1
|
Svenningsson AL, Bocancea DI, Stomrud E, van Loenhoud A, Barkhof F, Mattsson-Carlgren N, Palmqvist S, Hansson O, Ossenkoppele R. Biological mechanisms of resilience to tau pathology in Alzheimer's disease. Alzheimers Res Ther 2024; 16:221. [PMID: 39396028 PMCID: PMC11470552 DOI: 10.1186/s13195-024-01591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND In Alzheimer's disease (AD), the associations between tau pathology and brain atrophy and cognitive decline are well established, but imperfect. We investigate whether cerebrospinal fluid (CSF) biomarkers of biological processes (vascular, synaptic, and axonal integrity, neuroinflammation, neurotrophic factors) explain the disconnection between tau pathology and brain atrophy (brain resilience), and tau pathology and cognitive decline (cognitive resilience). METHODS We included 428 amyloid positive participants (134 cognitively unimpaired (CU), 128 with mild cognitive impairment (MCI), 166 with AD dementia) from the BioFINDER-2 study. At baseline, participants underwent tau positron emission tomography (tau-PET), magnetic resonance imaging (MRI), cognitive testing, and lumbar puncture. Longitudinal data were available for MRI (mean (standard deviation) follow-up 26.4 (10.7) months) and cognition (25.2 (11.4) months). We analysed 18 pre-selected CSF proteins, reflecting vascular, synaptic, and axonal integrity, neuroinflammation, and neurotrophic factors. Stratifying by cognitive status, we performed linear mixed-effects models with cortical thickness (brain resilience) and global cognition (cognitive resilience) as dependent variables to assess whether the CSF biomarkers interacted with tau-PET levels in its effect on cortical atrophy and cognitive decline. RESULTS Regarding brain resilience, interaction effects were observed in AD dementia, with vascular integrity biomarkers (VEGF-A (βinteraction = -0.009, pFDR = 0.047) and VEGF-B (βinteraction = -0.010, pFDR = 0.037)) negatively moderating the association between tau-PET signal and atrophy. In MCI, higher NfL levels were associated with more longitudinal cortical atrophy (β = -0.109, pFDR = 0.033) and lower baseline cortical thickness (β = -0.708, pFDR = 0.033) controlling for tau-PET signal. Cognitive resilience analyses in CU revealed interactions with tau-PET signal for inflammatory (GFAP, IL-15; βinteraction -0.073--0.069, pFDR 0.001-0.045), vascular (VEGF-A, VEGF-D, PGF; βinteraction -0.099--0.063, pFDR < 0.001-0.046), synaptic (14-3-3ζ/δ; βinteraction = -0.092, pFDR = 0.041), axonal (NfL; βinteraction = -0.079, pFDR < 0.001), and neurotrophic (NGF; βinteraction = 0.091, pFDR < 0.001) biomarkers. In MCI higher NfL levels (βmain = -0.690, pFDR = 0.025) were associated with faster cognitive decline independent of tau-PET signal. CONCLUSIONS Biomarkers of co-existing pathological processes, in particular vascular pathology and axonal degeneration, interact with levels of tau pathology on its association with the downstream effects of AD pathology (i.e. brain atrophy and cognitive decline). This indicates that vascular pathology and axonal degeneration could impact brain and cognitive resilience.
Collapse
Affiliation(s)
- Anna L Svenningsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden.
| | - Diana I Bocancea
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Anita van Loenhoud
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Center for Medical Image Computing, University College London, London, WC1N 3BG, UK
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Department of Neurology, Skåne University Hospital, 211 84, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, 211 46, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Haddad FL, Patel SV, Doornaert EE, De Oliveira C, Allman BL, Baines KJ, Renaud SJ, Schmid S. Interleukin 15 modulates the effects of poly I:C maternal immune activation on offspring behaviour. Brain Behav Immun Health 2022; 23:100473. [PMID: 35668725 PMCID: PMC9166394 DOI: 10.1016/j.bbih.2022.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 01/21/2023] Open
Abstract
Maternal infections during pregnancy are linked with an increased risk for disorders like Autism Spectrum Disorder and schizophrenia in the offspring. Although precise mechanisms are still unclear, clinical and preclinical evidence suggest a strong role for maternal immune activation (MIA) in the neurodevelopmental disruptions caused by maternal infection. Previously, studies using the Polyinosinic:Polycytidylic (Poly I:C) MIA preclinical model showed that cytokines like Interleukin 6 (Il6) are important mediators of MIA's effects. In this study, we hypothesized that Il15 may similarly act as a mediator of Poly I:C MIA, given its role in the antiviral immune response. To test this hypothesis, we induced Poly I:C MIA at gestational day 9.5 in wildtype (WT) and Il15−/− rat dams and tested their offspring in adolescence and adulthood. Poly I:C MIA and Il15 knockout produced both independent and synergistic effects on offspring behaviour. Poly I:C MIA decreased startle reactivity in adult WT offspring but resulted in increased adolescent anxiety and decreased adult locomotor activity in Il15−/− offspring. In addition, Poly I:C MIA led to genotype-independent effects on locomotor activity and prepulse inhibition. Finally, we showed that Il15−/− offspring exhibit distinct phenotypes that were unrelated to Poly I:C MIA including altered startle reactivity, locomotion and signal transduction in the auditory brainstem. Overall, our findings indicate that the lack of Il15 can leave offspring either more or less susceptible to Poly I:C MIA, depending on the phenotype in question. Future studies should examine the contribution of fetal versus maternal Il15 in MIA to determine the precise developmental mechanisms underlying these changes. Poly I:C MIA decreases startle reactivity in adult WT but not Il15−/− offspring. Il15−/− offspring exposed to Poly I:C MIA show altered PPI and open field exploration. Il15−/− rats exhibit distinct behavioural phenotypes independent from MIA.
Collapse
|
3
|
Zveik O, Rechtman A, Haham N, Adini I, Canello T, Lavon I, Brill L, Vaknin-Dembinsky A. Sera of Neuromyelitis Optica Patients Increase BID-Mediated Apoptosis in Astrocytes. Int J Mol Sci 2022; 23:ijms23137117. [PMID: 35806122 PMCID: PMC9266359 DOI: 10.3390/ijms23137117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Neuromyelitis optica (NMO) is a rare disease usually presenting with bilateral or unilateral optic neuritis with simultaneous or sequential transverse myelitis. Autoantibodies directed against aquaporin-4 (AQP4-IgG) are found in most patients. They are believed to cross the blood−brain barrier, target astrocytes, activate complement, and eventually lead to astrocyte destruction, demyelination, and axonal damage. However, it is still not clear what the primary pathological event is. We hypothesize that the interaction of AQP4-IgG and astrocytes leads to DNA damage and apoptosis. We studied the effect of sera from seropositive NMO patients and healthy controls (HCs) on astrocytes’ immune gene expression and viability. We found that sera from seropositive NMO patients led to higher expression of apoptosis-related genes, including BH3-interacting domain death agonist (BID), which is the most significant differentiating gene (p < 0.0001), and triggered more apoptosis in astrocytes compared to sera from HCs. Furthermore, NMO sera increased DNA damage and led to a higher expression of immunological genes that interact with BID (TLR4 and NOD-1). Our findings suggest that sera of seropositive NMO patients might cause astrocytic DNA damage and apoptosis. It may be one of the mechanisms implicated in the primary pathological event in NMO and provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Omri Zveik
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Ariel Rechtman
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Nitzan Haham
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Irit Adini
- Department of Surgery, Harvard Medical School, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA;
| | - Tamar Canello
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Leslie and Michael Gaffin Center for Neuro-Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Iris Lavon
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Leslie and Michael Gaffin Center for Neuro-Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Livnat Brill
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Correspondence: ; Tel.: +972-2-677-7741
| |
Collapse
|
4
|
Sanmarco LM, Polonio CM, Wheeler MA, Quintana FJ. Functional immune cell-astrocyte interactions. J Exp Med 2021; 218:212503. [PMID: 34292315 PMCID: PMC8302447 DOI: 10.1084/jem.20202715] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Astrocytes are abundant glial cells in the central nervous system (CNS) that control multiple aspects of health and disease. Through their interactions with components of the blood–brain barrier (BBB), astrocytes not only regulate BBB function, they also sense molecules produced by peripheral immune cells, including cytokines. Here, we review the interactions between immune cells and astrocytes and their roles in health and neurological diseases, with a special focus on multiple sclerosis (MS). We highlight known pathways that participate in astrocyte crosstalk with microglia, NK cells, T cells, and other cell types; their contribution to the pathogenesis of neurological diseases; and their potential value as therapeutic targets.
Collapse
Affiliation(s)
- Liliana M Sanmarco
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Carolina M Polonio
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Neuroimmune Interactions Laboratory, Immunology Department, Instituto de Ciências Biomédicas IV, University of São Paulo, São Paulo, Brazil
| | - Michael A Wheeler
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Francisco J Quintana
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
5
|
Wu L, Ji NN, Wang H, Hua JY, Sun GL, Chen PP, Hua R, Zhang YM. Domino Effect of Interleukin-15 and CD8 T-Cell-Mediated Neuronal Apoptosis in Experimental Traumatic Brain Injury. J Neurotrauma 2021; 38:1450-1463. [PMID: 30430911 DOI: 10.1089/neu.2017.5607] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The effects of local factors on activation of immune cells infiltrating the central nervous system (CNS) in a rat model of traumatic brain injury (TBI) remain elusive. The cytokine, interleukin (IL)-15, is crucial for development and activation of CD8 T lymphocytes, a prominent lymphocytic population present in TBI lesions. We investigated whether IL-15 originates from astrocytes and whether IL-15 can evoke the CD8 T-lymphocyte response in TBI. We observed that astrocytes were activated in a rat model of TBI and that IL-15 was overexpressed on the surface of astrocytes. Further, CD8 T lymphocytes infiltrating TBI lesions colocalized with IL-15-expressing astrocytes. Activated CD8 T lymphocytes released granzyme B (Gra-b), which, in turn, activated caspase-3-induced poly(ADP-ribose) polymerase cleavage and, ultimately, neuronal apoptosis. Conversely, inhibition of astrocyte activation by pre-treatment with the specific inhibitor, fluorocitrate (FC), that reduces carbon flux through the Krebs cycle in astrocytes resulted in improved neurological function and memory. FC pre-treatment was also associated with downregulated IL-15 expression and CD8 T-cell activation as well as decreased levels of neuronal apoptosis, suggesting that IL-15 initiated a domino effect toward apoptosis. In contrast, rats pre-treated with recombinant rat IL-15 showed upregulated CD8 T-cell numbers and Gra-b levels, in addition to induction of neuronal apoptosis. Together, our results indicated that IL-15 could induce neuronal apoptosis by enhancing CD8 T-cell function in a rat model of TBI.
