1
|
Mohammad ZB, Yudin SCY, Goldberg BJ, Serra KL, Klegeris A. Exploring neuroglial signaling: diversity of molecules implicated in microglia-to-astrocyte neuroimmune communication. Rev Neurosci 2025; 36:91-117. [PMID: 39240134 PMCID: PMC11717358 DOI: 10.1515/revneuro-2024-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Effective communication between different cell types is essential for brain health, and dysregulation of this process leads to neuropathologies. Brain glial cells, including microglia and astrocytes, orchestrate immune defense and neuroimmune responses under pathological conditions during which interglial communication is indispensable. Our appreciation of the complexity of these processes is rapidly increasing due to recent advances in molecular biology techniques, which have identified numerous phenotypic states of both microglia and astrocytes. This review focuses on microglia-to-astrocyte communication facilitated by secreted neuroimmune modulators. The combinations of interleukin (IL)-1α, tumor necrosis factor (TNF), plus complement component C1q as well as IL-1β plus TNF are already well-established microglia-derived stimuli that induce reactive phenotypes in astrocytes. However, given the large number of inflammatory mediators secreted by microglia and the rapidly increasing number of distinct functional states recognized in astrocytes, it can be hypothesized that many more intercellular signaling molecules exist. This review identifies the following group of cytokines and gliotransmitters that, while not established as interglial mediators yet, are known to be released by microglia and elicit functional responses in astrocytes: IL-10, IL-12, IL-18, transforming growth factor (TGF)-β, interferon (IFN)-γ, C-C motif chemokine ligand (CCL)5, adenosine triphosphate (ATP), l-glutamate, and prostaglandin E2 (PGE2). The review of molecular mechanisms engaged by these mediators reveals complex, partially overlapping signaling pathways implicated in numerous neuropathologies. Additionally, lack of human-specific studies is identified as a significant knowledge gap. Further research on microglia-to-astrocyte communication is warranted, as it could discover novel interglial signaling-targeted therapies for diverse neurological disorders.
Collapse
Affiliation(s)
- Zainab B. Mohammad
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Samantha C. Y. Yudin
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Benjamin J. Goldberg
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Kursti L. Serra
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
2
|
Iitani Y, Miki R, Imai K, Fuma K, Ushida T, Tano S, Yoshida K, Yokoi A, Kajiyama H, Kotani T. Interleukin-17A stimulation induces alterations in Microglial microRNA expression profiles. Pediatr Res 2024; 95:167-173. [PMID: 37758861 DOI: 10.1038/s41390-023-02825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Increased maternal interleukin (IL)-17A and activated microglia are pivotal factors contributing to the pathological phenotypes of maternal immune activation (MIA), developing neurodevelopmental disorders in offspring. This study aimed to determine whether IL-17A affects the microglial microRNA (miRNA) profiles. METHODS The miRNA expression profiles of primary cultured microglia stimulated with recombinant IL-17A were examined comprehensively using miRNA sequencing and validated through qRT-PCR. The expressions of miRNAs target genes identified using bioinformatics, were investigated in microglia transfected with mimic miRNA. The target gene's expression was also examined in the fetal brains of the MIA mouse model induced by maternal lipopolysaccharide (LPS) administration. RESULTS Primary cultured microglia expressed the IL-17A receptor and increased proinflammatory cytokines and nitric oxide synthase 2 upon treatment with IL-17A. Among the three miRNAs with |log2FC | >1, only mmu-miR-206-3p expression was significantly up-regulated by IL-17A. Transfection with the mmu-miR-206-3p mimic resulted in a significant decrease in the expression of Hdac4 and Igf1, target genes of mmu-miR-206-3p. Hdac4 expression also significantly decreased in the LPS-induced MIA model. CONCLUSIONS IL-17A affected microglial miRNA profiles with upregulated mmu-miR-206-3p. These findings suggest that targeting the IL-17A/mmu-miR-206-3p pathway may be a new strategy for predicting MIA-related neurodevelopmental deficits and providing preventive interventions. IMPACT Despite the growing evidence of interleukin (IL)-17A and microglia in the pathology of maternal immune activation (MIA), the downstream of IL-17A in microglia is not fully known. IL-17A altered microRNA profiles and upregulated the mmu-miR-206-3p expression in microglia. The mmu-miR-206-3p reduced autism spectrum disorder (ASD) related gene expressions, Hdac4 and Igf1. The Hdac4 expression was also reduced in the brain of MIA offspring. The hsa-miR-206 sequence is consistent with that of mmu-miR-206-3p. This study may provide clues to pathological mechanisms leading to predictions and interventions for ASD children born to mothers with IL-17A-related disorders.
Collapse
Affiliation(s)
- Yukako Iitani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Rika Miki
- Laboratory of Bell Research Center‑Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Kazuya Fuma
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
- Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, 466‑8560, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
- Nagoya University Institute for Advanced Research, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
- Nagoya University Institute for Advanced Research, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan.
- Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, 466‑8560, Japan.
| |
Collapse
|
3
|
Rosas Almanza J, Stehlik KE, Page JJ, Xiong SH, Tabor EG, Aperi B, Patel K, Kodali R, Kurpad S, Budde MD, Tarima S, Swartz K, Kroner A. IL-12p40 promotes secondary damage and functional impairment after spinal cord contusional injury. J Neurosci Res 2022; 100:2213-2231. [PMID: 36089917 DOI: 10.1002/jnr.25122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 01/07/2023]
Abstract
Secondary damage obstructs functional recovery for individuals who have sustained a spinal cord injury (SCI). Two processes significantly contributing to tissue damage after trauma are spinal cord hemorrhage and inflammation: more specifically, the recruitment and activation of immune cells, frequently driven by pro-inflammatory factors. Cytokines are inflammatory mediators capable of modulating the immune response. While cytokines are necessary to elicit inflammation for proper healing, excessive inflammation can result in destructive processes. The pro-inflammatory cytokines IL-12 and IL-23 are pathogenic in multiple autoimmune diseases. The cytokine subunit IL-12p40 is necessary to form bioactive IL-12 and IL-23. In this study, we examined the relationship between spinal cord hemorrhage and IL-12-related factors, as well as the impact of IL-12p40 (IL-12/IL-23) on secondary damage and functional recovery after SCI. Using in vivo magnetic resonance imaging and protein tissue analyses, we demonstrated a positive correlation between IL-12 and tissue hemorrhage. Receptor and ligand subunits of IL-12 were significantly upregulated after injury and colocalized with astrocytes, demonstrating a myriad of opportunities for IL-12 to induce an inflammatory response. IL-12p40-/- mice demonstrated significantly improved functional recovery and reduced lesion sizes compared to wild-type mice. Targeted gene array analysis in wild-type and IL-12p40-/- female mice after SCI revealed an upregulation of genes associated with worsened recovery after SCI. Taken together, our data reveal a pathogenic role of IL-12p40 in the secondary damage after SCI, hindering functional recovery. IL-12p40 (IL-12/IL-23) is thus an enticing neuroinflammatory target for further study as a potential therapeutic target to benefit recovery in acute SCI.
Collapse
Affiliation(s)
- Jose Rosas Almanza
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Kyle E Stehlik
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Justin J Page
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Shuana H Xiong
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Emma G Tabor
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Brandy Aperi
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Kishan Patel
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Rajiv Kodali
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Shekar Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Matthew D Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Sergey Tarima
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Karin Swartz
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
4
|
Ganguly U, Singh S, Chakrabarti S, Saini AK, Saini RV. Immunotherapeutic interventions in Parkinson's disease: Focus on α-Synuclein. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 129:381-433. [PMID: 35305723 DOI: 10.1016/bs.apcsb.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized classically by motor manifestations. However, nonmotor symptoms appear early in the course of the disease progression, making both diagnosis and treatment difficult. The pathology of PD is complicated by the accumulation and aggregation of misfolded proteins in intracellular cytoplasmic inclusions called Lewy bodies (LBs). The main toxic component of LBs is the protein α-Synuclein which plays a pivotal role in PD pathogenesis. α-Synuclein can propagate from cell-to-cell exhibiting prion-like properties and spread PD pathology throughout the central nervous system. Immunotherapeutic interventions in PD, both active and passive immunization, have targeted α-Synuclein in both experimental models and clinical trials. In addition, targeting the hyperactive inflammation in PD also holds promise in designing potential immunotherapeutics. The inflammatory and proteotoxic pathways are interlinked and contribute immensely to the disease pathology. In this chapter, we critically review the targets of immunotherapeutic interventions in PD, focusing on the pathogenetic mechanisms of PD, particularly neuroinflammation and α-Synuclein misfolding, aggregation, and propagation. We thoroughly summarized the various immunotherapeutic strategies designed to treat PD-in vitro, in vivo, and clinical trials. The development of these targeted immunotherapies could open a new avenue in the treatment of patients with PD.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, India
| | - Sukhpal Singh
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, India
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, India
| | - Adesh K Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, India
| | - Reena V Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, India.
| |
Collapse
|
5
|
Vedam-Mai V. Harnessing the immune system for the treatment of Parkinson's disease. Brain Res 2021; 1758:147308. [PMID: 33524380 DOI: 10.1016/j.brainres.2021.147308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/11/2020] [Accepted: 01/16/2021] [Indexed: 01/03/2023]
Abstract
Current treatment options for Parkinson's disease (PD) typically aim to replace dopamine, and hence only provide symptomatic relief. However, in the long run, this approach alone loses its efficacy as it is associated with debilitating side effects. Hence there is an unmet clinical need for addressing levodopa resistant symptoms, and an urgency to develop therapies that can halt or prevent the course of PD. The premise that α-syn can transmit from cell-to-cell in a prion like manner has opened up the possibility for the use of immunotherapy in PD. There is evidence for inflammation in PD as is evidenced by microglial activation, as well as the involvement of the peripheral immune system in PD, and peripheral inflammation can exacerbate dopaminergic degeneration as seen in animal models of the disease. However, mechanisms that link the immune system with PD are not clear, and the sequence of immune responses with respect to PD are still unknown. Nevertheless, our present knowledge offers avenues for the development of immune-based therapies for PD. In order to successfully employ such strategies, we must comprehend the state of the peripheral immune system during the course of PD. This review describes the developments in the field of both active and passive immunotherapies in the treatment of PD, and highlights the crucial need for future research for clarifying the role of inflammation and immunity in this debilitating disease.
