1
|
Jiang W, Liu X, Chen Y, Liu M, Yuan J, Nie M, Fan Y, Wu D, Qian Y, Sha Z, Dong S, Wu C, Liu T, Huang J, Zhang J, Gao C, Jiang R. CD4 + CD11b + T cells infiltrate and aggravate the traumatic brain injury depending on brain-to-cervical lymph node signaling. CNS Neurosci Ther 2024; 30:e14673. [PMID: 38468459 PMCID: PMC10928342 DOI: 10.1111/cns.14673] [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: 09/01/2023] [Revised: 12/28/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024] Open
Abstract
AIM We aim to identify the specific CD4+ T-cell subtype influenced by brain-to-CLN signaling and explore their role during the acute phase of traumatic brain injury (TBI). METHOD Cervical lymphadenectomy or cervical afferent lymphatic ligation was performed before TBI. Cytokine array and western blot were used to detect cytokines, while the motor function was assessed using mNss and rotarod test. CD4+ T-cell subtypes in blood, brain, and CLNs were analyzed with Cytometry by time-of-flight analysis (CyTOF) or fluorescence-activated cell sorting (FACS). Brain edema and volume changes were measured by 9.4T MRI. Neuronal apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. RESULTS Cervical lymphadenectomy and ligation of cervical lymphatic vessels resulted in a decreased infiltration of CD4+ T cells, specifically CD11b-positive CD4+ T cells, within the affected region. The population of CD4+ CD11b+ T cells increased in ligated CLNs, accompanied by a decrease in the average fluorescence intensity of sphingosine-1-phosphate receptor-1 (S1PR1) on these cells. Administration of CD4+ CD11b+ T cells sorted from CLNs into the lateral ventricle reversed the attenuated neurologic deficits, brain edema, and lesion volume following cervical lymphadenectomy. CONCLUSION The infiltration of CD4+ CD11b+ T cells exacerbates secondary brain damage in TBI, and this process is modulated by brain-to-CLN signaling.
Collapse
Affiliation(s)
- Weiwei Jiang
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Xuanhui Liu
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Yupeng Chen
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Mingqi Liu
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Jiangyuan Yuan
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Meng Nie
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Yibing Fan
- Department of NeurosurgeryTianjin First Central HospitalTianjinChina
| | - Di Wu
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Yu Qian
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Zhuang Sha
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Shiying Dong
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Chenrui Wu
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Tao Liu
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Jinhao Huang
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Jianning Zhang
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Chuang Gao
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| | - Rongcai Jiang
- Department of NeurosurgeryGeneral Hospital of Tianjin Medical UniversityTianjinChina
- State Key Laboratory of Experimental HematologyTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post‐Neuroinjury Neurorepair and Regeneration in Central Nervous SystemTianjin Medical University General Hospital, Ministry of EducationTianjinChina
| |
Collapse
|
2
|
Pischiutta F, Caruso E, Cavaleiro H, Salgado AJ, Loane DJ, Zanier ER. Mesenchymal stromal cell secretome for traumatic brain injury: Focus on immunomodulatory action. Exp Neurol 2022; 357:114199. [PMID: 35952763 DOI: 10.1016/j.expneurol.2022.114199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/14/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022]
Abstract
The severity and long-term consequences of brain damage in traumatic brain injured (TBI) patients urgently calls for better neuroprotective/neuroreparative strategies for this devastating disorder. Mesenchymal stromal cells (MSCs) hold great promise and have been shown to confer neuroprotection in experimental TBI, mainly through paracrine mechanisms via secreted bioactive factors (i.e. secretome), which indicates significant potential for a cell-free neuroprotective approach. The secretome is composed of cytokines, chemokines, growth factors, proteins, lipids, nucleic acids, metabolites, and extracellular vesicles; it may offer advantages over MSCs in terms of delivery, safety, and variability of therapeutic response for brain injury. Immunomodulation by molecular factors secreted by MSCs is considered to be a key mechanism involved in their multi-potential therapeutic effects. Regulated neuroinflammation is required for healthy remodeling of central nervous system during development and adulthood. Moreover, immune cells and their secreted factors can also contribute to tissue repair and neurological recovery following acute brain injury. However, a chronic and maladaptive neuroinflammatory response can exacerbate TBI and contribute to progressive neurodegeneration and long-term neurological impairments. Here, we review the evidence for MSC-derived secretome as a therapy for TBI. Our framework incorporates a detailed analysis of in vitro and in vivo studies investigating the effects of the secretome on clinically relevant neurological and histopathological outcomes. We also describe the activation of immune cells after TBI and the immunomodulatory properties exerted by mediators released in the secretome. We then describe how ageing modifies central and systemic immune responses to TBI and discuss challenges and opportunities of developing secretome based neuroprotective therapies for elderly TBI populations. Finally, strategies aimed at modulating the secretome in order to boost its efficacy for TBI will also be discussed.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| | - Enrico Caruso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy; Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Helena Cavaleiro
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; Stemmatters, Biotechnology and Regenerative Medicine, Guimarães, Portugal
| | - Antonio J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Elisa R Zanier
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy.