Collapse
Affiliation(s)
- Liang Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesiology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, China
| | - Ning-Ning Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesiology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, China
| | - Hang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jing-Yu Hua
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Guo-Lin Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Pan-Pan Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Rong Hua
- Faculty of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, China.,Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
6
|
Flores-Aguilar L, Iulita MF, Kovecses O, Torres MD, Levi SM, Zhang Y, Askenazi M, Wisniewski T, Busciglio J, Cuello AC. Evolution of neuroinflammation across the lifespan of individuals with Down syndrome. Brain 2021; 143:3653-3671. [PMID: 33206953 DOI: 10.1093/brain/awaa326] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/18/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Epidemiological and experimental studies suggest that a disease-aggravating neuroinflammatory process is present at preclinical stages of Alzheimer's disease. Given that individuals with Down syndrome are at increased genetic risk of Alzheimer's disease and therefore develop the spectrum of Alzheimer's neuropathology in a uniform manner, they constitute an important population to study the evolution of neuroinflammation across the Alzheimer's continuum. Therefore, in this cross-sectional study, we characterized the brain inflammatory profile across the lifespan of individuals with Down syndrome. Microglial morphology and inflammatory cytokine expression were analysed by immunohistochemistry and electrochemiluminescent-based immunoassays in the frontal cortex from foetuses to adults with Down syndrome and control subjects (16 gestational weeks to 64 years), totalling 127 cases. Cytokine expression in mixed foetal primary cultures and hippocampus of adults with Down syndrome, as well as the effects of sex on cytokine expression were also analysed. A higher microglial soma size-to-process length ratio was observed in the frontal cortex of children and young adults with Down syndrome before the development of full-blown Alzheimer's pathology. Moreover, young adults with Down syndrome also displayed increased numbers of rod-like microglia. Increased levels of interleukin-8 and interleukin-10 were observed in children with Down syndrome (1-10 years; Down syndrome n = 5, controls n = 10) and higher levels of interleukin-1β, interleukin-1α, interleukin-6, interleukin-8, interleukin-10, interleukin-15, eotaxin-3, interferon gamma-induced protein 10, macrophage-derived chemokine, and macrophage inflammatory protein-beta, were found in young adults with Down syndrome compared to euploid cases (13-25 years, Down syndrome n = 6, controls n = 24). Increased cytokine expression was also found in the conditioned media of mixed cortical primary cultures from second trimester foetuses with Down syndrome (Down syndrome n = 7, controls n = 7). Older adults with Down syndrome (39-68 years, Down syndrome n = 22, controls n = 16) displayed reduced levels of interleukin-10, interleukin-12p40, interferon-gamma and tumour necrosis factor-alpha. Microglia displayed larger somas and shorter processes. Moreover, an increase in dystrophic microglia and rod-like microglia aligning to neurons harbouring tau pathology were also observed. Sex stratification analyses revealed that females with Down syndrome had increased interleukin-6 and interleukin-8 levels compared to males with Down syndrome. Finally, multivariate projection methods identified specific cytokine patterns among individuals with Down syndrome. Our findings indicate the presence of an early and evolving neuroinflammatory phenotype across the lifespan in Down syndrome, a knowledge that is relevant for the discovery of stage-specific targets and for the design of possible anti-inflammatory trials against Alzheimer's disease in this population.
Collapse
Affiliation(s)
| | - M Florencia Iulita
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Alzheimer-Down Unit, Fundación Catalana Síndrome de Down, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Olivia Kovecses
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Maria D Torres
- Department of Neurobiology and Behavior, UCI-MIND Institute, and Center for the Neurobiology of Learning and Memory, University of California, Irvine, USA
| | - Sarah M Levi
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Yian Zhang
- Division of Biostatistics, New York University, Grossman School of Medicine, New York, USA
| | | | - Thomas Wisniewski
- Departments of Neurology, Pathology, and Psychiatry, Center for Cognitive Neurology, New York University, Grossman School of Medicine, New York, USA
| | - Jorge Busciglio
- Department of Neurobiology and Behavior, UCI-MIND Institute, and Center for the Neurobiology of Learning and Memory, University of California, Irvine, USA
| | - A Claudio Cuello
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Visiting Professor, Department of Pharmacology, Oxford University, Oxford, UK
| |
Collapse
|
7
|
Bottai D, Adami R, Paroni R, Ghidoni R. Brain Cancer-Activated Microglia: A Potential Role for Sphingolipids. Curr Med Chem 2020; 27:4039-4061. [PMID: 31057101 DOI: 10.2174/0929867326666190506120213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/27/2018] [Accepted: 01/12/2019] [Indexed: 02/06/2023]
Abstract
Almost no neurological disease exists without microglial activation. Microglia has exert a pivotal role in the maintenance of the central nervous system and its response to external and internal insults. Microglia have traditionally been classified as, in the healthy central nervous system, "resting", with branched morphology system and, as a response to disease, "activated", with amoeboid morphology; as a response to diseases but this distinction is now outmoded. The most devastating disease that hits the brain is cancer, in particular glioblastoma. Glioblastoma multiforme is the most aggressive glioma with high invasiveness and little chance of being surgically removed. During tumor onset, many brain alterations are present and microglia have a major role because the tumor itself changes microglia from the pro-inflammatory state to the anti-inflammatory and protects the tumor from an immune intervention. What are the determinants of these changes in the behavior of the microglia? In this review, we survey and discuss the role of sphingolipids in microglia activation in the progression of brain tumors, with a particular focus on glioblastoma.
Collapse
Affiliation(s)
- Daniele Bottai
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Raffaella Adami
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Rita Paroni
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Riccardo Ghidoni
- Department of Health Sciences, University of Milan, Milan, Italy,Aldo Ravelli Research Center, Milan, Italy
| |
Collapse
|
8
|
Lenk L, Alsadeq A, Schewe DM. Involvement of the central nervous system in acute lymphoblastic leukemia: opinions on molecular mechanisms and clinical implications based on recent data. Cancer Metastasis Rev 2020; 39:173-187. [PMID: 31970588 PMCID: PMC7098933 DOI: 10.1007/s10555-020-09848-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. One of the major clinical challenges is adequate diagnosis and treatment of central nervous system (CNS) involvement in this disease. Intriguingly, there is little solid evidence on the mechanisms sustaining CNS disease in ALL. Here, we present and discuss recent data on this topic, which are mainly derived from preclinical model systems. We thereby highlight sites and routes of leukemic CNS infiltration, cellular features promoting infiltration and survival of leukemic cells in a presumably hostile niche, and dormancy as a potential mechanism of survival and relapse in CNS leukemia. We also focus on the impact of ALL cytogenetic subtypes on features associated with a particular CNS tropism. Finally, we speculate on new perspectives in the treatment of ALL in the CNS, including ideas on the impact of novel immunotherapies.
Collapse
Affiliation(s)
- Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ameera Alsadeq
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Denis M Schewe
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
9
|
Shi SX, Li YJ, Shi K, Wood K, Ducruet AF, Liu Q. IL (Interleukin)-15 Bridges Astrocyte-Microglia Crosstalk and Exacerbates Brain Injury Following Intracerebral Hemorrhage. Stroke 2020; 51:967-974. [PMID: 32019481 DOI: 10.1161/strokeaha.119.028638] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Microglia are among the first cells to respond to intracerebral hemorrhage (ICH), but the mechanisms that underlie their activity following ICH remain unclear. IL (interleukin)-15 is a proinflammatory cytokine that orchestrates homeostasis and the intensity of the immune response following central nervous system inflammatory events. The goal of this study was to investigate the role of IL-15 in ICH injury. Methods- Using brain slices of patients with ICH, we determined the presence and cellular source of IL-15. A transgenic mouse line with targeted expression of IL-15 in astrocytes was generated to determine the role of astrocytic IL-15 in ICH. The expression of IL-15 was controlled by a glial fibrillary acidic protein promoter (GFAP-IL-15tg). ICH was induced by intraparenchymal injection of collagenase or autologous blood. Results- In patients with ICH and wild-type mice subjected to experimental ICH, we found a significant upregulation of IL-15 in astrocytes. In GFAP-IL-15tg mice, we found that astrocyte-targeted expression of IL-15 exacerbated brain edema and neurological deficits following ICH. This aggravated ICH injury in GFAP-IL-15tg mice is accompanied by increased microglial accumulation in close proximity to astrocytes in perihematomal tissues. Additionally, microglial expression of CD86, IL-1β, and TNF-α is markedly increased in GFAP-IL-15tg mice following ICH. Furthermore, depletion of microglia using a colony stimulating factor 1 receptor inhibitor diminishes the exacerbation of ICH injury in GFAP-IL-15tg mice. Conclusions- Our findings identify IL-15 as a mediator of the crosstalk between astrocytes and microglia that exacerbates brain injury following ICH.
Collapse
Affiliation(s)
- Samuel X Shi
- From the Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (S.X.S., K.S., K.W., A.F.D., Q.L.).,Interdisciplinary Graduate Program in Neuroscience, Arizona State University, Tempe (S.X.S.)
| | - Yu-Jing Li
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix (Y.-J.L.)
| | - Kaibin Shi
- From the Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (S.X.S., K.S., K.W., A.F.D., Q.L.)
| | - Kristofer Wood
- From the Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (S.X.S., K.S., K.W., A.F.D., Q.L.)
| | - Andrew F Ducruet
- From the Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (S.X.S., K.S., K.W., A.F.D., Q.L.)
| | - Qiang Liu
- From the Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (S.X.S., K.S., K.W., A.F.D., Q.L.)
| |
Collapse
|
10
|
Wang Y, Zhang JH, Sheng J, Shao A. Immunoreactive Cells After Cerebral Ischemia. Front Immunol 2019; 10:2781. [PMID: 31849964 PMCID: PMC6902047 DOI: 10.3389/fimmu.2019.02781] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
The immune system is rapidly activated after ischemic stroke. As immune cells migrate and infiltrate across the blood-brain barrier into the ischemic region, a cascade of cellular and molecular biological reactions occur, involving migrated immune cells, resident glial cells, and the vascular endothelium. These events regulate infarction evolution and thus influence the outcome of ischemic stroke. Most immune cells exert dual effects on cerebral ischemia, and some crucial cells may become central targets in ischemic stroke treatment and rehabilitation.
