chemotactic protein-1 (MCP-1), IFNγ-induced protein-10 (IP-10) and RANTES. This may have implications in neurodegenerative diseases where BBB permeability is compromised.
Collapse
|
16
|
Swardfager W, Lanctôt K, Rothenburg L, Wong A, Cappell J, Herrmann N. A meta-analysis of cytokines in Alzheimer's disease. Biol Psychiatry 2010; 68:930-41. [PMID: 20692646 DOI: 10.1016/j.biopsych.2010.06.012] [Citation(s) in RCA: 702] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 06/03/2010] [Accepted: 06/08/2010] [Indexed: 12/27/2022]
Abstract
BACKGROUND Studies suggest that inflammation is involved in the neurodegenerative cascade leading to Alzheimer's disease (AD) pathology and symptoms. This study sought to quantitatively summarize the clinical cytokine data. METHODS Original English language peer-reviewed studies measuring cytokine concentrations in AD and healthy control subjects were included. Mean (± standard deviation) cytokine concentrations for AD and control subjects were extracted. RESULTS Forty studies measuring peripheral blood cytokine concentrations and 14 measuring cerebrospinal fluid (CSF) cytokine concentrations were included. In peripheral blood, there were significantly higher concentrations (weighted mean difference [95% confidence interval]) of interleukin (IL)-6 (2.86 [1.68, 4.04] pg/mL, p < .00001, N[AD/control subjects] = 985/680, 14 studies), tumor necrosis factor (TNF)-α (3.25 [.76, 5.74] pg/mL, p = .01, N = 680/447, 14 studies), IL-1β (.55 [.32, .78] pg/mL, p < .00001, N = 574/370, 10 studies), transforming growth factor (TGF)-β (67.23 [28.62, 105.83] pg/mL, p = .0006, N = 190/158, 5 studies), IL-12 (7.60 [5.58, 9.62] pg/mL, p < .00001, N = 148/106, 5 studies), and IL-18 (15.82 [1.98, 29.66] pg/mL, p = .03, N = 131/94, 4 studies) but not of IL-4, IL-8, IL-10, interferon-γ, or C-reactive protein in AD subjects compared with control subjects. There were significantly higher concentrations of TGF-β (7.81 [2.27, 13.35] pg/mL, p =.006, N = 113/114, 5 studies) but not IL-6, TNF-α, and IL-1β in the CSF of AD subjects compared with control subjects. CONCLUSIONS These results strengthen the clinical evidence that AD is accompanied by an inflammatory response, particularly higher peripheral concentrations of IL-6, TNF-α, IL-1β, TGF-β, IL-12 and IL-18 and higher CSF concentrations of TGF-β.
Collapse
Affiliation(s)
- Walter Swardfager
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
17
|
A meta-analysis of cytokines in major depression. Biol Psychiatry 2010; 67:446-57. [PMID: 20015486 DOI: 10.1016/j.biopsych.2009.09.033] [Citation(s) in RCA: 3290] [Impact Index Per Article: 219.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 08/31/2009] [Accepted: 09/26/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND Major depression occurs in 4.4% to 20% of the general population. Studies suggest that major depression is accompanied by immune dysregulation and activation of the inflammatory response system (IRS). Our objective was to quantitatively summarize the data on concentrations of specific cytokines in patients diagnosed with a major depressive episode and controls. METHODS We performed a meta-analysis of studies measuring cytokine concentration in patients with major depression, with a database search of the English literature (to August 2009) and a manual search of references. RESULTS Twenty-four studies involving unstimulated measurements of cytokines in patients meeting DSM criteria for major depression were included in the meta-analysis; 13 for tumor necrosis factor (TNF)-alpha, 9 for interleukin (IL)-1beta, 16 for IL-6, 5 for IL-4, 5 for IL-2, 4 for IL-8, 6 for IL-10, and 4 for interferon (IFN)-gamma. There were significantly higher concentrations of TNF-alpha (p < .00001), weighted mean difference (WMD) (95% confidence interval) 3.97 pg/mL (2.24 to 5.71), in depressed subjects compared with control subjects (438 depressed/350 nondepressed). Also, IL-6 concentrations were significantly higher (p < .00001) in depressed subjects compared with control subjects (492 depressed/400 nondepressed) with an overall WMD of 1.78 pg/mL (1.23 to 2.33). There were no significant differences among depressed and nondepressed subjects for the other cytokines studied. CONCLUSIONS This meta-analysis reports significantly higher concentrations of the proinflammatory cytokines TNF-alpha and IL-6 in depressed subjects compared with control subjects. While both positive and negative results have been reported in individual studies, this meta-analytic result strengthens evidence that depression is accompanied by activation of the IRS.