Collapse
|
6
|
Kubota K, Ogawa M, Ji B, Watabe T, Zhang MR, Suzuki H, Sawada M, Nishi K, Kudo T. Basic Science of PET Imaging for Inflammatory Diseases. PET/CT FOR INFLAMMATORY DISEASES 2020. [PMCID: PMC7418531 DOI: 10.1007/978-981-15-0810-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
FDG-PET/CT has recently emerged as a useful tool for the evaluation of inflammatory diseases too, in addition to that of malignant diseases. The imaging is based on active glucose utilization by inflammatory tissue. Autoradiography studies have demonstrated high FDG uptake in macrophages, granulocytes, fibroblasts, and granulation tissue. Especially, activated macrophages are responsible for the elevated FDG uptake in some types of inflammation. According to one study, after activation by lipopolysaccharide of cultured macrophages, the [14C]2DG uptake by the cells doubled, reaching the level seen in glioblastoma cells. In activated macrophages, increase in the expression of total GLUT1 and redistributions from the intracellular compartments toward the cell surface have been reported. In one rheumatoid arthritis model, following stimulation by hypoxia or TNF-α, the highest elevation of the [3H]FDG uptake was observed in the fibroblasts, followed by that in macrophages and neutrophils. As the fundamental mechanism of elevated glucose uptake in both cancer cells and inflammatory cells, activation of glucose metabolism as an adaptive response to a hypoxic environment has been reported, with transcription factor HIF-1α playing a key role. Inflammatory cells and cancer cells seem to share the same molecular mechanism of elevated glucose metabolism, lending support to the notion of usefulness of FDGPET/CT for the evaluation of inflammatory diseases, besides cancer.
Collapse
|
7
|
de Abreu MS, Giacomini ACVV, Zanandrea R, Dos Santos BE, Genario R, de Oliveira GG, Friend AJ, Amstislavskaya TG, Kalueff AV. Psychoneuroimmunology and immunopsychiatry of zebrafish. Psychoneuroendocrinology 2018; 92:1-12. [PMID: 29609110 DOI: 10.1016/j.psyneuen.2018.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
Abstract
Despite the high prevalence of neural and immune disorders, their etiology and molecular mechanisms remain poorly understood. As the zebrafish (Danio rerio) is increasingly utilized as a powerful model organism in biomedical research, mounting evidence suggests these fish as a useful tool to study neural and immune mechanisms and their interplay. Here, we discuss zebrafish neuro-immune mechanisms and their pharmacological and genetic modulation, the effect of stress on cytokines, as well as relevant models of microbiota-brain interplay. As many human brain diseases are based on complex interplay between the neural and the immune system, here we discuss zebrafish models, as well as recent successes and challenges, in this rapidly expanding field. We particularly emphasize the growing utility of zebrafish models in translational immunopsychiatry research, as they improve our understanding of pathogenetic neuro-immune interactions, thereby fostering future discovery of potential therapeutic agents.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; Postgraduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Ana C V V Giacomini
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; Postgraduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria, Brazil; Postgraduate Program in Environmental Sciences, University of Passo Fundo (UPF), Passo Fundo, Brazil
| | - Rodrigo Zanandrea
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Bruna E Dos Santos
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Rafael Genario
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | | | - Ashton J Friend
- Tulane University School of Science and Engineering, New Orleans, LA, USA
| | - Tamara G Amstislavskaya
- Research Institute of Physiology and Basic Medicine SB RAS, and Department of Neuroscience, Novosibirsk State University, Novosibirsk, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia; ZENEREI Research Center, Slidell, LA, USA; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, St. Petersburg, Russia; Russian Research Center for Radiology and Surgical Technologies, Pesochny, Russia; Laboratory of Translational Biopsychiatry, Research Institute of Physiology and Basic Medicine SB RAS, Novosibirsk, Russia.
| |
Collapse
|
8
|
Chauhan P, Sheng WS, Hu S, Prasad S, Lokensgard JR. Nitrosative damage during retrovirus infection-induced neuropathic pain. J Neuroinflammation 2018; 15:66. [PMID: 29506535 PMCID: PMC5836380 DOI: 10.1186/s12974-018-1107-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/26/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Peripheral neuropathy is currently the most common neurological complication in HIV-infected individuals, occurring in 35-50% of patients undergoing combination anti-retroviral therapy. Data have shown that distal symmetric polyneuropathy develops in mice by 6 weeks following infection with the LP-BM5 retrovirus mixture. Previous work from our laboratory has demonstrated that glial cells modulate antiviral T-cell effector responses through the programmed death (PD)-1: PD-L1 pathway, thereby limiting the deleterious consequences of unrestrained neuroinflammation. METHODS Using the MouseMet electronic von Frey system, we assessed hind-paw mechanical hypersensitivity in LP-BM5-infected wild-type (WT) and PD-1 KO animals. Using multi-color flow cytometry, we quantitatively assessed cellular infiltration and microglial activation. Using real-time RT-PCR, we assessed viral load, expression of IFN-γ, iNOS, and MHC class II. Using western blotting, we measured protein nitrosylation within the lumbar spinal cord (LSC) and dorsal root ganglion (DRG). Histochemical staining was performed to analyze the presence of CD3, ionized calcium binding adaptor molecule (Iba)-1, MHCII, nitrotyrosine, isolectin B4 (IB4) binding, and neurofilament 200 (NF200). Statistical analyses were carried out using graphpad prism. RESULTS Hind-paw mechanical hypersensitivity observed in LP-BM5-infected animals was associated with significantly increased lymphocyte infiltration into the spinal cord and DRG. We also observed elevated expression of IFN-γ (in LSC and DRG) and MHC II (on resident microglia in LSC). We detected elevated levels of 3-nitrotyrosine within the LSC and DRG of LP-BM5-infected animals, an indicator of nitric oxide (NO)-induced protein damage. Moreover, we observed 3-nitrotyrosine in both small (IB4+) and large (NF200+) DRG sensory neurons. Additionally, infected PD-1 KO animals displayed significantly greater mechanical hypersensitivity than WT or uninfected mice at 4 weeks post-infection (p.i.). Accelerated onset of hind-paw hypersensitivity in PD-1 KO animals was associated with significantly increased infiltration of CD4+ and CD8+ T lymphocytes, macrophages, and microglial activation at early time points. Importantly, we also observed elevated levels of 3-nitrotyrosine and iNOS in infected PD-1 KO animals when compared with WT animals. CONCLUSIONS Results reported here connect peripheral immune cell infiltration and reactive gliosis with nitrosative damage. These data may help elucidate how retroviral infection-induced neuroinflammatory networks contribute to nerve damage and neuropathic pain.