Collapse
Affiliation(s)
- Yijie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Wnt-3a alleviates neuroinflammation after ischemic stroke by modulating the responses of microglia/macrophages and astrocytes. Int Immunopharmacol 2019; 75:105760. [DOI: 10.1016/j.intimp.2019.105760] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
|
12
|
Stratification and prediction of remission in first-episode psychosis patients: the OPTiMiSE cohort study. Transl Psychiatry 2019; 9:20. [PMID: 30655509 PMCID: PMC6336802 DOI: 10.1038/s41398-018-0366-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 12/10/2018] [Indexed: 11/09/2022] Open
Abstract
Early response to first-line antipsychotic treatments is strongly associated with positive long-term symptomatic and functional outcome in psychosis. Unfortunately, attempts to identify reliable predictors of treatment response in first-episode psychosis (FEP) patients have not yet been successful. One reason for this could be that FEP patients are highly heterogeneous in terms of symptom expression and underlying disease biological mechanisms, thereby impeding the identification of one-size-fits-all predictors of treatment response. We have used a clustering approach to stratify 325 FEP patients into four clinical subtypes, termed C1A, C1B, C2A and C2B, based on their symptoms assessed using the Positive and Negative Syndrome Scale (PANSS) scale. Compared to C1B, C2A and C2B patients, those from the C1A subtype exhibited the most severe symptoms and were the most at risk of being non-remitters when treated with the second-generation antipsychotic drug amisulpride. Before treatment, C1A patients exhibited higher serum levels of several pro-inflammatory cytokines and inflammation-associated biomarkers therefore validating our stratification approach on external biological measures. Most importantly, in the C1A subtype, but not others, lower serum levels of interleukin (IL)-15, higher serum levels of C-X-C motif chemokine 12 (CXCL12), previous exposure to cytomegalovirus (CMV), use of recreational drugs and being younger were all associated with higher odds of being non-remitters 4 weeks after treatment. The predictive value of this model was good (mean area under the curve (AUC) = 0.73 ± 0.10), and its specificity and sensitivity were 45 ± 0.09% and 83 ± 0.03%, respectively. Further validation and replication of these results in clinical trials would pave the way for the development of a blood-based assisted clinical decision support system in psychosis.
Collapse
|
13
|
Hersh J, Yang SH. Glia-immune interactions post-ischemic stroke and potential therapies. Exp Biol Med (Maywood) 2018; 243:1302-1312. [PMID: 30537868 DOI: 10.1177/1535370218818172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
IMPACT STATEMENT This article reviews glial cell interactions with the immune system post-ischemic stroke. Research has shown that glial cells in the brain play a role in altering phenotypes of other glial cells and have downstream immune cell targets ultimately regulating a neuroinflammatory response. These interactions may play a deleterious as well as beneficial role in stroke recovery. Furthermore, they may provide a novel way to approach potential therapies, since current stroke drug therapy is limited to only one Food and Drug Administration-approved drug complicated by a narrow therapeutic window. Until this point, most research has emphasized neuroimmune interactions, but little focus has been on bidirectional communication of glial-immune interactions in the ischemic brain. By expanding our understanding of these interactions through a compilation of glial cell effects, we may be able to pinpoint major modulating factors in brain homeostasis to maintain or discover ways to suppress irreversible ischemic damage and improve brain repair.
Collapse
Affiliation(s)
- Jessica Hersh
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shao-Hua Yang
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
14
|
NK cells in cerebral ischemia. Biomed Pharmacother 2018; 109:547-554. [PMID: 30399590 DOI: 10.1016/j.biopha.2018.10.103] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/18/2018] [Accepted: 10/20/2018] [Indexed: 01/03/2023] Open
Abstract
As a vital cell type in immune system and infiltrating cells in ischemic brain, NK cells can bridge the crosstalk between immune system and nervous system in stroke setting. The mechanism of action of NK cells is complicated, involving direct and indirect actions. NK cells are closely associated with poststroke inflammation, immunodepression and infections. The excessive inflammatory response in ischemic brain is one of the important causes for aggravating cerebral ischemic injury. Besides the inflammation induced by ischemia itself, thrombolytic drug tissue plasminogen activator (tPA) administration could also induce deteriorative inflammation, which is unfavorable for stroke control and recovery. Regulating NK cells may has the potential to modulate the immune response, limiting the development of ischemic damage and getting better outcome. In addition, post-stroke immunosuppression may lead to infections which contribute to higher severity and mortality of ischemic stroke (IS). Targeting NK cells may help to find novel pathways for IS therapy, which can both ameliorate the infarction itself, but also reduce the infectious complications. NK cells may also link IS and related diseases, suggesting NK cells can be used as a diagnostic or prognostic biomarker for IS prevention and treatment.
Collapse
|
15
|
Lee GA, Lin TN, Chen CY, Mau SY, Huang WZ, Kao YC, Ma RY, Liao NS. Interleukin 15 blockade protects the brain from cerebral ischemia-reperfusion injury. Brain Behav Immun 2018; 73:562-570. [PMID: 29959050 DOI: 10.1016/j.bbi.2018.06.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/02/2018] [Accepted: 06/22/2018] [Indexed: 01/08/2023] Open
Abstract
Acute ischemic stroke is followed by a complex interplay between the brain and the immune system in which ischemia-reperfusion leads to a detrimental inflammatory response that causes brain injury. In the brain, IL-15 is expressed by astrocytes, neurons and microglia. Previous study showed that ischemia-reperfusion induces expression of IL-15 by astrocytes. Transgenic over-expression of IL-15 in astrocytes aggravates ischemia-reperfusion brain damage by increasing the levels and promoting the effector functions of CD8+ T and NK cells. Treatment of neonatal rats with IL-15 neutralizing antibody before hypoxia-ischemia induction reduces the infarct volume. However, as stroke-induced inflammatory responses differ between neonate and adult brain, the effects of IL-15 blockade on the injury and immune response arising from stroke in adult animals has remained unclear. In this study, we examined the effect of post-ischemia/reperfusion IL-15 blockade on the pathophysiology of cerebral ischemia-reperfusion in adult mice. Using a cerebral ischemia-reperfusion model, we compared infarct size and the infiltrating immune cells in the brain of wild type (WT) mice and Il15-/- mice lacking NK and memory CD8+ T cells. We also evaluated the effects of IL-15 neutralizing antibody treatment on brain infarct volume, motor function, and the status of brain-infiltrating immune cells in WT mice. Il15-/- mice show a smaller infarct volume and lower numbers of activated brain-infiltrating NK, CD8+ T, and CD4+ T cells compared to WT mice after cerebral ischemia-reperfusion. Post-ischemia/reperfusion IL-15 blockade reduces infarct size and improves motor and locomotor activity. Furthermore, IL-15 blockade reduces the effector function of NK, CD8+ T, and CD4+ T cells in the ischemia-reperfusion brain of WT mice. Ablation of IL-15 responses after cerebral ischemia-reperfusion ameliorates brain injury in adult mice. Therefore, targeting IL-15 is a potential effective therapy for ischemic stroke.
Collapse
Affiliation(s)
- Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Cheng-Yu Chen
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shin-Yi Mau
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wan-Zhen Huang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yu-Chieh Kao
- Translational Imaging Research Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ruo-Yu Ma
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Nan-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
16
|
Wang N, Feng Z, Zhao W, Zhang Z, Zhang L. Ultrashortwave radiation promotes the recovery of spinal cord injury by inhibiting inflammation via suppression of the MK2/TNF‑α pathway. Int J Mol Med 2018; 42:1909-1916. [PMID: 30066830 PMCID: PMC6108855 DOI: 10.3892/ijmm.2018.3786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 07/12/2018] [Indexed: 01/10/2023] Open
Abstract
Mitogen-activated protein kinase-activated protein kinase 2 (MK2) and its mediated inflammation are involved in various diseases, including spinal cord injury (SCI). Ultrashortwave (USW) radiation has previously been reported to exert a protective effect on SCI. In the present study, through a series of reverse transcription-quantitative polymerase chain reaction (RT-qPCR), western blot and immunofluorescence assay, it was found that MK2 and tumor necrosis factor (TNF)-α/interleukin (IL)-1β were elevated in patients with SCI and in H2O2-treated C8-D1A cells. Through gene level and protein level detection by using of RT-qPCR, western blot, immunofluorescence assay and terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling assay, it was demonstrated that USW radiation inhibited the expression of MK2/TNF-α/IL-1β and suppressed the apoptosis of H2O2-treated C8-D1A cells. Furthermore, it was confirmed that the overexpression of MK2 reversed the protective effect of USW on C8-D1A cells, which indicated that USW achieved its function via regulation of the MK2/TNF-α/IL-1β pathway. Finally, using a constructed in vivo model and a series of RT-qPCR, western blot and IHC detection, it was confirmed that USW suppressed the expression of MK2 to promote functional recovery following SCI. The findings of the present study may provide a novel target and improve on the current understanding of how USW functions in the treatment of SCI.
Collapse
Affiliation(s)
- Nan Wang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Zhiping Feng
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Wei Zhao
- The 4th Department of Orthopedic Surgery, Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning 110024, P.R. China
| | - Zhiqiang Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Lixin Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| |
Collapse
|
17
|
Li Z, Han J, Ren H, Ma CG, Shi FD, Liu Q, Li M. Astrocytic Interleukin-15 Reduces Pathology of Neuromyelitis Optica in Mice. Front Immunol 2018; 9:523. [PMID: 29616032 PMCID: PMC5867910 DOI: 10.3389/fimmu.2018.00523] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/28/2018] [Indexed: 12/05/2022] Open
Abstract
Astrocyte loss induced by neuromyelitis optica (NMO)-IgG and complement-dependent cytotoxicity (CDC) is the hallmark of NMO pathology. The survival of astrocytes is thought to reflect astrocyte exposure to environmental factors in the CNS and the response of astrocytes to these factors. However, still unclear are how astrocytes respond to NMO-IgG and CDC, and what CNS environmental factors may impact the survival of astrocytes. In a murine model of NMO induced by intracerebral injection of NMO-IgG and human complement, we found dramatic upregulation of IL-15 in astrocytes. To study the role of astrocytic IL-15 in NMO, we generated a transgenic mouse line with targeted expression of IL-15 in astrocytes (IL-15tg), in which the expression of IL-15 is controlled by a glial fibrillary acidic protein promoter. We showed that astrocyte-targeted expression of IL-15 attenuates astrocyte injury and the loss of aquaporin-4 in the brain. Reduced blood–brain barrier leakage and immune cell infiltration are also found in the lesion of IL-15tg mice subjected to NMO induction. IL-15tg astrocytes are less susceptible to NMO-IgG-mediated CDC than their wild-type counterparts. The enhanced resistance of IL-15tg astrocytes to cytotoxicity and cell death involves NF-κB signaling pathway. Our findings suggest that IL-15 reduces astrocyte loss and NMO pathology.