| |
Collapse
|
3
|
Bao W, Lin Y, Chen Z. The Peripheral Immune System and Traumatic Brain Injury: Insight into the role of T-helper cells. Int J Med Sci 2021; 18:3644-3651. [PMID: 34790036 PMCID: PMC8579286 DOI: 10.7150/ijms.46834] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence suggests that immune-inflammatory processes are key elements in the physiopathological events associated with traumatic brain injury (TBI). TBI is followed by T-cell-specific immunological changes involving several subsets of T-helper cells and the cytokines they produce; these processes can have opposite effects depending on the disease course and cytokine concentrations. Efforts are underway to identify the T-helper cells and cytokine profiles associated with prognosis. These predictors may eventually serve as effective treatment targets to decrease morbidity and mortality and to improve the management of TBI patients. Here, we review the immunological response to TBI, the possible molecular mechanisms of this response, and therapeutic strategies to address it.
Collapse
Affiliation(s)
| | | | - Zuobing Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
A new inflammatory parameter can predict delayed intracranial hemorrhage following ventriculoperitoneal shunt. Sci Rep 2021; 11:13763. [PMID: 34215829 PMCID: PMC8253783 DOI: 10.1038/s41598-021-93315-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Delayed intracerebral hemorrhage (DICH) secondary to ventriculoperitoneal (VP) shunt is considered to be a potentially severe event. This study aimed to investigate the association between a ratio of postoperative neutrophil-to-lymphocyte ratio to preoperative neutrophil-to-lymphocyte ratio (NLRR) and DICH secondary to VP shunt. We performed a retrospective review of patients who underwent VP shunt between January 2016 and June 2020. Multivariable logistic regression analysis was used to assess the association of DICH and NLRR. Then patients were divided into two groups according to the optimal cut-off point of NLRR, propensity score matching (PSM) method was performed to reconfirm the result. A total of 130 patients were enrolled and DICH occurred in 29 patients. Elevated NLRR and history of craniotomy were independent risk factors for DICH secondary to VP shunt. The optimal cut off point of NLRR was 2.05, and the sensitivity was 89.7%, the specificity was 63.4%. Patients with NLRR > 2.05 had much higher incidence of DICH (40.6% vs 4.5%). Our finding suggested that DICH following VP shunt was not a rare complication and elevated NLRR could independently predict DICH. Inflammatory responses might play an important role in the development of DICH following VP shunt.