Collapse
Affiliation(s)
- Priyanka Chauhan
- Department of Medicine, Neurovirology Laboratory, University of Minnesota Medical School, 3-107 Microbiology Research Facility, 689 23rd Ave. S.E, Minneapolis, MN 55455 USA
| | - Wen S. Sheng
- Department of Medicine, Neurovirology Laboratory, University of Minnesota Medical School, 3-107 Microbiology Research Facility, 689 23rd Ave. S.E, Minneapolis, MN 55455 USA
| | - Shuxian Hu
- Department of Medicine, Neurovirology Laboratory, University of Minnesota Medical School, 3-107 Microbiology Research Facility, 689 23rd Ave. S.E, Minneapolis, MN 55455 USA
| | - Sujata Prasad
- Department of Medicine, Neurovirology Laboratory, University of Minnesota Medical School, 3-107 Microbiology Research Facility, 689 23rd Ave. S.E, Minneapolis, MN 55455 USA
| | - James R. Lokensgard
- Department of Medicine, Neurovirology Laboratory, University of Minnesota Medical School, 3-107 Microbiology Research Facility, 689 23rd Ave. S.E, Minneapolis, MN 55455 USA
| |
Collapse
|
9
|
Imai K, Kotani T, Tsuda H, Mano Y, Nakano T, Ushida T, Li H, Miki R, Sumigama S, Iwase A, Hirakawa A, Ohno K, Toyokuni S, Takeuchi H, Mizuno T, Suzumura A, Kikkawa F. Neuroprotective potential of molecular hydrogen against perinatal brain injury via suppression of activated microglia. Free Radic Biol Med 2016; 91:154-63. [PMID: 26709014 DOI: 10.1016/j.freeradbiomed.2015.12.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 12/15/2022]
Abstract
Exposure to inflammation in utero is related to perinatal brain injury, which is itself associated with high rates of long-term morbidity and mortality in children. Novel therapeutic interventions during the perinatal period are required to prevent inflammation, but its pathogenesis is incompletely understood. Activated microglia are known to play a central role in brain injury by producing a variety of pro-inflammatory cytokines and releasing oxidative products. The study is aimed to investigate the preventative potential of molecular hydrogen (H2), which is an antioxidant and anti-inflammatory agent without mutagenicity. Pregnant ICR mice were injected with lipopolysaccharide (LPS) intraperitoneally on embryonic day 17 to create a model of perinatal brain injury caused by prenatal inflammation. In this model, the effect of maternal administration of hydrogen water (HW) on pups was also evaluated. The levels of pro-inflammatory cytokines, oxidative damage and activation of microglia were determined in the fetal brains. H2 reduced the LPS-induced expression of pro-inflammatory cytokines, oxidative damage and microglial activation in the fetal brains. Next, we investigated how H2 contributes to neuroprotection, focusing on microglia, using primary cultured microglia and neurons. H2 prevented LPS- or cytokine-induced generation of reactive oxidative species by microglia and reduced LPS-induced microglial neurotoxicity. Finally, we identified several molecules influenced by H2, involved in the process of activating microglia. These results suggested that H2 holds promise for the prevention of inflammation related to perinatal brain injury.
Collapse
Affiliation(s)
- Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Hiroyuki Tsuda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yukio Mano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomoko Nakano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hua Li
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Rika Miki
- Laboratory of Bell Research Centre-Department of Obstetrics and Gynecology Collaborative Research, Bell Research Centre for Reproductive Health and Cancer, Department of Reproduction, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Seiji Sumigama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akihiro Hirakawa
- Biostatistics Section, Center for Advanced Medicine and Clinical Research Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
10
|
Sumida M, Hane M, Yabe U, Shimoda Y, Pearce OMT, Kiso M, Miyagi T, Sawada M, Varki A, Kitajima K, Sato C. Rapid Trimming of Cell Surface Polysialic Acid (PolySia) by Exovesicular Sialidase Triggers Release of Preexisting Surface Neurotrophin. J Biol Chem 2015; 290:13202-14. [PMID: 25750127 PMCID: PMC4505574 DOI: 10.1074/jbc.m115.638759] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/23/2015] [Indexed: 01/08/2023] Open
Abstract
As acidic glycocalyx on primary mouse microglial cells and a mouse microglial cell line Ra2, expression of polysialic acid (polySia/PSA), a polymer of the sialic acid Neu5Ac (N-acetylneuraminic acid), was demonstrated. PolySia is known to modulate cell adhesion, migration, and localization of neurotrophins mainly on neural cells. PolySia on Ra2 cells disappeared very rapidly after an inflammatory stimulus. Results of knockdown and inhibitor studies indicated that rapid surface clearance of polySia was achieved by secretion of endogenous sialidase Neu1 as an exovesicular component. Neu1-mediated polySia turnover was accompanied by the release of brain-derived neurotrophic factor normally retained by polySia molecules. Introduction of a single oxygen atom change into polySia by exogenous feeding of the non-neural sialic acid Neu5Gc (N-glycolylneuraminic acid) caused resistance to Neu1-induced polySia turnover and also inhibited the associated release of brain-derived neurotrophic factor. These results indicate the importance of rapid turnover of the polySia glycocalyx by exovesicular sialidases in neurotrophin regulation.
Collapse
Affiliation(s)
- Mizuki Sumida
- From the Bioscience and Biotechnology Center and School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Masaya Hane
- From the Bioscience and Biotechnology Center and School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Uichiro Yabe
- From the Bioscience and Biotechnology Center and School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Yasushi Shimoda
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomiokamachi, Nagaoka 940-2188 Japan
| | - Oliver M T Pearce
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0687
| | - Makoto Kiso
- Department of Applied Bioorganic Chemistry, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Taeko Miyagi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 981-8558, Sendai, Japan, and
| | - Makoto Sawada
- Department of Brain Functions, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Ajit Varki
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0687
| | - Ken Kitajima
- From the Bioscience and Biotechnology Center and School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan,
| | - Chihiro Sato
- From the Bioscience and Biotechnology Center and School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan,
| |
Collapse
|
11
|
Oligomeric amyloid β induces IL-1β processing via production of ROS: implication in Alzheimer's disease. Cell Death Dis 2013; 4:e975. [PMID: 24357806 PMCID: PMC3877570 DOI: 10.1038/cddis.2013.503] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 10/29/2013] [Accepted: 11/12/2013] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive neuronal loss and cognitive decline. Oligomeric amyloid β (oAβ) is involved in the pathogenesis of AD by affecting synaptic plasticity and inhibiting long-term potentiation. Although several lines of evidence suggests that microglia, the resident immune cells in the central nervous system (CNS), are neurotoxic in the development of AD, the mechanism whether or how oAβ induces microglial neurotoxicity remains unknown. Here, we show that oAβ promotes the processing of pro-interleukin (IL)-1β into mature IL-1β in microglia, which then enhances microglial neurotoxicity. The processing is induced by an increase in activity of caspase-1 and NOD-like receptor family, pyrin domain containing 3 (NLRP3) via mitochondrial reactive oxygen species (ROS) and partially via NADPH oxidase-induced ROS. The caspase-1 inhibitor Z-YVAD-FMK inhibits the processing of IL-1β, and attenuates microglial neurotoxicity. Our results indicate that microglia can be activated by oAβ to induce neuroinflammation through processing of IL-1β, a pro-inflammatory cytokine, in AD.
Collapse
|
12
|
Zhang XM, Zhu J. Kainic Acid-induced neurotoxicity: targeting glial responses and glia-derived cytokines. Curr Neuropharmacol 2012; 9:388-98. [PMID: 22131947 PMCID: PMC3131729 DOI: 10.2174/157015911795596540] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 09/28/2010] [Accepted: 10/18/2010] [Indexed: 01/01/2023] Open
Abstract
Glutamate excitotoxicity contributes to a variety of disorders in the central nervous system, which is triggered primarily by excessive Ca2+ influx arising from overstimulation of glutamate receptors, followed by disintegration of the endoplasmic reticulum (ER) membrane and ER stress, the generation and detoxification of reactive oxygen species as well as mitochondrial dysfunction, leading to neuronal apoptosis and necrosis. Kainic acid (KA), a potent agonist to the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate class of glutamate receptors, is 30-fold more potent in neuro-toxicity than glutamate. In rodents, KA injection resulted in recurrent seizures, behavioral changes and subsequent degeneration of selective populations of neurons in the brain, which has been widely used as a model to study the mechanisms of neurodegenerative pathways induced by excitatory neurotransmitter. Microglial activation and astrocytes proliferation are the other characteristics of KA-induced neurodegeneration. The cytokines and other inflammatory molecules secreted by activated glia cells can modify the outcome of disease progression. Thus, anti-oxidant and anti-inflammatory treatment could attenuate or prevent KA-induced neurodegeneration. In this review, we summarized updated experimental data with regard to the KA-induced neurotoxicity in the brain and emphasized glial responses and glia-oriented cytokines, tumor necrosis factor-α, interleukin (IL)-1, IL-12 and IL-18.
Collapse
Affiliation(s)
- Xing-Mei Zhang
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
13
|
Furr SR, Marriott I. Viral CNS infections: role of glial pattern recognition receptors in neuroinflammation. Front Microbiol 2012; 3:201. [PMID: 22723794 PMCID: PMC3379540 DOI: 10.3389/fmicb.2012.00201] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/15/2012] [Indexed: 12/13/2022] Open
Abstract
Viruses are the major causative agents of central nervous system (CNS) infection worldwide. RNA and DNA viruses trigger broad activation of glial cells including microglia and astrocytes, eliciting the release of an array of mediators that can promote innate and adaptive immune responses. Such responses can limit viral replication and dissemination leading to infection resolution. However, a defining feature of viral CNS infection is the rapid onset of severe neuroinflammation and overzealous glial responses are associated with significant neurological damage or even death. The mechanisms by which microglia and astrocytes perceive neurotropic RNA and DNA viruses are only now becoming apparent with the discovery of a variety of cell surface and cytosolic molecules that serve as sensors for viral components. In this review we discuss the role played by members of the Toll-like family of pattern recognition receptors (PRRs) in the inflammatory responses of glial cells to the principle causative agents of viral encephalitis. Importantly, we also describe the evidence for the involvement of a number of newly described intracellular PRRs, including retinoic acid-inducible gene I and DNA-dependent activator of IFN regulatory factors, that are thought to function as intracellular sensors of RNA and DNA viruses, respectively. Finally, we explore the possibility that cross-talk exists between these disparate viral sensors and their signaling pathways, and describe how glial cytosolic and cell surface/endosomal PRRs could act in a cooperative manner to promote the fulminant inflammation associated with acute neurotropic viral infection.