Collapse
Affiliation(s)
- Zhiguo Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinrui Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Cun-Gen Ma
- Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Dreyer-Andersen N, Almeida AS, Jensen P, Kamand M, Okarmus J, Rosenberg T, Friis SD, Martínez Serrano A, Blaabjerg M, Kristensen BW, Skrydstrup T, Gramsbergen JB, Vieira HLA, Meyer M. Intermittent, low dose carbon monoxide exposure enhances survival and dopaminergic differentiation of human neural stem cells. PLoS One 2018; 13:e0191207. [PMID: 29338033 PMCID: PMC5770048 DOI: 10.1371/journal.pone.0191207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/30/2017] [Indexed: 12/18/2022] Open
Abstract
Exploratory studies using human fetal tissue have suggested that intrastriatal transplantation of dopaminergic neurons may become a future treatment for patients with Parkinson's disease. However, the use of human fetal tissue is compromised by ethical, regulatory and practical concerns. Human stem cells constitute an alternative source of cells for transplantation in Parkinson's disease, but efficient protocols for controlled dopaminergic differentiation need to be developed. Short-term, low-level carbon monoxide (CO) exposure has been shown to affect signaling in several tissues, resulting in both protection and generation of reactive oxygen species. The present study investigated the effect of CO produced by a novel CO-releasing molecule on dopaminergic differentiation of human neural stem cells. Short-term exposure to 25 ppm CO at days 0 and 4 significantly increased the relative content of β-tubulin III-immunoreactive immature neurons and tyrosine hydroxylase expressing catecholaminergic neurons, as assessed 6 days after differentiation. Also the number of microtubule associated protein 2-positive mature neurons had increased significantly. Moreover, the content of apoptotic cells (Caspase3) was reduced, whereas the expression of a cell proliferation marker (Ki67) was left unchanged. Increased expression of hypoxia inducible factor-1α and production of reactive oxygen species (ROS) in cultures exposed to CO may suggest a mechanism involving mitochondrial alterations and generation of ROS. In conclusion, the present procedure using controlled, short-term CO exposure allows efficient dopaminergic differentiation of human neural stem cells at low cost and may as such be useful for derivation of cells for experimental studies and future development of donor cells for transplantation in Parkinson's disease.
Collapse
Affiliation(s)
- Nanna Dreyer-Andersen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ana Sofia Almeida
- Instituto de Biologia Experimental e Tecnológica (IBET), Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica (ITQB), Oeiras, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Pia Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morad Kamand
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tine Rosenberg
- Department of Pathology, Odense University Hospital, Denmark & Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Stig Düring Friis
- Center for Insoluble Protein Structures (inSPIN), Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Alberto Martínez Serrano
- Department of Molecular Biology and Center of Molecular Biology Severo Ochoa, University Autonoma Madrid-C.S.I.C Campus Cantoblanco, Madrid, Spain
| | - Morten Blaabjerg
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Denmark & Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Troels Skrydstrup
- Center for Insoluble Protein Structures (inSPIN), Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Jan Bert Gramsbergen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Helena L. A. Vieira
- Instituto de Biologia Experimental e Tecnológica (IBET), Oeiras, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
19
|
Gao L, Dai C, Feng Z, Zhang L, Zhang Z. MiR-137 inhibited inflammatory response and apoptosis after spinal cord injury via targeting of MK2. J Cell Biochem 2017; 119:3280-3292. [PMID: 29125882 DOI: 10.1002/jcb.26489] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/09/2017] [Indexed: 12/29/2022]
Abstract
Spinal cord injuries are common and troublesome disorder, which is mediated by various signal pathways and mechanisms. MK2 is also involved in numerous inflammatory diseases including spinal cord injury. The role of microRNA-137 (miR-137) and its detailed working mechanism in spinal cord injuries remain unclear. In the present study, we found that an elevated MK2 but a decreased miR-137 was expressed in serum specimens of patients with spinal cord injury and in hydrogen peroxide-treated C8-D1A and C8-B4 cells. Meanwhile, we suggested that upregulation of miR-137 could inhibit the expression of TNF-α and IL-6, two markers of inflammatory response after SCI, and apoptosis in hydrogen peroxide-treated C8-D1A and C8-B4 cells. Furthermore, we verified that MK2 was a direct target of miR-137 thorough a constructed luciferase assay. Even further, we elucidated that miR-137 could suppress the inflammatory response and apoptosis via negative regulation of MK2. Finally, through an animal model trial performed using mice, we demonstrated the protective effect of how miR-137 works on inflammatory response and apoptosis after spinal cord injury. Considering all the forementioned, our findings revealed that miR-137 inhibited inflammatory response and apoptosis after spinal cord injury via the targeting of MK2. The outcomes of the present study might indicate a new target in molecular treatment of SCI.
Collapse
Affiliation(s)
- Lin Gao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Chenfei Dai
- Department of Orthopedics, The Eleventh People's Hospital of Shenyang, Shenyang, P.R. China
| | - Zhiping Feng
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Lixin Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Zhiqiang Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
20
|
White EJ, Trigatti BL, Igdoura SA. Suppression of NK and CD8+ T cells reduces astrogliosis but accelerates cerebellar dysfunction and shortens life span in a mouse model of Sandhoff disease. J Neuroimmunol 2017; 306:55-67. [DOI: 10.1016/j.jneuroim.2017.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/15/2017] [Accepted: 03/06/2017] [Indexed: 01/09/2023]
|
21
|
Chronic ethanol intake induces partial microglial activation that is not reversed by long-term ethanol withdrawal in the rat hippocampal formation. Neurotoxicology 2017; 60:107-115. [PMID: 28408342 DOI: 10.1016/j.neuro.2017.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/12/2022]
Abstract
Neuroinflammation has been implicated in the pathogenesis of several disorders. Activation of microglia leads to the release of pro-inflammatory mediators and microglial-mediated neuroinflammation has been proposed as one of the alcohol-induced neuropathological mechanisms. The present study aimed to examine the effect of chronic ethanol exposure and long-term withdrawal on microglial activation and neuroinflammation in the hippocampal formation. Male rats were submitted to 6 months of ethanol treatment followed by a 2-month withdrawal period. Stereological methods were applied to estimate the total number of microglia and activated microglia detected by CD11b immunohistochemistry in the hippocampal formation. The expression levels of the pro-inflammatory cytokines TNF-α, COX-2 and IL-15 were measured by qRT-PCR. Alcohol consumption was associated with an increase in the total number of activated microglia but morphological assessment indicated that microglia did not exhibit a full activation phenotype. These data were supported by functional evidence since chronic alcohol consumption produced no changes in the expression of TNF-α or COX-2. The levels of IL-15 a cytokine whose expression is increased upon activation of both astrocytes and microglia, was induced by chronic alcohol treatment. Importantly, the partial activation of microglia induced by ethanol was not reversed by long-term withdrawal. This study suggests that chronic alcohol exposure induces a microglial phenotype consistent with partial activation without significant increase in classical cytokine markers of neuroinflammation in the hippocampal formation. Furthermore, long-term cessation of alcohol intake is not sufficient to alter the microglial partial activation phenotype induced by ethanol.
Collapse
|
22
|
|
23
|
Pardo CA, Farmer CA, Thurm A, Shebl FM, Ilieva J, Kalra S, Swedo S. Serum and cerebrospinal fluid immune mediators in children with autistic disorder: a longitudinal study. Mol Autism 2017; 8:1. [PMID: 28070266 PMCID: PMC5217649 DOI: 10.1186/s13229-016-0115-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The causes of autism likely involve genetic and environmental factors that influence neurobiological changes and the neurological and behavioral features of the disorder. Immune factors and inflammation are hypothesized pathogenic influences, but have not been examined longitudinally. METHODS In a cohort of 104 participants with autism, we performed an assessment of immune mediators such as cytokines, chemokines, or growth factors in serum and cerebrospinal fluid (n = 67) to determine potential influences of such mediators in autism. RESULTS As compared with 54 typically developing controls, we found no evidence of differences in the blood profile of immune mediators supportive of active systemic inflammation mechanisms in participants with autism. Some modulators of immune function (e.g., EGF and soluble CD40 ligand) were increased in the autism group; however, no evidence of group differences in traditional markers of active inflammation (e.g., IL-6, TNFα, IL-1β) were observed in the serum. Further, within-subject stability (measured by estimated intraclass correlations) of most analytes was low, indicating that a single measurement is not a reliable prospective indicator of concentration for most analytes. Additionally, in participants with autism, there was little correspondence between the blood and CSF profiles of cytokines, chemokines, and growth factors, suggesting that peripheral markers may not optimally reflect the immune status of the central nervous system. Although the relatively high fraction of intrathecal production of selected chemokines involved in monocyte/microglia function may suggest a possible relationship with the homeostatic role of microglia, control data are needed for further interpretation of its relevance in autism. CONCLUSIONS These longitudinal observations fail to provide support for the hypothesized role of disturbances in the expression of circulating cytokines and chemokines as an indicator of systemic inflammation in autism. ClinicalTrials.gov, NCT00298246.