Collapse
|
18
|
Chen K, Iribarren P, Huang J, Zhang L, Gong W, Cho EH, Lockett S, Dunlop NM, Wang JM. Induction of the formyl peptide receptor 2 in microglia by IFN-gamma and synergy with CD40 ligand. THE JOURNAL OF IMMUNOLOGY 2007; 178:1759-66. [PMID: 17237425 DOI: 10.4049/jimmunol.178.3.1759] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human formyl peptide receptor (FPR)-like 1 (FPRL1) and its mouse homologue mFPR2 are functional receptors for a variety of exogenous and host-derived chemotactic peptides, including amyloid beta 1-42 (Abeta(42)), a pathogenic factor in Alzheimer's disease. Because mFPR2 in microglial cells is regulated by proinflammatory stimulants including TLR agonists, in this study we investigated the capacity of IFN-gamma and the CD40 ligand (CD40L) to affect the expression and function of mFPR2. We found that IFN-gamma, when used alone, induced mFPR2 mRNA expression in a mouse microglial cell line and primary microglial cells in association with increased cell migration in response to mFPR2 agonists, including Abeta(42). IFN-gamma also increased the endocytosis of Abeta(42) by microglial cells via mFPR2. The effect of IFN-gamma on mFPR2 expression in microglial cells was dependent on activation of MAPK and IkappaB-alpha. IFN-gamma additionally increased the expression of CD40 by microglial cells and soluble CD40L significantly promoted cell responses to IFN-gamma during a 6-h incubation period by enhancing the activation of MAPK and IkappaB-alpha signaling pathways. We additionally found that the effect of IFN-gamma and its synergy with CD40L on mFPR2 expression in microglia was mediated in part by TNF-alpha. Our results suggest that IFN-gamma and CD40L, two host-derived factors with increased concentrations in inflammatory central nervous system diseases, may profoundly affect microglial cell responses in the pathogenic process in which mFPR2 agonist peptides are elevated.
Collapse
Affiliation(s)
- Keqiang Chen
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kawanokuchi J, Mizuno T, Takeuchi H, Kato H, Wang J, Mitsuma N, Suzumura A. Production of interferon-gamma by microglia. Mult Scler 2006; 12:558-64. [PMID: 17086900 DOI: 10.1177/1352458506070763] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neural cells do not usually interact with immune cells because of the lack of major histocompatibility complex (MHC) antigen expression. Interferon-gamma (IFN-gamma) enables this interaction via induction of MHC antigen expression in neural cells. Thus, IFN-gamma is a critical cytokine for the development of central nervous system (CNS) pathologies. IFN-gamma, however, is considered to be produced exclusively by lymphoid cells. Here, we show for the first time that murine microglia produce IFN-gamma in response to IL-12 and/or IL-18, using RT-PCR detection of IFN-gamma mRNA and Western blotting and immunohistochemical analysis for cytoplasmic expression of IFN-gamma. Stimulation of microglia with IL-12 and IL-18 resulted in MHC class II mRNA expression in microglia. Since IL-12 and IL-18 are produced in the CNS by glial cells, these cytokines may play a critical role in the initiation of neural-immune cell interaction and the induction of autoimmune processes in the CNS via induction of IFN-gamma and MHC antigens.
Collapse
Affiliation(s)
- J Kawanokuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusaku, Nagoya 464-8601, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Miyatake Y, Ikeda H, Ishizu A, Baba T, Ichihashi T, Suzuki A, Tomaru U, Kasahara M, Yoshiki T. Role of neuronal interferon-gamma in the development of myelopathy in rats infected with human T-cell leukemia virus type 1. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:189-99. [PMID: 16816372 PMCID: PMC1698768 DOI: 10.2353/ajpath.2006.051225] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of not only adult T-cell leukemia but also HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Among the rat strains infected with HTLV-1, chronic progressive myelopathy, named HAM rat disease, occurs exclusively in WKAH rats. In the present study, we found that HTLV-1 infection induces interferon (IFN)-gamma production in the spinal cords of HAM-resistant strains but not in those of WKAH rats. Neurons were the major cells that produced IFN-gamma in HTLV-1-infected, HAM-resistant strains. Administration of IFN-gamma suppressed expression of pX, the gene critically involved in the onset of HAM rat disease, in an HTLV-1-immortalized rat T-cell line, indicating that IFN-gamma protects against the development of HAM rat disease. The inability of WKAH spinal cord neurons to produce IFN-gamma after infection appeared to stem from defects in signaling through the interleukin (IL)-12 receptor. Specifically, WKAH-derived spinal cord cells were unable to up-regulate the IL-12 receptor beta2 gene in response to IL-12 stimulation. We suggest that the failure of spinal cord neurons to produce IFN-gamma through the IL-12 pathway is involved in the development of HAM rat disease.