Collapse
Affiliation(s)
| | - Ian Marriott
- Department of Biology, University of North Carolina at Charlotte,Charlotte, NC, USA
| |
Collapse
|
14
|
Chastain EML, Duncan DS, Rodgers JM, Miller SD. The role of antigen presenting cells in multiple sclerosis. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1812:265-74. [PMID: 20637861 PMCID: PMC2970677 DOI: 10.1016/j.bbadis.2010.07.008] [Citation(s) in RCA: 188] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 07/06/2010] [Accepted: 07/07/2010] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a debilitating T cell mediated autoimmune disease of the central nervous system (CNS). Animal models of MS, such as experimental autoimmune encephalomyelitis (EAE) and Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) have given light to cellular mechanisms involved in the initiation and progression of this organ-specific autoimmune disease. Within the CNS, antigen presenting cells (APC) such as microglia and astrocytes participate as first line defenders against infections or inflammation. However, during chronic inflammation they can participate in perpetuating the self-destructive environment by secretion of inflammatory factors and/or presentation of myelin epitopes to autoreactive T cells. Dendritic cells (DC) are also participants in the presentation of antigen to T cells, even within the CNS. While the APCs alone are not solely responsible for mediating the destruction to the myelin sheath, they are critical players in perpetuating the inflammatory milieu. This review will highlight relevant studies which have provided insight to the roles played by microglia, DCs and astrocytes in the context of CNS autoimmunity.
Collapse
Affiliation(s)
- Emily M L Chastain
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | |
Collapse
|
15
|
Litteljohn D, Mangano E, Clarke M, Bobyn J, Moloney K, Hayley S. Inflammatory mechanisms of neurodegeneration in toxin-based models of Parkinson's disease. PARKINSONS DISEASE 2010; 2011:713517. [PMID: 21234362 PMCID: PMC3018622 DOI: 10.4061/2011/713517] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 12/09/2010] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) has been associated with exposure to a variety of environmental agents, including pesticides, heavy metals, and organic pollutants; and inflammatory processes appear to constitute a common mechanistic link among these insults. Indeed, toxin exposure has been repeatedly demonstrated to induce the release of oxidative and inflammatory factors from immunocompetent microglia, leading to damage and death of midbrain dopamine (DA) neurons. In particular, proinflammatory cytokines such as tumor necrosis factor-α and interferon-γ, which are produced locally within the brain by microglia, have been implicated in the loss of DA neurons in toxin-based models of PD; and mounting evidence suggests a contributory role of the inflammatory enzyme, cyclooxygenase-2. Likewise, immune-activating bacterial and viral agents were reported to have neurodegenerative effects themselves and to augment the deleterious impact of chemical toxins upon DA neurons. The present paper will focus upon the evidence linking microglia and their inflammatory processes to the death of DA neurons following toxin exposure. Particular attention will be devoted to the possibility that environmental toxins can activate microglia, resulting in these cells adopting a “sensitized” state that favors the production of proinflammatory cytokines and damaging oxidative radicals.
Collapse
Affiliation(s)
- Darcy Litteljohn
- Institute of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, Canada K1S 5B6
| | | | | | | | | | | |
Collapse
|
16
|
Seki Y, Suzuki SO, Masui K, Harada S, Nakamura S, Kanba S, Iwaki T. A simple and high-yield method for preparation of rat microglial cultures utilizing Aclar plastic film. Neuropathology 2010; 31:215-22. [PMID: 21092060 DOI: 10.1111/j.1440-1789.2010.01163.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Microglia are implicated in both neuroprotection and neurodegeneration, and are a key area of interest with respect to various CNS diseases. Until now, primary microglia prepared by various isolation methods have been widely used to investigate their role in CNS diseases. However, there are some problems with the current isolation methods, such as the numbers of animals required in order to obtain sufficient numbers of microglial cells due to low yields, and also the long periods of culture required. We herein describe a simple, high-yield method for isolating not only primary microglia, but also immortalized microglial cells. Our method allows for the isolation of an almost pure population of microglia with only two steps. First, a primary mixed neural culture was prepared from the brains of 3-day-old postnatal rats. Next, primary microglia were collected for 2 h by adhesion to Aclar plastic film. The average yield by this method was approximately 50 times higher than that of the conventional shaking method. Immortalized microglial cells could also be prepared based on this procedure. A plasmid vector encoding the SV40 large T antigen was transfected into the mixed neural culture using a calcium phosphate precipitation method. Then, proliferating immortalized microglia were collected after several weeks in a similar fashion. Several clones were obtained by limited dilution and one of the immortalized cell lines was designated SMK. The SMK cells exhibited markers specific for the microglia lineage, including Iba-1, CD11b, CD45, CD68, major histocompatibility complex (MHC) class I and MHC class II, but not for the astrocyte-specific markers, GFAP and glutamate aspartate transporter. SMK also showed phagocytic activity. In conclusion, this method resulted in a high-yield preparation of microglial cultures with ease and reproducibility.
Collapse
Affiliation(s)
- Yoshihiro Seki
- Department of Neuropathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
Ono K, Suzuki H, Sawada M. Delayed neural damage is induced by iNOS-expressing microglia in a brain injury model. Neurosci Lett 2010; 473:146-50. [DOI: 10.1016/j.neulet.2010.02.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 02/12/2010] [Accepted: 02/13/2010] [Indexed: 11/25/2022]
|
18
|
Stone DK, Reynolds AD, Mosley RL, Gendelman HE. Innate and adaptive immunity for the pathobiology of Parkinson's disease. Antioxid Redox Signal 2009; 11:2151-66. [PMID: 19243239 PMCID: PMC2788126 DOI: 10.1089/ars.2009.2460] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Innate and adaptive immunity affect the pathogenesis of Parkinson's disease (PD). In particular, activation of microglia influences degeneration of dopaminergic neurons. Cell-to-cell interactions and immune regulation critical for neuronal homeostasis also influence immune responses. The links between T cell immunity and nigrostriatal degeneration are supported by laboratory, animal model, and human pathologic investigations. Immune-associated biomarkers in spinal fluids and brain tissue of patients with idiopathic or familial forms of PD provide means to improve diagnosis and therapeutic monitoring. Relationships between oxidative stress, inflammation, and immune-mediated cell death pathways are examined in this review as they are linked to PD pathogenesis. Harnessing the immune system by drugs or by vaccination remain promising future therapeutic options.
Collapse
Affiliation(s)
- David K Stone
- Center for Neurovirology and Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | |
Collapse
|
19
|
Marked induction of inducible nitric oxide synthase and tumor necrosis factor-alpha in rat CD40+ microglia by comparison to CD40- microglia. J Neuroimmunol 2009; 208:70-9. [PMID: 19211155 DOI: 10.1016/j.jneuroim.2009.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 01/05/2009] [Accepted: 01/07/2009] [Indexed: 01/21/2023]
Abstract
There may be two subtypes of microglia (MG) at least in the CNS. We separated the two types from rat mixed glial culture. mRNAs and proteins for inducible nitric oxide synthase (iNOS) and tumor necrosis factor-alpha (TNFalpha) were more induced in CD40(+) MG than CD40(-) MG after LPS stimulation. Although the expression level of LPS receptors showed a little difference between the subtypes, LPS-induced degradation of phosphorylated IkappaBalpha was marked in CD40(+) MG. These results strongly suggest that CD40(+) MG produce larger amount of NO and TNFalpha to exhibit neurotoxic action under certain pathological conditions in brains.
Collapse
|
20
|
Yaguchi M, Ohta S, Toyama Y, Kawakami Y, Toda M. Functional recovery after spinal cord injury in mice through activation of microglia and dendritic cells after IL‐12 administration. J Neurosci Res 2008; 86:1972-80. [DOI: 10.1002/jnr.21658] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
21
|
Suzumura A. Immune Response in the Brain: Glial Response and Cytokine Production. CYTOKINES AND THE BRAIN 2008. [PMCID: PMC7185635 DOI: 10.1016/s1567-7443(07)10014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
22
|
Markus T, Hansson S, Amer-Wåhlin I, Hellström-Westas L, Saugstad OD, Ley D. Cerebral inflammatory response after fetal asphyxia and hyperoxic resuscitation in newborn sheep. Pediatr Res 2007; 62:71-7. [PMID: 17519806 DOI: 10.1203/pdr.0b013e31811ead6e] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Resuscitation with pure oxygen at birth after fetal asphyxia may aggravate brain damage by inducing pro-inflammation. The toll-like receptors (TLRs) may serve a pro-inflammatory role in hyperoxemia during ischemia-reperfusion. Sixteen near-term fetal sheep (132-136 d) were subjected to 10 min of cord occlusion, delivery and mechanical ventilation with 100% O2 (n = 8), or 21% O2 (n = 8) for 30 min followed by normoxemia for 90 min. Eight sheep fetuses were delivered immediately with inspired O2 targeted at normoxemia for 120 min (controls). Levels and distributions of mRNAs for IL-1beta, TNF-alpha, IL-12p40, IL-18, IL-6, IL-10, IFN-gamma, TLR-2, -3 and -4 in cerebral tissue at 2 h after birth were evaluated with real-time polymerase chain reaction (PCR) and in situ hybridization. Expressions of IL-1beta, IL-12p40, TLR-2, and TLR-4 were increased in cortex/subcortex after resuscitation with 100% O2 compared with 21% O2 (all p < 0.05) and to controls (all p < 0.05). Increased cellular expression of IL-1beta was localized to sub-meningeal cortical layers and to sub-cortical white matter. Hyperoxic resuscitation at birth following fetal asphyxia induces a cerebral pro-inflammatory response with an up-regulation of TLR-2 and -4. These may be early events leading to increased tissue damage after exposure to hyperoxemia at birth.