Collapse
Affiliation(s)
- Carlos A Pardo
- Johns Hopkins University School of Medicine, 627 Pathology Bld., 6000 North Wolfe Street, Baltimore, MD 21287 USA
| | - Cristan A Farmer
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| | - Audrey Thurm
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| | - Fatma M Shebl
- Yale School of Public Health, Yale University, New Haven, CT USA
| | - Jorjetta Ilieva
- Johns Hopkins University School of Medicine, 627 Pathology Bld., 6000 North Wolfe Street, Baltimore, MD 21287 USA
| | - Simran Kalra
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| | - Susan Swedo
- Pediatrics and Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD USA
| |
Collapse
|
24
|
Astrocyte-derived interleukin-15 exacerbates ischemic brain injury via propagation of cellular immunity. Proc Natl Acad Sci U S A 2016; 114:E396-E405. [PMID: 27994144 DOI: 10.1073/pnas.1612930114] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Astrocytes are believed to bridge interactions between infiltrating lymphocytes and neurons during brain ischemia, but the mechanisms for this action are poorly understood. Here we found that interleukin-15 (IL-15) is dramatically up-regulated in astrocytes of postmortem brain tissues from patients with ischemic stroke and in a mouse model of transient focal brain ischemia. We generated a glial fibrillary acidic protein (GFAP) promoter-controlled IL-15-expressing transgenic mouse (GFAP-IL-15tg) line and found enlarged brain infarcts, exacerbated neurodeficits after the induction of brain ischemia. In addition, knockdown of IL-15 in astrocytes attenuated ischemic brain injury. Interestingly, the accumulation of CD8+ T and natural killer (NK) cells was augmented in these GFAP-IL-15tg mice after brain ischemia. Of note, depletion of CD8+ T or NK cells attenuated ischemic brain injury in GFAP-IL-15tg mice. Furthermore, knockdown of the IL-15 receptor α or blockade of cell-to-cell contact diminished the activation and effector function of CD8+ T and NK cells in GFAP-IL-15tg mice, suggesting that astrocytic IL-15 is delivered in trans to target cells. Collectively, these findings indicate that astrocytic IL-15 could aggravate postischemic brain damage via propagation of CD8+ T and NK cell-mediated immunity.
Collapse
|
25
|
Patidar M, Yadav N, Dalai SK. Interleukin 15: A key cytokine for immunotherapy. Cytokine Growth Factor Rev 2016; 31:49-59. [PMID: 27325459 DOI: 10.1016/j.cytogfr.2016.06.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/20/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-15, a member of the immunoregulatory cytokines family, is a pluripotent molecule with therapeutic potential. It is predominantly expressed by the myeloid cells, as well as other cell types. IL-15 serves multiple functions including dictating T cell response, regulating tissue repair and B cell homing, modulating inflammation, and activating NK cells. Among cytokines, IL-15 is unique because of its wide expression, tightly regulated secretion, trans-presentation, and therapeutic potential. IL-15 has been investigated for its therapeutic potential for the induction and maintenance of T cell responses. In addition, IL-15 can be targeted by antibody- or mutant IL-15 therapy to reduce inflammation. Its multifaceted biological applications are crucial in immunotherapy. In this article, we review the functions, expression, and regulation of IL-15 for designing an improved IL-15-based therapy targeting the IL-15 signaling pathway.
Collapse
Affiliation(s)
- Manoj Patidar
- Institute of Science, Nirma University, Ahmedabad 382481, India.
| | - Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad 382481, India.
| | - Sarat K Dalai
- Institute of Science, Nirma University, Ahmedabad 382481, India.
| |
Collapse
|
26
|
Olmos-Alonso A, Schetters STT, Sri S, Askew K, Mancuso R, Vargas-Caballero M, Holscher C, Perry VH, Gomez-Nicola D. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer's-like pathology. Brain 2016; 139:891-907. [PMID: 26747862 PMCID: PMC4766375 DOI: 10.1093/brain/awv379] [Citation(s) in RCA: 349] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/29/2015] [Indexed: 01/24/2023] Open
Abstract
The proliferation and activation of microglial cells is a hallmark of several neurodegenerative conditions. This mechanism is regulated by the activation of the colony-stimulating factor 1 receptor (CSF1R), thus providing a target that may prevent the progression of conditions such as Alzheimer’s disease. However, the study of microglial proliferation in Alzheimer’s disease and validation of the efficacy of CSF1R-inhibiting strategies have not yet been reported. In this study we found increased proliferation of microglial cells in human Alzheimer’s disease, in line with an increased upregulation of the CSF1R-dependent pro-mitogenic cascade, correlating with disease severity. Using a transgenic model of Alzheimer’s-like pathology (APPswe, PSEN1dE9; APP/PS1 mice) we define a CSF1R-dependent progressive increase in microglial proliferation, in the proximity of amyloid-β plaques. Prolonged inhibition of CSF1R in APP/PS1 mice by an orally available tyrosine kinase inhibitor (GW2580) resulted in the blockade of microglial proliferation and the shifting of the microglial inflammatory profile to an anti-inflammatory phenotype. Pharmacological targeting of CSF1R in APP/PS1 mice resulted in an improved performance in memory and behavioural tasks and a prevention of synaptic degeneration, although these changes were not correlated with a change in the number of amyloid-β plaques. Our results provide the first proof of the efficacy of CSF1R inhibition in models of Alzheimer’s disease, and validate the application of a therapeutic strategy aimed at modifying CSF1R activation as a promising approach to tackle microglial activation and the progression of Alzheimer’s disease.
Collapse
Affiliation(s)
- Adrian Olmos-Alonso
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | | | - Sarmi Sri
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Katharine Askew
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Renzo Mancuso
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Mariana Vargas-Caballero
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK 2 Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christian Holscher
- 3 Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YQ, UK
| | - V Hugh Perry
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Diego Gomez-Nicola
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
27
|
Bortell N, Morsey B, Basova L, Fox HS, Marcondes MCG. Phenotypic changes in the brain of SIV-infected macaques exposed to methamphetamine parallel macrophage activation patterns induced by the common gamma-chain cytokine system. Front Microbiol 2015; 6:900. [PMID: 26441851 PMCID: PMC4568411 DOI: 10.3389/fmicb.2015.00900] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/17/2015] [Indexed: 12/12/2022] Open
Abstract
One factor in the development of neuroAIDS is the increase in the migration of pro-inflammatory CD8 T cells across the blood–brain barrier. Typically these cells are involved with keeping the viral load down. However, the persistence of above average numbers of CD8 T cells in the brain, not necessarily specific to viral peptides, is facilitated by the upregulation of IL15 from astrocytes, in the absence of IL2, in the brain environment. Both IL15 and IL2 are common gamma chain (γc) cytokines. Here, using the non-human primate model of neuroAIDS, we have demonstrated that exposure to methamphetamine, a powerful illicit drug that has been associated with HIV exposure and neuroAIDS severity, can cause an increase in molecules of the γc system. Among these molecules, IL15, which is upregulated in astrocytes by methamphetamine, and that induces the proliferation of T cells, may also be involved in driving an inflammatory phenotype in innate immune cells of the brain. Therefore, methamphetamine and IL15 may be critical in the development and aggravation of central nervous system immune-mediated inflammatory pathology in HIV-infected drug abusers.
Collapse
Affiliation(s)
- Nikki Bortell
- Department of Molecular and Cellular Neurosciences, The Scripps Research Institute La Jolla, CA, USA
| | - Brenda Morsey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center Omaha, NE, USA
| | - Liana Basova
- Department of Molecular and Cellular Neurosciences, The Scripps Research Institute La Jolla, CA, USA
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center Omaha, NE, USA
| | | |
Collapse
|
28
|
Cardoso FL, Herz J, Fernandes A, Rocha J, Sepodes B, Brito MA, McGavern DB, Brites D. Systemic inflammation in early neonatal mice induces transient and lasting neurodegenerative effects. J Neuroinflammation 2015; 12:82. [PMID: 25924675 PMCID: PMC4440597 DOI: 10.1186/s12974-015-0299-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/10/2015] [Indexed: 12/11/2022] Open
Abstract
Background The inflammatory mediator lipopolysaccharide (LPS) has been shown to induce acute gliosis in neonatal mice. However, the progressive effects on the murine neurodevelopmental program over the week that follows systemic inflammation are not known. Thus, we investigated the effects of repeated LPS administration in the first postnatal week in mice, a condition mimicking sepsis in late preterm infants, on the developing central nervous system (CNS). Methods Systemic inflammation was induced by daily intraperitoneal administration (i.p.) of LPS (6 mg/kg) in newborn mice from postnatal day (PND) 4 to PND6. The effects on neurodevelopment were examined by staining the white matter and neurons with Luxol Fast Blue and Cresyl Violet, respectively. The inflammatory response was assessed by quantifying the expression/activity of matrix metalloproteinases (MMP), toll-like receptor (TLR)-4, high mobility group box (HMGB)-1, and autotaxin (ATX). In addition, B6 CX3CR1gfp/+ mice combined with cryo-immunofluorescence were used to determine the acute, delayed, and lasting effects on myelination, microglia, and astrocytes. Results LPS administration led to acute body and brain weight loss as well as overt structural changes in the brain such as cerebellar hypoplasia, neuronal loss/shrinkage, and delayed myelination. The impaired myelination was associated with alterations in the proliferation and differentiation of NG2 progenitor cells early after LPS administration, rather than with excessive phagocytosis by CNS myeloid cells. In addition to disruptions in brain architecture, a robust inflammatory response to LPS was observed. Quantification of inflammatory biomarkers revealed decreased expression of ATX with concurrent increases in HMGB1, TLR-4, and MMP-9 expression levels. Acute astrogliosis (GFAP+ cells) in the brain parenchyma and at the microvasculature interface together with parenchymal microgliosis (CX3CR1+ cells) were also observed. These changes preceded the migration/proliferation of CX3CR1+ cells around the vessels at later time points and the subsequent loss of GFAP+ astrocytes. Conclusion Collectively, our study has uncovered a complex innate inflammatory reaction and associated structural changes in the brains of neonatal mice challenged peripherally with LPS. These findings may explain some of the neurobehavioral abnormalities that develop following neonatal sepsis.
Collapse
Affiliation(s)
- Filipa L Cardoso
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Jasmin Herz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892-1430, USA.
| | - Adelaide Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Bruno Sepodes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Maria A Brito
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Dorian B McGavern
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892-1430, USA.
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
29
|
Bishnoi RJ, Palmer RF, Royall DR. Serum interleukin (IL)-15 as a biomarker of Alzheimer's disease. PLoS One 2015; 10:e0117282. [PMID: 25710473 PMCID: PMC4339977 DOI: 10.1371/journal.pone.0117282] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 12/19/2014] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL-15), a pro-inflammatory cytokine has been studied as a possible marker of Alzheimer’s disease (AD); however its exact role in neuro-inflammation or the pathogenesis AD is not well understood yet. A Multiple Indicators Multiple Causes (MIMIC) approach was used to examine the relationship between serum IL-15 levels and AD in a well characterized AD cohort, the Texas Alzheimer's Research and Care Consortium (TARCC). Instead of categorical diagnoses, we used two latent construct d (for dementia) and g’ (for cognitive impairments not contributing to functional impairments) in our analysis. The results showed that the serum IL-15 level has significant effects on cognition, exclusively mediated by latent construct d and g’. Contrasting directions of association lead us to speculate that IL-15’s effects in AD are mediated through functional networks as d scores have been previously found to be specifically related to default mode network (DMN). Our finding warrants the need for further research to determine the changes in structural and functional networks corresponding to serum based biomarkers levels.