Collapse
Affiliation(s)
- Yukiko Miyatake
- Department of Pathology/Pathophysiology, Division of Pathophysiological Science, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dafny N, Yang PB. Interferon and the central nervous system. Eur J Pharmacol 2005; 523:1-15. [PMID: 16226745 DOI: 10.1016/j.ejphar.2005.08.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Revised: 07/01/2005] [Accepted: 08/18/2005] [Indexed: 01/14/2023]
Abstract
Interferons (IFNs) were discovered as natural antiviral substances produced during viral infection and were initially characterized for their ability to "interfere" with viral replication, slow cell proliferation, and profound alteration of immunity. The IFNs are synthesized and secreted by monocytes, macrophages, T-lymphocytes, neurons, and glia cells. The different IFNs are classified into three classes: alpha, beta, and gamma. alpha-IFN produced in the brain exerts direct effects on the brain and endocrine system by activating the neurosecretory hypothalamic neurons and regulates the hypothalamic-pituitary-adrenocortical axis. IFNs modulate neurophysiological activities of many brain region involving in pain, temperature, and food intake regulation. alpha-IFN administration activates the sympathetic nerves innervating components of the immune system. IFNs may serve as regulatory mediators between the central nervous system, the immune system, and endocrine system. IFN is used as immunologic therapy to treat various hematologic malignancies and infectious ailments and autoimmune diseases.
Collapse
Affiliation(s)
- Nachum Dafny
- Department of Neurobiology and Anatomy, The University of Texas, Medical School, P.O. Box 20708, Houston, Texas 77225, USA.
| | | |
Collapse
|
22
|
Bolin LM, Zhaung A, Strychkarska-Orczyk I, Nelson E, Huang I, Malit M, Nguyen Q. Differential inflammatory activation of IL-6 (−/−) astrocytes. Cytokine 2005; 30:47-55. [PMID: 15804595 DOI: 10.1016/j.cyto.2004.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Revised: 11/22/2004] [Accepted: 11/29/2004] [Indexed: 10/25/2022]
Abstract
IL-6 is a major immunomodulatory cytokine with neuroprotective activity. The absence of interleukin-6 (IL-6) results in increased vulnerability of dopaminergic neurons to the neurotoxicant, MPTP, and a compromised reactive microgliosis. To determine how astrogliosis may contribute to nigrostriatal degeneration in IL-6 (-/-) mice, the inflammatory profiles of astrocytes of IL-6 genotype were compared. Fourteen cytokines and four chemokines were simultaneously assayed in the supernatants of LPS-stimulated primary astrocyte cultures. In a time course of 6, 18 and 48 h and LPS stimulations of 0, 0.1, 1, 10 and 100 ng/ml, IL-6 (-/-) astrocytes secreted significantly greater amounts of the pro-inflammatory cytokines IL-1alpha, IL-1beta and TNFalpha than did IL-6 (+/+) cells. Elevated levels of IL-10 and IL-12p40 were only detected at 48 h post-stimulation with greater IL-10 in IL-6 (-/-) supernatants and greater IL-12p40 in IL-6 (+/+) supernatants. IL-6 (+/+) astrocytes produced more G-CSF and GM-CSF when compared with IL-6 (-/-) astrocytes. Chemokine levels were greater in supernatants of IL-6 (+/+) astrocytes than IL-6 (-/-) cells prior to 48 h post-stimulation. At that time, higher levels of MIP-1alpha were maintained in IL-6 (+/+) supernatant, while similar levels of MCP-1 in supernatants of both IL-6 (+/+) and IL-6 (-/-) cells were measured. Additionally, LPS (100 ng/ml) resulted in greater levels of KC and Rantes in IL-6 (-/-) astrocyte supernatants compared with IL-6 (+/+) supernatants at that time. These results suggest that the autocrine modulatory activities of IL-6 affect multiple cytokine secretory pathways, which could participate in neurodegenerative processes.