Collapse
Affiliation(s)
- Tina Markus
- Department of Pediatrics, Lund University, 221 85 Lund, Sweden
| | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
Ireland DDC, Reiss CS. Gene expression contributing to recruitment of circulating cells in response to vesicular stomatitis virus infection of the CNS. Viral Immunol 2006; 19:536-45. [PMID: 16987071 PMCID: PMC2562241 DOI: 10.1089/vim.2006.19.536] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
During acute Vesicular Stomatitis Virus (VSV) infection of the mouse central nervous system, neutrophils, natural killer (NK) cells, macrophages, and CD4+ and CD8+ T cells are recruited from the circulation in response to chemokines and cytokines. This study elucidated the production of these factors and infiltration of these peripheral cells. Chemokines that were observed included CCL1, CXCL10 (IP-10), CCL5 (RANTES), CCL3 (MIP-1alpha), CCL4 (MIP-1beta), CXCL1 (MIP-2), CCL2 (MCP-1), and CCL11 (eotaxin). Cytokines produced in response to the infection include IL-1 and interferon-gamma, but not type I interferons. Neutrophils are the first recruited cell type, appearing as early as 24 h after intranasal application of the virus. NK cells follow, but T cells are not detected until 6 days postinfection.
Collapse
Affiliation(s)
- Derek D C Ireland
- Department of Biology, New York University, New York, New York 10003-6688, USA
| | | |
Collapse
|
25
|
Takeuchi H, Wang J, Kawanokuchi J, Mitsuma N, Mizuno T, Suzumura A. Interferon-gamma induces microglial-activation-induced cell death: a hypothetical mechanism of relapse and remission in multiple sclerosis. Neurobiol Dis 2005; 22:33-9. [PMID: 16386911 DOI: 10.1016/j.nbd.2005.09.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 08/19/2005] [Accepted: 09/30/2005] [Indexed: 12/25/2022] Open
Abstract
Relapse and remission are characteristics of multiple sclerosis (MS). The underlying mechanisms, however, remain uncertain. Interferon-gamma (IFN-gamma) disturbs the immunological privilege of the central nervous system (CNS) by inducing major histocompatibility complex antigen expression in CNS cells and activating microglia to become antigen-presenting and effector cells. Thus, IFN-gamma and microglia are thought to play important roles in the initiation and development of MS. Here, we show that IFN-gamma induces microglial apoptosis as the activation-induced cell death. This microglial apoptosis was associated with the up-regulation of pro-apoptosis proteins, especially Bax. Microglial apoptosis was also observed in peak EAE mice, but not in early EAE mice. Therefore, IFN-gamma may act on microglia as part of a self-limiting negative feedback system. The activation and subsequent death of microglia induced by IFN-gamma may play pivotal roles in the mechanism of MS relapse and remission.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/immunology
- Cells, Cultured
- Central Nervous System/immunology
- Central Nervous System/pathology
- Central Nervous System/physiopathology
- Coculture Techniques
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Feedback, Physiological/immunology
- Gliosis/immunology
- Gliosis/metabolism
- Gliosis/physiopathology
- In Situ Nick-End Labeling
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C57BL
- Microglia/immunology
- Microglia/metabolism
- Models, Neurological
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/metabolism
- Multiple Sclerosis, Relapsing-Remitting/physiopathology
- Spinal Cord/immunology
- Spinal Cord/pathology
- Spinal Cord/physiopathology
- Up-Regulation/immunology
- bcl-2-Associated X Protein/immunology
Collapse
Affiliation(s)
- Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
Nakamichi K, Saiki M, Sawada M, Yamamuro Y, Morimoto K, Kurane I. Double-stranded RNA stimulates chemokine expression in microglia through vacuolar pH-dependent activation of intracellular signaling pathways. J Neurochem 2005; 95:273-83. [PMID: 16181431 DOI: 10.1111/j.1471-4159.2005.03354.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
During neurotropic virus infection, microglia act as a source of chemokines, thereby regulating the recruitment of peripheral leukocytes and the multicellular immune response within the CNS. Herein, we present a comprehensive study on the chemokine production by microglia in response to double-stranded RNA (dsRNA), a conserved molecular pattern of virus infection. Transcriptional analyses of chemokine genes revealed that dsRNA strongly induces the expression of CXC chemokine ligand 10 (CXCL10) and CC chemokine ligand 5 (CCL5) in microglia. We also observed that the dsRNA stimulation triggered the activation of signaling pathways mediated by nuclear factor kappaB (NF-kappaB) and mitogen-activated protein kinases (MAPK), including extracellular signal-regulated kinases 1 and 2 (ERK1/2), p38, and c-Jun N-terminal kinase (JNK). The microglial CXCL10 response to dsRNA was induced via NF-kappaB, p38, and JNK pathways, whereas the dsRNA-induced CCL5 production was dependent on JNK, but not on the other signal-transducing molecules tested. In addition, the acidic environment of intracellular vesicles was required for the activation of cellular signaling in response to dsRNA. Taken together, these results suggest that the recognition of dsRNA structure selectively induces the CXCL10 and CCL5 responses in microglia through vacuolar pH-dependent activation of NF-kappaB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Kazuo Nakamichi
- Department of Virology 1, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Nakamichi K, Saiki M, Sawada M, Takayama-Ito M, Yamamuro Y, Morimoto K, Kurane I. Rabies virus-induced activation of mitogen-activated protein kinase and NF-kappaB signaling pathways regulates expression of CXC and CC chemokine ligands in microglia. J Virol 2005; 79:11801-12. [PMID: 16140757 PMCID: PMC1212600 DOI: 10.1128/jvi.79.18.11801-11812.2005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Following virus infection of the central nervous system, microglia, the ontogenetic and functional equivalents of macrophages in somatic tissues, act as sources of chemokines, thereby recruiting peripheral leukocytes into the brain parenchyma. In the present study, we have systemically examined the growth characteristics of rabies virus (RV) in microglia and the activation of cellular signaling pathways leading to chemokine expression upon RV infection. In RV-inoculated microglia, the synthesis of the viral genome and the production of virus progenies were significantly impaired, while the expression of viral proteins was observed. Transcriptional analyses of the expression profiles of chemokine genes revealed that RV infection, but not exposure to inactivated virions, strongly induces the expression of CXC chemokine ligand 10 (CXCL10) and CC chemokine ligand 5 (CCL5) in microglia. RV infection triggered the activation of signaling pathways mediated by mitogen-activated protein kinases, including p38, extracellular signal-regulated kinases 1 and 2 (ERK1/2), and c-Jun N-terminal kinase, and nuclear factor kappaB (NF-kappaB). RV-induced expression of CXCL10 and CCL5 was achieved by the activation of p38 and NF-kappaB pathways. In contrast, the activation of ERK1/2 was found to down-regulate CCL5 expression in RV-infected microglia, despite the fact that it was involved in partial induction of CXCL10 expression. Furthermore, NF-kappaB signaling upon RV infection was augmented via a p38-mediated mechanism. Taken together, these results indicate that the strong induction of CXCL10 and CCL5 expression in microglia is precisely regulated by the activation of multiple signaling pathways through the recognition of RV infection.
Collapse
Affiliation(s)
- Kazuo Nakamichi
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Ireland DD, Palian BM, Reiss CS. Interleukin (IL)-12 receptor beta1 or IL-12 receptor beta 2 deficiency in mice indicates that IL-12 and IL-23 are not essential for host recovery from viral encephalitis. Viral Immunol 2005; 18:397-402. [PMID: 16035952 PMCID: PMC1237019 DOI: 10.1089/vim.2005.18.397] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vesicular stomatitis virus (VSV), a negative-sense, single-stranded RNA rhabdovirus, causes acute viral encephalitis when administered intranasally to mice. Interleukin-12 (IL-12) is a key pro-inflammatory cytokine that is produced largely by the antigen presenting cells (APC) and that bridges the innate and acquired immune responses. IL-12 is efficacious in enhancing recovery from VSV infection of the murine central nervous system. This effect is mediated by nitric oxide (NO) produced by the neuronal isoform of nitric oxide synthase (NOS-1), and is independent of the pro-inflammatory cytokines interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). These data implied a link between IL-12 and NOS-1. Here we investigate the role of the IL-12R during VSV pathogenesis, using IL-12R beta2 and IL-12R beta1-deficient mice. We showed that a deficiency in either IL-12R beta2 or IL-12R beta1 had no effect on the outcome of VSV infection of the CNS or on the clearance of VSV from the CNS. Furthermore, these data indicate that IL-23 is not acting redundantly in the absence of IL-12 during VSV-induced encephalitis.