Collapse
Affiliation(s)
- Ram J. Bishnoi
- Department of Psychiatry, University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail:
| | - Raymond F. Palmer
- Department of Family and Community Medicine, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Donald R. Royall
- Department of Psychiatry, Family and Community Medicine, and Medicine, University of Texas Health Science Center, South Texas Veterans’ Health System Audie L. Murphy Division, Geriatric Research Education and Clinical Centers, San Antonio, Texas, United States of America
| |
Collapse
|
30
|
Fathali N, Ostrowski RP, Hasegawa Y, Lekic T, Tang J, Zhang JH. Splenic immune cells in experimental neonatal hypoxia-ischemia. Transl Stroke Res 2014; 4:208-19. [PMID: 23626659 DOI: 10.1007/s12975-012-0239-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuroimmune processes contribute to hypoxic-ischemic damage in the immature brain and may play a role in the progression of particular variants of neonatal encephalopathy. The present study was designed to elucidate molecular mediators of interactions between astrocytes, neurons and infiltrating peripheral immune cells after experimental neonatal hypoxia-ischemia (HI). Splenectomy was performed on postnatal day-7 Sprague-Dawley rats 3 days prior to HI surgery; in which the right common carotid artery was permanently ligated followed by 2 hours of hypoxia (8% O2). Quantitative analysis showed that natural killer (NK) and T cell expression was reduced in spleen but increased in the brain following HI. Elevations in cyclooxygenase-2 (COX-2) expression after HI by immune cells promoted interleukin-15 expression in astrocytes and infiltration of inflammatory cells to site of injury; additionally, down-regulated the pro-survival protein, phosphoinositide-3-kinase, resulting in caspase-3 mediated neuronal death. The removal of the largest pool of peripheral immune cells in the body by splenectomy, COX-2 inhibitors, as well as rendering NK cells inactive by CD161 knockdown, significantly ameliorated cerebral infarct volume at 72 hours, diminished body weight loss and brain and systemic organ atrophy, and reduced neurobehavioral deficits at 3 weeks. Herein we demonstrate with the use of surgical approach (splenectomy), with pharmacological loss-gain function approach using COX-2 inhibitors/agonists, as well as with NK cell-type specific siRNA that after neonatal HI, the infiltrating peripheral immune cells may modulate downstream targets of cell death and neuroinflammation by COX-2 regulated signals.
Collapse
Affiliation(s)
- Nancy Fathali
- Department of Human Anatomy and Pathology, Loma Linda University, Loma Linda, California, USA
| | | | | | | | | | | |
Collapse
|
31
|
Gomez-Nicola D, Suzzi S, Vargas-Caballero M, Fransen NL, Al-Malki H, Cebrian-Silla A, Garcia-Verdugo JM, Riecken K, Fehse B, Perry VH. Temporal dynamics of hippocampal neurogenesis in chronic neurodegeneration. ACTA ACUST UNITED AC 2014; 137:2312-28. [PMID: 24941947 PMCID: PMC4107745 DOI: 10.1093/brain/awu155] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Increased neurogenesis has been reported in neurodegenerative disease, but its significance is unclear. In a mouse model of prion disease, Gomez-Nicola et al. detect increased neurogenesis in the dentate gyrus that partially counteracts neuronal loss. Targeting neurogenesis may have therapeutic potential. The study of neurogenesis during chronic neurodegeneration is crucial in order to understand the intrinsic repair mechanisms of the brain, and key to designing therapeutic strategies. In this study, using an experimental model of progressive chronic neurodegeneration, murine prion disease, we define the temporal dynamics of the generation, maturation and integration of new neurons in the hippocampal dentate gyrus, using dual pulse-chase, multicolour γ-retroviral tracing, transmission electron microscopy and patch-clamp. We found increased neurogenesis during the progression of prion disease, which partially counteracts the effects of chronic neurodegeneration, as evidenced by blocking neurogenesis with cytosine arabinoside, and helps to preserve the hippocampal function. Evidence obtained from human post-mortem samples, of both variant Creutzfeldt-Jakob disease and Alzheimer’s disease patients, also suggests increased neurogenic activity. These results open a new avenue into the exploration of the effects and regulation of neurogenesis during chronic neurodegeneration, and offer a new model to reproduce the changes observed in human neurodegenerative diseases.
Collapse
Affiliation(s)
- Diego Gomez-Nicola
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Stefano Suzzi
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Mariana Vargas-Caballero
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Nina L Fransen
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Hussain Al-Malki
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | | | | | - Kristoffer Riecken
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Boris Fehse
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - V Hugh Perry
- 1 Centre for Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
32
|
Gómez-Nicola D, Schetters STT, Perry VH. Differential role of CCR2 in the dynamics of microglia and perivascular macrophages during prion disease. Glia 2014; 62:1041-52. [PMID: 24648328 PMCID: PMC4324129 DOI: 10.1002/glia.22660] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/28/2014] [Accepted: 03/04/2014] [Indexed: 01/27/2023]
Abstract
The expansion of the microglial population is one of the hallmarks of numerous brain disorders. The addition of circulating progenitors to the pool of brain macrophages can contribute to the progression of brain disease and needs to be precisely defined to better understand the evolution of the glial and inflammatory reactions in the brain. We have analyzed the degree of infiltration/recruitment of circulating monocytes to the microglial pool, in a prion disease model of chronic neurodegeneration. Our results indicate a minimal/absent level of CCR2-dependent recruitment of circulating monocytes, local proliferation of microglia is the main driving force maintaining the amplification of the population. A deficiency in CCR2, and thus the absence of recruitment of circulating monocytes, does not impact microglial dynamics, the inflammatory profile or the temporal behavioral course of prion disease. However, the lack of CCR2 has unexpected effects including the failure to recruit perivascular macrophages in diseased but not healthy CNS and a small reduction in microglia proliferation. These data define the composition of the CNS-resident macrophage populations in prion disease and will help to understand the dynamics of the CNS innate immune response during chronic neurodegeneration.
Collapse
Affiliation(s)
- Diego Gómez-Nicola
- Centre for Biological Sciences, University of Southampton, United Kingdom
| | | | | |
Collapse
|
33
|
Dooley D, Vidal P, Hendrix S. Immunopharmacological intervention for successful neural stem cell therapy: New perspectives in CNS neurogenesis and repair. Pharmacol Ther 2013; 141:21-31. [PMID: 23954656 DOI: 10.1016/j.pharmthera.2013.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/11/2022]
Abstract
The pharmacological support and stimulation of endogenous and transplanted neural stem cells (NSCs) is a major challenge in brain repair. Trauma to the central nervous system (CNS) results in a distinct inflammatory response caused by local and infiltrating immune cells. This makes NSC-supported regeneration difficult due to the presence of inhibitory immune factors which are upregulated around the lesion site. The continual and dual role of the neuroinflammatory response leaves it difficult to decipher upon a single modulatory strategy. Therefore, understanding the influence of cytokines upon regulation of NSC self-renewal, proliferation and differentiation is crucial when designing therapies for CNS repair. There is a plethora of partially conflicting data in vitro and in vivo on the role of cytokines in modulating the stem cell niche and the milieu around NSC transplants. This is mainly due to the pleiotropic role of many factors. In order for cell-based therapy to thrive, treatment must be phase-specific to the injury and also be personalized for each patient, i.e. taking age, sex, neuroimmune and endocrine status as well as other key parameters into consideration. In this review, we will summarize the most relevant information concerning interleukin (IL)-1, IL-4, IL-10, IL-15, IFN-γ, the neuropoietic cytokine family and TNF-α in order to extract promising therapeutic approaches for further research. We will focus on the consequences of neuroinflammation on endogenous brain stem cells and the transplantation environment, the effects of the above cytokines on NSCs, as well as immunopharmacological manipulation of the microenvironment for potential therapeutic use.
Collapse
Affiliation(s)
- Dearbhaile Dooley
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium
| | - Pia Vidal
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium
| | - Sven Hendrix
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium.
| |
Collapse
|
34
|
Abstract
An important component of chronic neurodegenerative diseases is the generation of an innate inflammatory response within the CNS. Microglial and astroglial cells play a key role in the development and maintenance of this inflammatory response, showing enhanced proliferation and activation. We studied the time course and regulation of microglial proliferation, using a mouse model of prion disease. Our results show that the proliferation of resident microglial cells accounts for the expansion of the population during the development of the disease. We identify the pathway regulated by the activation of CSF1R and the transcription factors PU.1 and C/EBPα as the molecular regulators of the proliferative response, correlating with the chronic human neurodegenerative conditions variant Creutzfeldt-Jakob disease and Alzheimer's disease. We show that targeting the activity of CSF1R inhibits microglial proliferation and slows neuronal damage and disease progression. Our results demonstrate that microglial proliferation is a major component in the evolution of chronic neurodegeneration, with direct implications for understanding the contribution of the CNS innate immune response to disease progression.
Collapse
|
35
|
Abstract
Interleukin (IL)-15 is a ubiquitously expressed cytokine existing in both intracellular and secretory forms. Here we review the expression, regulation, and functions of IL15 and its receptors in the brain. IL15 receptors show robust upregulation after neuroinflammation, suggesting a major role of IL15 signaling in cerebral function. Involvement of the IL15 system in neuropsychiatric behavior is reflected by the effects of IL15, IL15Rα, and IL2Rγ deletions on neurobehavior and neurotransmitters, the effects of IL15 treatment on neuronal activity, and the potential role of IL15 in neuroplasticity/neurogenesis. The results show that IL15 modulates GABA and serotonin transmission. This may underlie deficits in mood (depressive-like behavior and decreased normal anxiety) and memory, as well as activity level, sleep, and thermoregulation. Although IL15 has only a low level of permeation across the blood-brain barrier, peripheral IL15 is able to activate multiple signaling pathways in neurons widely distributed in CNS regions. The effects of IL15 in "preventing" neuropsychiatric symptoms in normal mice implicate a potential therapeutic role of this polypeptide cytokine.