Collapse
Affiliation(s)
- L M Bolin
- The Parkinson's Institute, 1170 Morse Avenue, Sunnyvale, CA 94089-1605, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Häusler KG, Prinz M, Nolte C, Weber JR, Schumann RR, Kettenmann H, Hanisch UK. Interferon-gamma differentially modulates the release of cytokines and chemokines in lipopolysaccharide- and pneumococcal cell wall-stimulated mouse microglia and macrophages. Eur J Neurosci 2002; 16:2113-22. [PMID: 12473079 DOI: 10.1046/j.1460-9568.2002.02287.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During bacterial infections of the CNS, activated microglia could support leucocyte recruitment to the brain through the synthesis of cyto- and chemokines. In turn, invading leucocytes may feedback on microglial cells to influence their chemokine release pattern. Here, we analyzed the capacity of interferon-gamma (IFNgamma) to serve as such a leucocyte-to-microglia signal. Production of cyto- and chemokines was stimulated in mouse microglia cultures by treatments with lipopolysaccharide (LPS) from Gram-negative Escherichia coli or cell walls from Gram-positive Streptococcus pneumoniae (PCW). IFNgamma presence during the stimulation (0.1-100 ng/mL) modulated the patterns of LPS- and PCW-induced cyto- and chemokine release in a dose-dependent, potent and complex manner. While amounts of TNFalpha and IL-6 remained nearly unchanged, IFNgamma enhanced the production of IL-12, MCP-1 and RANTES, but attenuated that of KC, MIP-1alpha and MIP-2. Release modulation was obtained with IFNgamma preincubation (treatment of cells before LPS or PCW administration), coincubation and even delayed addition to an ongoing LPS or PCW stimulation. Together the changes observed for the microglial chemokine release under IFNgamma would shift the chemoattractive profile from favouring neutrophils to a preferential attraction of monocytes and T lymphocyte populations--as actually seen during the course of bacterial meningitis. The findings support the view of activated microglia as a major intrinsic source for an instant production of a variety of chemokines and suggest that leucocyte-derived IFNgamma could potentially regulate the microglial chemokine release pattern.
Collapse
Affiliation(s)
- Karl Georg Häusler
- Max Delbrück Center for Molecular Medicine, Cellular Neurosciences, Robert-Rössle-Strasse 10, D-13092 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
24
|
Kubota T, Majde JA, Brown RA, Krueger JM. Tumor necrosis factor receptor fragment attenuates interferon-gamma-induced non-REM sleep in rabbits. J Neuroimmunol 2001; 119:192-8. [PMID: 11585621 DOI: 10.1016/s0165-5728(01)00382-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although the somnogenic actions of interferon-alpha (IFNalpha) and IFNbeta have been reported, the sleep effects of IFNgamma remained unknown. Thus, we investigated the effects of intracerebroventricular injection of human IFNgamma on sleep in rabbits. IFNgamma dose-dependently increased nonrapid eye movement sleep (NREMS), electroencephalographic slow wave activity and brain temperature (Tbr). These effects were markedly attenuated after the heat treatment of IFNgamma. IFNgamma suppressed rapid eye movement sleep if given during the light period, but not if given at dark onset. Although a tumor necrosis factor receptor fragment did not affect any sleep parameters when given at dark onset, it significantly attenuated IFNgamma-induced NREMS and Tbr. These data suggest that IFNgamma may be involved in the sleep responses during infection. Further, IFNgamma may have a synergistic interaction with intrinsic TNFalpha in the brain.