Collapse
Affiliation(s)
- Derek D.C. Ireland
- Department of Neurology, Keck School of Medicine, University of Southern California, 1333 San Pablo St., Mail Code 9153, Los Angeles, CA 91030
| | - Beth M. Palian
- Department of Neurology, Keck School of Medicine, University of Southern California, 1333 San Pablo St., Mail Code 9153, Los Angeles, CA 91030
| | - Carol Shoshkes Reiss
- Department of Biology
- Center for Neural Science
- Department of Microbiology and
- Kaplan Comprehensive Cancer Center, New York University
- Department of Microbiology, Mt. Sinai School of Medicine 100 Washington Square East, mail code 5181 New York, NY 10003-6688
- Corresponding author: Carol Shoshkes Reiss,
, Phone: 212-998-8243, Fax: 212-995-4015
| |
Collapse
|
29
|
Sonobe Y, Yawata I, Kawanokuchi J, Takeuchi H, Mizuno T, Suzumura A. Production of IL-27 and other IL-12 family cytokines by microglia and their subpopulations. Brain Res 2005; 1040:202-7. [PMID: 15804443 DOI: 10.1016/j.brainres.2005.01.100] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2004] [Revised: 01/24/2005] [Accepted: 01/25/2005] [Indexed: 01/17/2023]
Abstract
Production of IL-27 and other IL-12 family cytokines by murine microglia were examined using RT-PCR, real-time RT-PCR and Western blot analysis. We show for the first time that murine microglia produce IL-27 in response to lipopolysaccharide (LPS) and/or interferon-gamma. Primary microglia, but not their cell lines, also induce IL-12 and IL-23 upon above stimulation. Therefore, microglia may play a critical role initiating Th1 responses via producing IL-12 family cytokines in the brain.
Collapse
Affiliation(s)
- Yoshifumi Sonobe
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Chen Z, Duan RS, Concha QH, Wu Q, Mix E, Winblad B, Ljunggren HG, Zhu J. IL-12p35 deficiency alleviates kainic acid-induced hippocampal neurodegeneration in C57BL/6 mice. Neurobiol Dis 2004; 17:171-8. [PMID: 15474355 DOI: 10.1016/j.nbd.2004.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Revised: 06/23/2004] [Accepted: 07/30/2004] [Indexed: 11/29/2022] Open
Abstract
The role of IL-12 in excitotoxic neurodegeneration of brain is largely unknown. To address this issue, we used the model of kainic acid (KA)-induced hippocampal injury in IL-12p35 knockout (KO) mice, a well-characterized model for human neurodegenerative diseases. After KA treatment, hippocampal neurodegeneration was significantly less severe in the IL-12p35 KO mice than in wild-type mice as demonstrated by reduced pathological changes and astrogliosis. One day after KA treatment, levels of F4/80 and CD86 expression on microglia were significantly lower in IL-12p35 KO mice than in wild-type mice analyzed by flow cytometry, indicating that IL-12p35 deficiency resulted in lower levels of microglial activation. Five days after KA treatment, CD86 expression on microglia of wild-type mice was still higher, whereas F4/80 expression in wild-type mice decreased and was similar to that in IL-12p35 KO mice. Because microglial activation is necessary for KA-induced neurodegeneration, the lower level of microglial activation in the absence of IL-12p35 may alleviate hippocampal injury in KO mice. In summary, this study indicates that IL-12 may play a critical role in excitotoxin-induced brain injury.
Collapse
Affiliation(s)
- Zhiguo Chen
- Division of Experimental Geriatrics, Department of Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kawahara K, Mori M, Nakayama H. [NO-induced apoptosis and ER stress in microglia]. Nihon Yakurigaku Zasshi 2004; 124:399-406. [PMID: 15572844 DOI: 10.1254/fpj.124.399] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Nitric Oxide (NO) produced by activated microglia is an important contributor to neuronal damage. NO toxicity is generally thought to be mediated by the DNA damage-p53 pathway or mitochondrial dysfunction. We investigated the mechanism of NO toxicity by using microglial MG5 cells established from p53-deficient mouse. When MG5 cells were exposed to LPS plus IFN-gamma, mRNA and protein for inducible NO synthase (iNOS) were markedly induced and apoptosis occurred. Under these conditions, we found that mRNA and protein for CHOP/GADD153, a C/EBP family transcription factor that is involved in ER stress-induced apoptosis, were induced. These results suggest that NO-induced apoptosis in MG5 cells occurs through the ER stress pathway involving CHOP, but is independent of p53. Overactivation-induced apoptosis may be an essential self-regulatory mechanism for microglia in order to limit bystander killing of vulnerable neurons. On the other hand, recent reports suggest that there may exist two subtypes of microglia at least in the CNS. We found activated rat type-1 microglia induced expression of iNOS and exhibited neurotoxic to rat hippocampal neurons. By contrast, activated type-2 microglia hardly exhibited neurotoxicity in this co-culture system. These results suggest that the two subtype(s) of microglia may regulate differently the inflammatory response in the CNS.
Collapse
Affiliation(s)
- Kohichi Kawahara
- Department of Molecular Cell Function, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | | | | |
Collapse
|
32
|
Abstract
Interleukin (IL)-12, a key cytokine bridging innate and acquired immunity, is efficacious in enhancing recovery from experimental vesicular stomatitis virus (VSV) infection of the mouse central nervous system (CNS). This response is associated with the upregulation of neuronal nitric oxide synthase (NOS-1), independent of IFN-gamma and TNF-alpha. We hypothesized that neurons may respond directly IL-12. Our data are consistent with the expression of a functional IL-12 receptor (IL-12R) by neurons in culture and this receptor-ligand interaction results in the induction of an innate antiviral immune response. N18 cells, which did not express IL-12Rbeta2 were transfected with the IL-12Rbeta2 receptor gene; Koch's postulates were fulfilled, as clones derived from this transfection were reconstituted for IL-12 responsiveness.
Collapse
|
33
|
Kawanokuchi J, Mizuno T, Kato H, Mitsuma N, Suzumura A. Effects of interferon-β on microglial functions as inflammatory and antigen presenting cells in the central nervous system. Neuropharmacology 2004; 46:734-42. [PMID: 14996551 DOI: 10.1016/j.neuropharm.2003.11.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2003] [Revised: 10/14/2003] [Accepted: 11/12/2003] [Indexed: 11/19/2022]
Abstract
Interferon-beta (IFNbeta) reduces exacerbations of the relapsing-remitting form of multiple sclerosis (MS), but the exact mechanisms by which it exerts its beneficial effects are unknown. In this study, we examined the effects of IFNbeta on microglial functions, as either antigen presenting cells or effector cells for inflammatory demyelination. IFNbeta significantly suppressed the expression of class II MHC antigen and the co-stimulatory molecule B7-1 in microglia. It also suppressed microglial IL-12 production and differentiation of myelin oligodendrocyte glycoprotein (MOG)-sensitized T cells into the T helper 1 phenotype, which use microglia as antigen presenting cells. However, IFNbeta significantly and dose-dependently enhanced the production of inflammatory mediators for demyelination, such as TNFalpha, IL-1beta, IL-6, and nitric oxide (NO). The upregulation of inflammatory mediators was effectively suppressed with a phosphodiesterase inhibitor. Thus, IFNbeta may exert its suppressive effects in the induction phase, but not in the effector phase of MS. Side effects of IFNbeta treatment may be due to elevation of pro-inflammatory cytokines, and may be reduced by co-treatment with phosphodiesterase inhibitors.