Collapse
|
36
|
Krishnan G, Chatterjee N. Endocannabinoids alleviate proinflammatory conditions by modulating innate immune response in muller glia during inflammation. Glia 2012; 60:1629-45. [DOI: 10.1002/glia.22380] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/05/2012] [Indexed: 12/29/2022]
|
37
|
Cannabinoid agonist WIN 55,212-2 prevents the development of paclitaxel-induced peripheral neuropathy in rats. Possible involvement of spinal glial cells. Eur J Pharmacol 2012; 682:62-72. [PMID: 22374260 DOI: 10.1016/j.ejphar.2012.02.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 02/02/2012] [Accepted: 02/08/2012] [Indexed: 02/07/2023]
Abstract
Spinal glial activation contributes to the development and maintenance of chronic pain states, including neuropathic pain of diverse etiologies. Cannabinoid compounds have shown antinociceptive properties in a variety of neuropathic pain models and are emerging as a promising class of drugs to treat neuropathic pain. Thus, the effects of repeated treatment with WIN 55,212-2, a synthetic cannabinoid agonist, were examined throughout the development of paclitaxel-induced peripheral neuropathy. Painful neuropathy was induced in male Wistar rats by intraperitoneal (i.p.) administration of paclitaxel (1mg/kg) on four alternate days. Paclitaxel-treated animals received WIN 55,212-2 (1mg/kg, i.p.) or minocycline (15 mg/kg, i.p.), a microglial inhibitor, daily for 14 days, simultaneous with the antineoplastic. The development of hypersensitive behaviors was assessed on days 1, 7, 14, 21 and 28 following the initial administration of drugs. Both the activation of glial cells (microglia and astrocytes) at day 29 and the time course of proinflammatory cytokine release within the spinal cord were also determined. Similar to minocycline, repeated administration of WIN 55,212-2 prevented the development of thermal hyperalgesia and mechanical allodynia in paclitaxel-treated rats. WIN 55,212-2 treatment also prevented spinal microglial and astrocytic activation evoked by paclitaxel at day 29 and attenuated the early production of spinal proinflammatory cytokines (interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α). Our results confirm changes in the reactivity of glial cells during the development of peripheral neuropathy induced by paclitaxel and support a preventive effect of WIN 55,212-2, probably via glial cells reactivity inactivation, on the development of this neuropathy.
Collapse
|
38
|
Abstract
IL-15 is a proinflammatory cytokine. It is produced by activated blood monocytes, macrophages, dendritic cells, and activated glial cells. It promotes T-cell proliferation, induction of cytolytic effector cells including natural killer and cytotoxic cells and stimulates B-cell to proliferate and secrete immunoglobulins. Little information is available on the exact role of IL-15 in the neurological diseases. Microglial cells are the main regulators of both innate and adaptive immune responses in the central nervous system (CNS). IL-15 may be involved in the inflammatory reactions and microglial activation of some common CNS disorders such as multiple sclerosis, Alzheimer's and Parkinson's disease, but its exact role in their pathogenesis is not clear.
Collapse
Affiliation(s)
- M Rentzos
- Department of Neurology, Aeginition Hospital, Athens National University, School of Medicine, Greece.
| | | |
Collapse
|
39
|
David S, Zarruk JG, Ghasemlou N. Inflammatory pathways in spinal cord injury. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 106:127-52. [PMID: 23211462 DOI: 10.1016/b978-0-12-407178-0.00006-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Injury to the spinal cord results in direct damage to axons, neuronal cell bodies, and glia that cause functional loss below the site of injury. In addition, the injury also triggers an inflammatory response that contributes to secondary tissue damage that leads to further functional loss. Reducing inflammation after spinal cord injury (SCI) is therefore a worthy therapeutic goal. Inflammation in the injured spinal cord is a complex response that involves resident cells of the central nervous system as well as infiltrating immune cells, and is mediated by a variety of molecular pathways and signaling molecules. Here, we discuss approaches we have used to identify novel therapeutic targets to modulate the inflammatory response after SCI to reduce tissue damage and promote recovery. Effective treatments for SCI will likely require a combination of approaches to reduce inflammation and secondary damage with those that promote axon regeneration.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
40
|
Monnet‐Tschudi F, Defaux A, Braissant O, Cagnon L, Zurich M. Methods to Assess Neuroinflammation. ACTA ACUST UNITED AC 2011; Chapter 12:Unit12.19. [DOI: 10.1002/0471140856.tx1219s50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Florianne Monnet‐Tschudi
- Department of Physiology, University of Lausanne Lausanne Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT) Lausanne Switzerland
| | - Antoinette Defaux
- Department of Physiology, University of Lausanne Lausanne Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT) Lausanne Switzerland
| | - Olivier Braissant
- Clinical Chemistry Laboratory, Centre Hospitalier Universitaire Vaudois Lausanne Switzerland
| | - Laurène Cagnon
- Clinical Chemistry Laboratory, Centre Hospitalier Universitaire Vaudois Lausanne Switzerland
| | - Marie‐Gabrielle Zurich
- Department of Physiology, University of Lausanne Lausanne Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT) Lausanne Switzerland
| |
Collapse
|
41
|
Granado N, Lastres-Becker I, Ares-Santos S, Oliva I, Martin E, Cuadrado A, Moratalla R. Nrf2 deficiency potentiates methamphetamine-induced dopaminergic axonal damage and gliosis in the striatum. Glia 2011; 59:1850-63. [PMID: 21882243 DOI: 10.1002/glia.21229] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 07/20/2011] [Indexed: 12/22/2022]
Abstract
Oxidative stress that correlates with damage to nigrostriatal dopaminergic neurons and reactive gliosis in the basal ganglia is a hallmark of methamphetamine (METH) toxicity. In this study, we analyzed the protective role of the transcription factor Nrf2 (nuclear factor-erythroid 2-related factor 2), a master regulator of redox homeostasis, in METH-induced neurotoxicity. We found that Nrf2 deficiency exacerbated METH-induced damage to dopamine neurons, shown by an increase in loss of tyrosine hydroxylase (TH)- and dopamine transporter (DAT)-containing fibers in striatum. Consistent with these effects, Nrf2 deficiency potentiated glial activation, indicated by increased striatal expression of markers for microglia (Mac-1 and Iba-1) and astroglia (GFAP) one day after METH administration. At the same time, Nrf2 inactivation dramatically potentiated the increase in TNFα mRNA and IL-15 protein expression in GFAP+ cells in the striatum. In sharp contrast to the potentiation of striatal damage, Nrf2 deficiency did not affect METH-induced dopaminergic neuron death or expression of glial markers or proinflammatory molecules in the substantia nigra. This study uncovers a new role for Nrf2 in protection against METH-induced inflammatory and oxidative stress and striatal degeneration.
Collapse
|
42
|
Gómez-Nicola D, Valle-Argos B, Pallas-Bazarra N, Nieto-Sampedro M. Interleukin-15 regulates proliferation and self-renewal of adult neural stem cells. Mol Biol Cell 2011; 22:1960-70. [PMID: 21508317 PMCID: PMC3113763 DOI: 10.1091/mbc.e11-01-0053] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of IL-15 in the regulation of neural stem cell biology appears as a key mechanism in the control of adult neurogenesis, with direct implications for the development of pathologies with a neuroimmune component. The impact of inflammation is crucial for the regulation of the biology of neural stem cells (NSCs). Interleukin-15 (IL-15) appears as a likely candidate for regulating neurogenesis, based on its well-known mitogenic properties. We show here that NSCs of the subventricular zone (SVZ) express IL-15, which regulates NSC proliferation, as evidenced by the study of IL-15−/− mice and the effects of acute IL-15 administration, coupled to 5-bromo-2′-deoxyuridine/5-ethynyl-2′-deoxyuridine dual-pulse labeling. Moreover, IL-15 regulates NSC differentiation, its deficiency leading to an impaired generation of neuroblasts in the SVZ–rostral migratory stream axis, recoverable through the action of exogenous IL-15. IL-15 expressed in cultured NSCs is linked to self-renewal, proliferation, and differentiation. IL-15–/– NSCs presented deficient proliferation and self-renewal, as evidenced in proliferation and colony-forming assays and the analysis of cell cycle–regulatory proteins. Moreover, IL-15–deficient NSCs were more prone to differentiate than wild-type NSCs, not affecting the cell population balance. Lack of IL-15 led to a defective activation of the JAK/STAT and ERK pathways, key for the regulation of proliferation and differentiation of NSCs. The results show that IL-15 is a key regulator of neurogenesis in the adult and is essential to understanding diseases with an inflammatory component.
Collapse
Affiliation(s)
- Diego Gómez-Nicola
- Functional and Systems Neurobiology Department, Cajal Institute (CSIC), Madrid, Spain.
| | | | | | | |
Collapse
|
43
|
The evolving landscape of neuroinflammation after neonatal hypoxia-ischemia. ACTA NEUROCHIRURGICA. SUPPLEMENT 2011; 111:93-100. [PMID: 21725737 DOI: 10.1007/978-3-7091-0693-8_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxic-ischemic brain injury remains a leading cause of mortality and morbidity in neonates. The inflammatory response, which is characterized in part by activation of local immune cells, has been implicated as a core component for the progression of damage to the immature brain following hypoxia-ischemia (HI). However, mounting evidence implicates circulating immune cells recruited to the site of damage as orchestrators of neuron-glial interactions and perpetuators of secondary brain injury. This suggests that re-directing our attention from the local inflammatory response toward the molecular mediators believed to link brain-immune cell interactions may be a more effective approach to mitigating the inflammatory sequelae of perinatal HI. In this review, we focus our attention on cyclooxygenase-2, a mediator by which peripheral immune cells may modulate signaling pathways in the brain that lead to a worsened outcome. Additionally, we present an overview of emerging therapeutic modalities that target mechanisms of neuroinflammation in the hypoxic-ischemic neonate.