Collapse
Affiliation(s)
- T Kubota
- Department of Veterinary, Comparative Anatomy, Pharmacology and Physiology, College of Veterinary Medicine, Washington State University, P.O. Box 646520, Pullman, WA 99164-6520, USA
| | | | | | | |
Collapse
|
25
|
Wakita T, Shintani F, Yagi G, Asai M, Nozawa S. Combination of inflammatory cytokines increases nitrite and nitrate levels in the paraventricular nucleus of conscious rats. Brain Res 2001; 905:12-20. [PMID: 11423074 DOI: 10.1016/s0006-8993(01)02346-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inflammatory cytokines stimulate glial cells in vitro to produce nitric oxide (NO) from inducible NO synthase (iNOS). Whether the stimulation with cytokines produces NO derived from iNOS has not hitherto been demonstrated in the vivo brain. Nitrite and nitrate (NOx(-)) levels in the rat paraventricular nucleus (PVN) were measured before and after intraparenchymal microinjection of cytokines with a microdialysis technique. The cytokines, tumor necrosis factor (TNF)-alpha (10 ng), interleukin (IL)-1 beta (2 ng), and interferon (IFN)-gamma (2 ng) were microinjected. None of the cytokines alone had any effect on the NOx(-) levels for 8 h. But a combination of TNF-alpha and IFN-gamma gradually increased NOx(-) levels beginning at 140 min after the microinjection, and NOx(-) levels reached 1.8 times the basal level at 380 min. A combination of TNF-alpha and IL-1 beta increased NOx(-) beginning at 340 min, reaching 1.7 times the basal level at 440 min, whereas a combination of IL-1 beta and IFN-gamma had no effect. Microinjection of a mixture of all three cytokines increased NOx(-) levels beginning at 120 min, reaching 3.3 times the basal level at 400 min. Aminoguanidine, which is a selective inhibitor of iNOS, reduced NOx(-) levels induced by the mixed cytokine treatment. Semi-quantitative RT-PCR for iNOS mRNA was done. The intensity of the iNOS mRNA band for the cytokine-treated PVN was stronger than that for the vehicle-treated PVN. These results suggest that the increased NOx(-) after the treatment with mixed cytokines were dependent on iNOS activity. This is the first report to indicate that only cytokines induce NOS in vivo in the brain.
Collapse
Affiliation(s)
- T Wakita
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | | | | | | | | |
Collapse
|
26
|
Ishizaka M, Ohe Y, Senbongi T, Wakabayashi K, Ishikawa K. Inflammatory stimuli increase prostaglandin D synthase levels in cerebrospinal fluid of rats. Neuroreport 2001; 12:1161-5. [PMID: 11338184 DOI: 10.1097/00001756-200105080-00022] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To examine the possibility that prostaglandin D synthase in CSF is involved in the brain responses to inflammation, we monitored the enzyme levels before and after administration of a bacterial endotoxin lipopolysaccharide (LPS) or proinflammatory cytokines. After an i.p. injection of LPS, the synthase levels began to rise within 1 day, peaked at 2 days and returned to the basal by 5 days after injection. The enzyme levels increased in a similar manner in response to tumor necrosis factor-alpha or interleukin (IL)-1 beta injected i.v. Centrally administered IL-1 beta had a similar effect. These findings support the hypothesis that PGD synthase in CSF participates in the brain responses to inflammation.
Collapse
Affiliation(s)
- M Ishizaka
- Institute for Molecular and Cellular Regulation, Gunma University, Showa-machi, Maebashi, Gunma 371, Japan
| | | | | | | | | |
Collapse
|
27
|
Blasko I, Ransmayr G, Veerhuis R, Eikelenboom P, Grubeck-Loebenstein B. Does IFNgamma play a role in neurodegeneration? J Neuroimmunol 2001; 116:1-4. [PMID: 11311323 DOI: 10.1016/s0165-5728(01)00279-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- I Blasko
- Department of Neurology, University Hospital of Innsbruck, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
28
|
Melcangi RC, Cavarretta I, Magnaghi V, Ciusani E, Salmaggi A. Corticosteroids protect oligodendrocytes from cytokine-induced cell death. Neuroreport 2000; 11:3969-72. [PMID: 11192611 DOI: 10.1097/00001756-200012180-00013] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The present data show that the simultaneous exposure to tumor necrosis factor-alpha (TNFalpha) and interferon-gamma (IFNgamma) induces cell death with characteristics of apoptosis in cultured rat oligodendrocytes; TNFalpha alone was ineffective. We have also demonstrated that different corticosteroids (aldosterone, deoxycorticosterone, dexamethasone and corticosterone) protect rat oligodendrocytes in culture from apoptosis induced by TNFalpha plus IFNgamma. This effect seems to be exerted via the interaction with both type I and type II corticosteroid receptors since all steroids considered are effective. Since oligodendrocyte apoptosis represents an important event in multiple sclerosis and in several demyelinating diseases, the present observations might be considered an interesting background for further researches directed to the possibility of controlling in vivo the death of these cells.