Collapse
Affiliation(s)
- Jun Kawanokuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | | | | | | | | |
Collapse
|
34
|
Suzumura A, Ito A, Mizuno T. Phosphodiesterase inhibitors suppress IL-12 production with microglia and T helper 1 development. Mult Scler 2004; 9:574-8. [PMID: 14664469 DOI: 10.1191/1352458503ms970oa] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The effects of phosphodiesterase inhibitors (PDEIs) on interleukin (IL)-12 production by microglia, antigen-presenting cells in the central nervous system (CNS), were examined to learn how they affect T cell differentiation in the CNS. PDEIs significantly suppressed the microglial IL-12 production, as determined by reverse transcriptase-polymerase chain reaction for IL-12 p35 and p40 mRNA expression and by an ELISA specific for IL-12 functional heterodimer, p70. In addition, the PDEI ibudilast also suppressed interferon-gamma, but not IL-4 or IL-10, production by myelin oligodendrocyte glycoprotein (MOG)-specific T cells reactivated with MOG in the presence of microglia. Thus, PDEIs may also suppress differentiation of T helper 1 (Th1) in the CNS. PDEIs can be of use for future therapeutic strategy to treat Th1-mediated diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Akio Suzumura
- Department of Neuroimmunology, Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8601, Japan.
| | | | | |
Collapse
|
35
|
Li J, Gran B, Zhang GX, Ventura ES, Siglienti I, Rostami A, Kamoun M. Differential expression and regulation of IL-23 and IL-12 subunits and receptors in adult mouse microglia. J Neurol Sci 2004; 215:95-103. [PMID: 14568135 DOI: 10.1016/s0022-510x(03)00203-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
IL-23 and IL-12 are functionally related heterodimeric cytokines that share the IL-12p40 subunit. IL-23 and IL-12 function through heterodimeric receptors, which share the IL-12Rbeta1 subunit. Production of IL-23, a heterodimer of IL-12p40 and IL-23p19, by CNS antigen-presenting cells (APC) is critical for susceptibility to experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis (MS). We report that the expression of IL-23p19 mRNA is highly induced by stimulation with IFN-gamma and LPS in adult mouse microglia and a microglia cell line, EOC13. Expression of the IL-12R subunits, IL-12Rbeta1 and IL-12Rbeta2, is upregulated in both microglia and splenic macrophages upon stimulation with LPS or IFN-gamma and LPS, whereas the IL-23R subunit is upregulated only in macrophages. In EAE, an early peak of IL-23p19 mRNA expression is found in CD11b(+) CNS APC, compared with peripheral macrophages. In contrast, IL-12p40 and IL-12p35 mRNA maximum levels in the CNS are detected at peak of disease. The expression of IL-12p35 mRNA is more sustained than that of IL-12p40 and IL-23p19. Thus, IL-23 produced by CNS microglia/macrophages may contribute to the early induction of EAE. In the CNS, IL-23 may preferentially target infiltrating mononuclear cells, which upregulate IL-23R, rather than parenchymal microglia.
Collapse
Affiliation(s)
- Jifen Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Nagata N, Kitaura H, Yoshida N, Nakayama K. Inhibition of RANKL-induced osteoclast formation in mouse bone marrow cells by IL-12: involvement of IFN-gamma possibly induced from non-T cell population. Bone 2003; 33:721-32. [PMID: 14555278 DOI: 10.1016/s8756-3282(03)00213-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IL-12 was shown to have the potential to inhibit osteoclast formation in mouse bone marrow cells treated with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-kappaB ligand (RANKL). When bone marrow macrophages (BMM) were used as osteoclast precursors, IL-12 failed to inhibit M-CSF/RANKL-induced osteoclast formation from BMM. In coculture experiments using transwells, IL-12 did inhibit osteoclast formation from BMM cocultured with whole bone marrow cells. These results indicated that IL-12 indirectly affected M-CSF/RANKL-induced osteoclastogenesis in bone marrow cells and that the inhibition of IL-12 on osteoclast formation was caused by a humoral factor from bone marrow cells treated with IL-12. Experiments with anti-interferon (IFN)-gamma antibody and bone marrow cells from IFN-gamma receptor knockout mice revealed that IFN-gamma might be involved in the inhibition of osteoclast formation in this system. The expression of osteoprotegerin mRNA in bone marrow cells was not affected by treatment with IL-12. The inhibitory effect of IL-12 on osteoclast formation was also seen in the T cell-depleted bone marrow cells of normal mice and the whole bone marrow cells of athymic nude mice, while the inhibitory effect of IL-12 was partially suppressed in the B cell-depleted bone marrow cells. The inhibitory effect of IL-12 on M-CSF/RANKL-induced osteoclastogenesis was not accompanied with cell death, in contrast with our previous finding that the inhibitory effect of IL-12 on M-CSF/TNF-alpha-induced osteoclastogenesis is attributable to Fas and FasL-mediated apoptosis.
Collapse
Affiliation(s)
- Noriko Nagata
- Division of Orthodontic and Biomedical Engineering, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | | | | | | |
Collapse
|
37
|
Abstract
Cytokines constitute a significant portion of the immuno- and neuromodulatory messengers that can be released by activated microglia. By virtue of potent effects on resident and invading cells, microglial cyto- and chemokines regulate innate defense mechanisms, help the initiation and influence the type of immune responses, participate in the recruitment of leukocytes to the CNS, and support attempts of tissue repair and recovery. Microglia can also receive cyto- and chemokine signals as part of auto- and paracrine communications with astrocytes, neurons, the endothelium, and leukocyte infiltrates. Strong responses and modulatory influences can be demonstrated, adding to the emerging view that microglial behavior is highly dependent on the (cytokine) environment and that reactions to a challenge may vary with the stimulation context. In principle, microglial activation aims at CNS protection. However, failed microglial engagement due to excessive or sustained activation could significantly contribute to acute and chronic neuropathologies. Dysregulation of microglial cytokine production could thereby promote harmful actions of the defense mechanisms, result in direct neurotoxicity, as well as disturb neural cell functions as they are sensitive to cytokine signaling.
Collapse
Affiliation(s)
- Uwe-Karsten Hanisch
- Department of Cellular Neurosciences, Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
- University of Applied Sciences Lausitz, Senftenberg, Germany
| |
Collapse
|
38
|
Abstract
Interleukin (IL)-12p75 is a heterodimeric cytokine composed of the product of two different genes that specify p35 and p40 subunits. The prevailing view is that IL-12 acts as a proinflammatory cytokine that bridges the innate and adaptive immune responses and skews T-cell reactivity toward a TH1 cytokine pattern. Though the terms IL-12 and IL-12p40 are often used interchangeably, and measurements of the p40 chain are often interpreted as measurements of the intact p75 heterodimer, such interchangeable usage may be incorrect. In the following discussion, I will delineate an alternative hypothesis for the roles of the p40 and p75 proteins, suggesting specifically, that: (1) in vivo, secretion of free p40 precedes that of p75 in response to pathogens; (2) induction of p40 is a T-independent response by antigen presenting cells (APCs) to early host-pathogen interactions; and (3) IL-12p75 is a late product, whose induction requires T-dependent signals. It is made as a result, rather than as a cause, of TH1 differentiation. Thus, it is the p40 protein, either alone or paired with other polypeptides, rather than p75, that acts as an interface between the innate and adaptive immune responses.
Collapse
Affiliation(s)
- K Abdi
- Ghost Laboratory, Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville Pike, Bethesda, MD 20892-0420, USA.
| |
Collapse
|
39
|
Abstract
During the past decade, mechanisms involved in the immune surveillance of the central nervous system (CNS) have moved to the forefront of neuropathological research mainly because of the recognition that most neurological disorders involve activation and, possibly, dysregulation of microglia, the intrinsic macrophages of the CNS. Increasing evidence indicates that, in addition to their well-established phagocytic function, microglia may also participate in the regulation of non specific inflammation as well as adaptive immune responses. This article focuses on the signals regulating microglia innate immune functions, the role of microglia in antigen presentation, and their possible involvement in the development of CNS immunopathology.
Collapse
Affiliation(s)
- F Aloisi
- Neurophysiology Unit, Laboratory of Organ and System Pathophysiology, Istituto Superiore di Sanità, Roma, Italy.
| |
Collapse
|
40
|
Abstract
Microglia are the resident tissue macrophages of the central nervous system (CNS) parenchyma and are key players in the initiation of an inflammatory response. Microglia rapidly transform from a resting to an activated morphology in response to a variety of disease states. However, they can also be the target of infections, as in the case of HIV. Many of the effector properties of microglia can be attributed to the array of substances they secrete in response to stimuli such as bacterial lipopolysaccharide, cytokines, and chemokines. The products of activated microglia include: cytokines (pro- and anti-inflammatory), chemokines, nitric oxide, superoxide radicals, and proteases. Furthermore, microglia have the ability to present antigen to T cells, migrate in response to chemotactic stimuli, and phagocytose cell debris. This report focuses on the immunomodulatory functions of microglia, with particular attention to chemokines, and highlights their pivotal role in the CNS.
Collapse
Affiliation(s)
- A K Cross
- Division of Biomedical Sciences, Sheffield Hallam University, United Kingdom
| | | |
Collapse
|
41
|
Katoh Y, Niimi M, Yamamoto Y, Kawamura T, Morimoto-Ishizuka T, Sawada M, Takemori H, Yamatodani A. Histamine production by cultured microglial cells of the mouse. Neurosci Lett 2001; 305:181-4. [PMID: 11403935 DOI: 10.1016/s0304-3940(01)01835-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We previously reported that cells other than mast cells or neurons could synthesize histamine in response to lipopolysaccharide (LPS) or interleukin 1beta in the rat brain. To identify the responsible cells, we examined histidine decarboxylase (HDC) activity and the expression of HDC mRNA in GMI 6-3 mouse microglial cells. Both the activity and mRNA for HDC in GMI 6-3 cells were induced by LPS treatment, and the induction was sensitive to calmodulin-dependent kinase II inhibitor, KN62. These findings indicate that microglia is a third cell type producing histamine in the brain.