Collapse
|
44
|
Valle-Argos B, Gómez-Nicola D, Nieto-Sampedro M. Neurostatin blocks glioma cell cycle progression by inhibiting EGFR activation. Mol Cell Neurosci 2011; 46:89-100. [DOI: 10.1016/j.mcn.2010.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Accepted: 08/11/2010] [Indexed: 01/08/2023] Open
|
45
|
Saikali P, Antel JP, Pittet CL, Newcombe J, Arbour N. Contribution of astrocyte-derived IL-15 to CD8 T cell effector functions in multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2010; 185:5693-703. [PMID: 20926794 DOI: 10.4049/jimmunol.1002188] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The contribution of local factors to the activation of immune cells infiltrating the CNS of patients with multiple sclerosis (MS) remains to be defined. The cytokine IL-15 is pivotal in the maintenance and activation of CD8 T lymphocytes, a prominent lymphocyte population found in MS lesions. We investigated whether astrocytes are a functional source of IL-15 sufficient to enhance CD8 T lymphocyte responses and whether they provide IL-15 in the inflamed CNS of patients with MS. We observed that human astrocytes in primary cultures increased surface IL-15 levels upon activation with combinations of proinflammatory cytokines. Expanded human myelin autoreactive CD8 T lymphocytes cultured with such activated astrocytes displayed elevated lytic enzyme content, NKG2D expression, and Ag-specific cytotoxicity. These functional enhancements were abrogated by anti-IL-15-blocking Abs. Immunohistochemical analysis of brain tissue sections obtained from patients with MS demonstrated colocalization for IL-15 and the astrocyte marker glial fibrillary acidic protein within white matter lesions. The majority of astrocytes (80-90%) present in demyelinating MS lesions expressed IL-15, whereas few astrocytes in normal control brain sections had detectable IL-15. IL-15 could be detected in the majority of Iba-1-expressing microglia in the control sections, albeit in lower numbers when compared with microglia/macrophages in MS lesions. Furthermore, infiltrating CD8 T lymphocytes in MS lesions were in close proximity to IL-15-expressing cells. Astrocyte production of IL-15 resulting in the activation of CD8 T lymphocytes ascribes a role for these cells as contributors to the exacerbation of tissue damage during MS pathogenesis.
Collapse
Affiliation(s)
- Philippe Saikali
- Département de Médecine, Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal-Notre-Dame Hospital, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
46
|
CCR7 is expressed in astrocytes and upregulated after an inflammatory injury. J Neuroimmunol 2010; 227:87-92. [PMID: 20638137 DOI: 10.1016/j.jneuroim.2010.06.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 05/19/2010] [Accepted: 06/21/2010] [Indexed: 11/20/2022]
Abstract
Neurodegenerative or autoimmune diseases are frequently regulated by chemokines and their receptors, controlling both glial activation and immune cell infiltration. CCL19 and CCL21 have been described to mediate crucial functions during CNS pathological states, regulating both immune cell traffic to the CNS and communication between glia and neurons. Here, we describe the expression pattern and cellular sources of CCR7, receptor of CCL19 and CCL21, in the normal mouse brain. Moreover, we found that CCR7 is upregulated in reactive astrocytes upon intracerebral LPS, regulating early glial reactivity through its ligands CCL19 and CCL21. Our results indicate that CCR7 is playing an important role for the intercellular communication during the inflammatory activation in the CNS.
Collapse
|
47
|
Valle-Argos B, Gómez-Nicola D, Nieto-Sampedro M. Glioma growth inhibition by neurostatin and O-But GD1b. Neuro Oncol 2010; 12:1135-46. [PMID: 20615925 DOI: 10.1093/neuonc/noq073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In spite of their low incidence, central nervous system tumors have elevated morbidity and mortality, being responsible for 2.3% of total cancer deaths. The ganglioside O-acetylated GD1b (O-Ac GD1b; neurostatin), present in the mammalian brain, and the semi-synthetic O-butyrylated GD1b (O-But GD1b) are potent glioma proliferation inhibitors, appearing as possible candidates for the treatment of nervous system tumors. Tumoral cell division inhibitory activity in culture correlated with growth inhibition of glioma xenotransplants in Foxn1(nu) nude mice and intracranial glioma allotransplants. Both O-Ac GD1b and O-But GD1b inhibited in vivo cell proliferation, induced cell cycle arrest, and potentiated immune cell response to the tumor. Furthermore, the increased stability of the butyrylated compound (O-But GD1b) enhanced its activity with respect to the acetylated ganglioside (neurostatin). These results are the first report of the antitumoral activity of neurostatin and a neurostatin-like compound in vivo and indicate that semi-synthetic O-acetylated and O-butyrylated gangliosides are potent antitumoral compounds that should be considered in strategies for brain tumor treatment.
Collapse
Affiliation(s)
- Beatriz Valle-Argos
- Neural Plasticity Group, Functional and Systems Neurobiology Department, Instituto Cajal, Madrid, Spain
| | | | | |
Collapse
|
48
|
Butchi NB, Du M, Peterson KE. Interactions between TLR7 and TLR9 agonists and receptors regulate innate immune responses by astrocytes and microglia. Glia 2010; 58:650-64. [PMID: 19998480 DOI: 10.1002/glia.20952] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors 7 (TLR7) and 9 (TLR9) are important mediators of innate immune responses. Both receptors are located in endosomal compartments, recognize nucleic acids, and signal via Myeloid differentiation factor 88 (MyD88). In the current study, we analyzed TLR7 and TLR9 induced activation of astrocytes and microglia, two cell types that contribute to innate immune responses in the CNS. TLR7 and TLR9 agonists induced similar cytokine profiles within each cell type. However, there were notable differences in the cytokine profile between astrocytes and microglia, including the production of the anti-inflammatory cytokine IL-10 and antiapoptotic cytokines G-CSF and IL-9 by microglia but not astrocytes. Costimulation studies demonstrated that the TLR7 agonist, imiquimod, could inhibit TLR9 agonist-induced innate immune responses, in both cell types, in a concentration-dependent manner. Surprisingly, this inhibition was not mediated by TLR7, as deficiency in TLR7 did not alter suppression of the TLR9 agonist-induced responses. The suppression of innate immune responses was also not due to an inhibition of TLR9 agonist uptake. This suggested that imiquimod suppression may be a direct effect, possibly by blocking CpG-ODN binding and/or signaling with TLR9, thus limiting cell activation. An antagonistic relationship was also observed between the two receptors in microglia, with TLR7 deficiency resulting in enhanced cytokine responses to CpG-ODN stimulation. Thus, both TLR7 and its agonist can have inhibitory effects on TLR9-induced cytokine responses in glial cells.
Collapse
Affiliation(s)
- Niranjan B Butchi
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | | | | |
Collapse
|
49
|
Gomez-Nicola D, Valle-Argos B, Nieto-Sampedro M. Blockade of IL-15 activity inhibits microglial activation through the NFkappaB, p38, and ERK1/2 pathways, reducing cytokine and chemokine release. Glia 2010; 58:264-76. [PMID: 19610094 DOI: 10.1002/glia.20920] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Reactive glia formation is one of the hallmarks of damage to the CNS, but little information exists on the signals that direct its activation. Microglial cells are the main regulators of both innate and adaptative immune responses in the CNS. The proinflammatory cytokine IL-15 is involved in regulating the response of T and B cells, playing a key role in regulating nervous system inflammatory events. We have used a microglial culture model of inflammation induced by LPS and IFNgamma to evaluate the role of IL-15 in the proinflammatory response. Our results indicate that IL-15 is necessary for the reactive response, its deficiency (IL-15-/-) leading to the development of a defective proinflammatory response. Blockade of IL-15, both with blocking antibodies or with the ganglioside Neurostatin, inhibited the activation of the NFkappaB pathway, decreasing iNOS expression and NO production. Inhibiting IL-15 signaling also blocked the activation of the mitogen-activated protein kinase (MAPK) pathways ERK1/2 and p38. The major consequence of these inhibitory effects, analyzed using cytokine antibody arrays, was a severe decrease in the production of chemokines, cytokines and growth factors, like CCL17, CCL19, IL-12, or TIMP-1, that are essential for the development of the phenotypic changes of glial activation. In conclusion, activation of the IL-15 system seems a necessary step for the development of glial reactivity and the regulation of the physiology of glial cells. Modulating IL-15 activity opens the possibility of developing new strategies to control gliotic events upon inflammatory stimulation.
Collapse
Affiliation(s)
- Diego Gomez-Nicola
- Experimental Neurology Unit, Hospital Nacional de Parapléjicos, Toledo, Spain
| | | | | |
Collapse
|
50
|
Gomez-Nicola D, Spagnolo A, Guaza C, Nieto-Sampedro M. Aggravated experimental autoimmune encephalomyelitis in IL-15 knockout mice. Exp Neurol 2010; 222:235-42. [PMID: 20070942 DOI: 10.1016/j.expneurol.2009.12.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/24/2009] [Accepted: 12/30/2009] [Indexed: 12/22/2022]
Abstract
IL-15 initially identified as a T proliferating cytokine has several structural and biological similarities with IL-2 and has been associated with a number of autoimmune diseases. Because of the scarcity of information available on the role of IL-15 in MS pathogenesis, we have investigated how the absence of IL-15 affected the development of experimental autoimmune encephalomyelitis, a mouse model of MS. Following immunization of IL-15(-/-) and C57BL/6 mice with MOG(35-55), we observed a more severe neurological impairment in the IL-15 knockout mice than in the wild-type group. The enhanced disease severity in IL-15(-/-) mice was associated with greater demyelination in the spinal cord, increased immune cell infiltration and inflammation. These events may be related to the higher CD4/CD8 ratio and the almost absent NK cell activity, congenital immune features of IL-15KO mice. Moreover, we found that the fractalkine receptor CX3CR1 was overexpressed in the spinal cord of IL-15(-/-) mice, mainly localized on infiltrating CD8(+) T cells. How these findings are contributing to the aggravated EAE development in IL-15 KO mice remain unclear and need to be further investigated.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- CD4 Antigens/metabolism
- CD8 Antigens/metabolism
- CX3C Chemokine Receptor 1
- Cytokines/metabolism
- Demyelinating Diseases/etiology
- Demyelinating Diseases/pathology
- Eliminative Behavior, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Flow Cytometry
- Gene Expression Regulation
- Glycoproteins
- Interleukin-15/deficiency
- Killer Cells, Natural/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin Basic Protein/metabolism
- Myelin-Oligodendrocyte Glycoprotein
- Neutrophil Infiltration/immunology
- Peptide Fragments
- Receptors, Chemokine/metabolism
- Spinal Cord/immunology
- Spinal Cord/physiopathology
Collapse
Affiliation(s)
- Diego Gomez-Nicola
- Neural Plasticity Group, Functional and Systems Neurobiology Department, Instituto Cajal, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | | | | | | |
Collapse
|