Collapse
Affiliation(s)
- R C Melcangi
- Department of Endocrinology, University of Milan, Italy
| | | | | | | | | |
Collapse
|
29
|
Blasko I, Veerhuis R, Stampfer-Kountchev M, Saurwein-Teissl M, Eikelenboom P, Grubeck-Loebenstein B. Costimulatory effects of interferon-gamma and interleukin-1beta or tumor necrosis factor alpha on the synthesis of Abeta1-40 and Abeta1-42 by human astrocytes. Neurobiol Dis 2000; 7:682-9. [PMID: 11114266 DOI: 10.1006/nbdi.2000.0321] [Citation(s) in RCA: 184] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic inflammation and astrocytosis are characteristic histopathological features of Alzheimer's Disease (AD). Astrocytes are one of the predominant cell types in the brain. In AD they are activated and produce inflammatory components such as complement components, acute phase proteins, and cytokines. In this study we analyzed the effect of cytokines on the production of amyloid beta (Abeta) in the astrocytoma cell line U373 and in primary human astrocytes isolated postmortem from healthy aged persons as well as from patients with AD. Astrocytes did not produce Abeta in the absence of stimuli or following stimulation with IL-1beta, TNFalpha, IL-6, and TGF-beta1. Neither did combinations of TNFalpha and IL-1beta, IL-6 or TGF-beta1, or the coadministration of IFNgamma and IL-6 or TGF-beta1 induce Abeta production. In contrast, pronounced production of Abeta1-40 and Abeta1-42 was observed when primary astrocytes or astrocytoma cells were stimulated with combinations of IFNgamma and TNFalpha or IFNgamma and IL-1beta. Induction of Abeta production was accompanied by decreased glycosylation of APP as well as by increased secretion of APPsbeta. Our results suggest that astrocytes may be an important source of Abeta in the presence of certain combinations of inflammatory cytokines. IFNgamma in combination with TNFalpha or IL-1beta seems to trigger Abeta production by supporting beta-secretase cleavage of the immature APP molecule.
Collapse
Affiliation(s)
- I Blasko
- Institute for Biomedical Aging Research of the Austrian Academy of Sciences, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
30
|
Jander S, Schroeter M, Fischer J, Stoll G. Differential regulation of microglial keratan sulfate immunoreactivity by proinflammatory cytokines and colony-stimulating factors. Glia 2000; 30:401-10. [PMID: 10797620 DOI: 10.1002/(sici)1098-1136(200006)30:4<401::aid-glia90>3.0.co;2-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Resident microglia of the rat CNS express a unique type of keratan sulfate immunoreactivity (KS-IR) that is lacking on peripheral monocytes/macrophages and associated with a so far unknown proteoglycan core protein. Microglial KS-IR is downregulated during T-cell-mediated autoimmune inflammation but largely preserved in degenerative lesion paradigms. This study addresses the role of cytokines and colony-stimulating factors in the regulation of microglial KS-IR. In vitro, ramified microglia in coculture with astrocytes, but not isolated microglia, constitutively expressed KS-IR under control conditions. In both culture paradigms, KS-IR was increased significantly by macrophage- (M-CSF) and granulocyte/macrophage colony-stimulating factors (GM-CSF), as well as tumor necrosis factor-alpha (TNF-alpha). By contrast, the Th1 cytokine interferon-gamma (IFN-gamma) downregulated KS-IR, both when applied alone or in combination with either GM-CSF, M-CSF, or TNF-alpha. In vivo, the intracerebroventricular administration of IFN-gamma, but not TNF-alpha, to healthy rats led to an almost complete disappearance of KS-IR from ramified brain microglia. Our data suggest that the expression of microglial KS-IR is under dominant negative control by the Th1 cell cytokine IFN-gamma and represent the first evidence of cytokine-dependent proteoglycan regulation in the CNS.
Collapse
Affiliation(s)
- S Jander
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany.
| | | | | | | |
Collapse
|
31
|
Yuan L, Neufeld AH. Tumor necrosis factor-?: A potentially neurodestructive cytokine produced by glia in the human glaucomatous optic nerve head. Glia 2000. [DOI: 10.1002/1098-1136(200010)32:1<42::aid-glia40>3.0.co;2-3] [Citation(s) in RCA: 193] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|