Collapse
Affiliation(s)
- Y Katoh
- Department of Medical Physics, School of Allied Health Sciences, Faculty of Medicine, Osaka University, 1-7 Yamadaoka, Suita, 565-0871, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Prinz M, Häusler KG, Kettenmann H, Hanisch U. beta-adrenergic receptor stimulation selectively inhibits IL-12p40 release in microglia. Brain Res 2001; 899:264-70. [PMID: 11311890 DOI: 10.1016/s0006-8993(01)02174-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The cytokine interleukin-12 (IL-12) is mainly produced in response to bacterial or parasitic infections. We examined the capacity of mouse brain microglia to release IL-12 forms upon challenge with bacterial lipopolysaccharide (LPS) and studied its modulation by sympathomimetics. LPS evoked the release of IL-12p40 whereas the heterodimeric form, IL-12p70 was virtually undetectable. Sympathomimetics such as salbutamol dose-dependently inhibited IL-12p40 release, whereas the production of IL-6, TNFalpha and MIP-1alpha was only marginally influenced. The inhibitory effect of salbutamol could be abolished by beta-antagonists, such as oxprenolol. The cAMP-elevating agent forskolin could mimic the effects of beta-agonists, indicating that IL-12p40 release inhibition involves intracellular cAMP accumulation. While microglial IL-12p40 may play a role in the regulation of IL-12p70 bioactivity, microglial release is itself modulated by IL-12p70. Recombinant IL-12p70 was found to enhance the LPS-evoked release of MIP-1alpha and to have a biphasic effect on both TNFalpha and MIP-1alpha with release augmentation at lower and attenuation at higher doses. Finally, no functional correlation was found between the release of IL-12p40 and the induction of Kv1.3 potassium channels, another marker of microglial activation. Taken together, beta(2)-adrenoreceptor-mediated effects on microglial cyto- and chemokine release via cAMP accumulation could modulate inflammatory cascades during bacterial infections.
Collapse
Affiliation(s)
- M Prinz
- Department of Cellular Neurosciences, Max Delbrück Center (MDC) for Molecular Medicine, Robert-Rössle-Strasse 10, D-13092, Berlin, Germany.
| | | | | | | |
Collapse
|
43
|
Pahan K, Sheikh FG, Liu X, Hilger S, McKinney M, Petro TM. Induction of nitric-oxide synthase and activation of NF-kappaB by interleukin-12 p40 in microglial cells. J Biol Chem 2001; 276:7899-905. [PMID: 11110796 PMCID: PMC2041874 DOI: 10.1074/jbc.m008262200] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interleukin-12 (IL-12) is composed of two different subunits, p40 and p35. Expression of p40 mRNA but not that of p35 mRNA in excessive amount in the central nervous system of patients with multiple sclerosis (MS) suggests that IL-12 p40 may have a role in the pathogenesis of the disease. However, the mode of action of p40 is completely unknown. Because nitric oxide produced from the induction of nitric-oxide synthase (iNOS) also plays a vital role in the pathophysiology of MS, the present study was undertaken to explore the role of p40 in the induction of NO production and the expression of iNOS in microglia. Both IL-12 and p40(2), the p40 homodimer, dose-dependently induced the production of NO in BV-2 microglial cells. This induction of NO production was accompanied by an induction of iNOS protein and mRNA. Induction of NO production by the expression of mouse p40 cDNA but not that of the mouse p35 cDNA suggests that the p40 but not the p35 subunit of IL-12 is involved in the expression of iNOS. In addition to BV-2 glial cells, p40(2) also induced the production of NO in mouse primary microglia and peritoneal macrophages. However, both IL-12 and p40(2) were unable to induce the production of NO in mouse primary astrocytes. Because activation of NF-kappaB is important for the expression of iNOS, we investigated the effect of p40(2) on the activation of NF-kappaB. Induction of the DNA binding as well as the transcriptional activity of NF-kappaB by p40(2) and inhibition of p40(2)-induced expression of iNOS by SN50, a cell-permeable peptide carrying the nuclear localization sequence of p50 NF-kappaB, but not by SN50M, a nonfunctional peptide mutant, suggests that p40(2) induces the expression of iNOS through the activation of NF-kappaB. This study delineates a novel role of IL-12 p40 in inducing the expression of iNOS in microglial cells, which may participate in the pathogenesis of neuroinflammatory diseases.
Collapse
Affiliation(s)
- K Pahan
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, Nebraska 68583, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Fantuzzi L, Puddu P, Varano B, Del Cornò M, Belardelli F, Gessani S. IFN‐α and IL‐18 exert opposite regulatory effects on the IL‐12 receptor expression and IL‐12‐induced IFN‐γ production in mouse macrophages: novel pathways in the regulation of the inflammatory response of macrophages. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.5.707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | - Patrizia Puddu
- Immunology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | | | | | | |
Collapse
|
45
|
Kanzawa T, Sawada M, Kato K, Yamamoto K, Mori H, Tanaka R. Differentiated regulation of allo-antigen presentation by different types of murine microglial cell lines. J Neurosci Res 2000; 62:383-8. [PMID: 11054807 DOI: 10.1002/1097-4547(20001101)62:3<383::aid-jnr8>3.0.co;2-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We established granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent murine microglial clones and investigated the immune properties of four individual clones. All four clones expressed MHC class I and CD54 (ICAM-1) at similar levels. The 5-2, Ra2, and 6-3 clones expressed CD80 (B7-1), CD86 (B7-2), and MHC class II at low, medium, and high levels, respectively. Only the 6-3 clone expressed CD40. Generally, the levels of co-stimulation and CD 40 signals had a profound effect on the response to antigens. The 5-2, Ra2, and 6-3 clones, however, stimulated allogenic T-cell proliferation to the same extent or less compared to spleen cells. Although the 6-1 clone expressed co-stimulatory and MHC molecules at levels similar to Ra2, it suppressed allogenic T-cell proliferation, unlike Ra2. Thus, allo-antigen presentation by microglial clones was not correlated with the expression of CD40 and co-stimulatory molecules. When microglial clones were fixed with paraformaldehyde, they enhanced IL-2-dependent T-cell proliferation according to the level of their expression of co-stimulatory molecules. Furthermore, conditioned medium from the 6-1 clone inhibited the T-cell response to allo-antigen. This indicates that some factor(s) derived from a microglial subtype may play an important role in the regulation of T-cell proliferation in addition to the molecules involved in antigen presentation. Moreover, these results also suggest that there may be specialized subtypes of microglia that regulate the immune response in the CNS.
Collapse
Affiliation(s)
- T Kanzawa
- Department of Neurosurgery, Niigata Brain Research Institute, Niigata University, Niigata, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Liu J, Chen M, Wang X. Calcitonin gene-related peptide inhibits lipopolysaccharide-induced interleukin-12 release from mouse peritoneal macrophages, mediated by the cAMP pathway. Immunology 2000; 101:61-7. [PMID: 11012754 PMCID: PMC2327047 DOI: 10.1046/j.1365-2567.2000.00082.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previously we showed that calcitonin gene-related peptide (CGRP), a neuropeptide, inhibited lipopolysaccharide (LPS)-induced tumour necrosis factor-alpha (TNF-alpha) production and increased interleukin (IL)-6 release at low concentrations via activation of the cAMP pathway in mouse peritoneal macrophages (Mphi). In this study we examined whether CGRP could modulate IL-12 release from mouse peritoneal Mphi, and if so, what signal transduction pathway was involved. Mphi were obtained from the peritoneal exudate of male BALB/c mice. The cells were plated on culture dishes at a density of 5 x 105 cells per well and allowed to adhere for 2 hr. After incubation for 24 hr, the Mphi were cultured with 0.1 microg/ml of LPS, alone or together with CGRP (1-1000 nM) for 24 hr. The amount of IL-12 in the cell medium was measured by enzyme-linked immunosorbent assay (ELISA). The results showed that CGRP attenuated LPS-induced IL-12 release in a concentration-dependent manner. Production of IL-12 was decreased from 95.9+/-4.6 to 73.4+/-5.7 pg/ml by 100 nM CGRP. The two cAMP phosphodiesterase (PDE) inhibitors, 3-isobutyl-1-methyl-xanthine (IBMX) and rolipram, significantly potentiated the CGRP response, and the level of IL-12 was further decreased by 28% and 47%, respectively. However, CGRP had no effect on IL-12 production from unstimulated Mphi. The LPS-induced IL-12 release from Mphi could also be reduced by forskolin, an activator of adenylate cyclase, and 8-Br-cAMP, an analogue of cAMP. Using the reverse transcription-polymerase chain reaction (RT-PCR), we found that CGRP also decreased the LPS-induced IL-12 p40 mRNA levels. Furthermore, pretreatment with H89 (0.1 microM or 1 microM), an inhibitor of cAMP-dependent protein kinase, diminished CGRP effects, IL-12 production and gene expression. These data suggest that LPS-induced IL-12 release and gene expression were attenuated by CGRP via an activated cAMP-protein kinase A (PKA) pathway in mouse peritoneal Mphi.
Collapse
Affiliation(s)
- J Liu
- Institute of Vascular Medicine, The Third Hospital, Beijing Medical University, Beijing, China
| | | | | |
Collapse
|