Collapse
|
5
|
Hegde RN, Chiki A, Petricca L, Martufi P, Arbez N, Mouchiroud L, Auwerx J, Landles C, Bates GP, Singh-Bains MK, Dragunow M, Curtis MA, Faull RL, Ross CA, Caricasole A, Lashuel HA. TBK1 phosphorylates mutant Huntingtin and suppresses its aggregation and toxicity in Huntington's disease models. EMBO J 2020; 39:e104671. [PMID: 32757223 PMCID: PMC7459410 DOI: 10.15252/embj.2020104671] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/17/2022] Open
Abstract
Phosphorylation of the N‐terminal domain of the huntingtin (HTT) protein has emerged as an important regulator of its localization, structure, aggregation, clearance and toxicity. However, validation of the effect of bona fide phosphorylation in vivo and assessing the therapeutic potential of targeting phosphorylation for the treatment of Huntington's disease (HD) require the identification of the enzymes that regulate HTT phosphorylation. Herein, we report the discovery and validation of a kinase, TANK‐binding kinase 1 (TBK1), that efficiently phosphorylates full‐length and N‐terminal HTT fragments in vitro (at S13/S16), in cells (at S13) and in vivo. TBK1 expression in HD models (cells, primary neurons, and Caenorhabditis elegans) increases mutant HTT exon 1 phosphorylation and reduces its aggregation and cytotoxicity. We demonstrate that the TBK1‐mediated neuroprotective effects are due to phosphorylation‐dependent inhibition of mutant HTT exon 1 aggregation and an increase in autophagic clearance of mutant HTT. These findings suggest that upregulation and/or activation of TBK1 represents a viable strategy for the treatment of HD by simultaneously lowering mutant HTT levels and blocking its aggregation.
Collapse
Affiliation(s)
- Ramanath Narayana Hegde
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Anass Chiki
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lara Petricca
- Department of Neuroscience, IRBM Science Park, Rome, Italy
| | - Paola Martufi
- Department of Neuroscience, IRBM Science Park, Rome, Italy
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry and Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laurent Mouchiroud
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Christian Landles
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - Gillian P Bates
- Huntington's Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - Malvindar K Singh-Bains
- Centre for Brain Research, Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard Lm Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
6
|
Sabouri E, Majdi A, Jangjui P, Rahigh Aghsan S, Naseri Alavi SA. Neutrophil-to-Lymphocyte Ratio and Traumatic Brain Injury: A Review Study. World Neurosurg 2020; 140:142-147. [DOI: 10.1016/j.wneu.2020.04.185] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 11/28/2022]
|
7
|
The immunological response to traumatic brain injury. J Neuroimmunol 2019; 332:112-125. [DOI: 10.1016/j.jneuroim.2019.04.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/30/2022]
|
8
|
Van Dyken P, Lacoste B. Impact of Metabolic Syndrome on Neuroinflammation and the Blood-Brain Barrier. Front Neurosci 2018; 12:930. [PMID: 30618559 PMCID: PMC6297847 DOI: 10.3389/fnins.2018.00930] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022] Open
Abstract
Metabolic syndrome, which includes diabetes and obesity, is one of the most widespread medical conditions. It induces systemic inflammation, causing far reaching effects on the body that are still being uncovered. Neuropathologies triggered by metabolic syndrome often result from increased permeability of the blood-brain-barrier (BBB). The BBB, a system designed to restrict entry of toxins, immune cells, and pathogens to the brain, is vital for proper neuronal function. Local and systemic inflammation induced by obesity or type 2 diabetes mellitus can cause BBB breakdown, decreased removal of waste, and increased infiltration of immune cells. This leads to disruption of glial and neuronal cells, causing hormonal dysregulation, increased immune sensitivity, or cognitive impairment depending on the affected brain region. Inflammatory effects of metabolic syndrome have been linked to neurodegenerative diseases. In this review, we discuss the effects of obesity and diabetes-induced inflammation on the BBB, the roles played by leptin and insulin resistance, as well as BBB changes occurring at the molecular level. We explore signaling pathways including VEGF, HIFs, PKC, Rho/ROCK, eNOS, and miRNAs. Finally, we discuss the broader implications of neural inflammation, including its connection to Alzheimer's disease, multiple sclerosis, and the gut microbiome.
Collapse
Affiliation(s)
- Peter Van Dyken
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Bhattacharya A, Derecki NC, Lovenberg TW, Drevets WC. Role of neuro-immunological factors in the pathophysiology of mood disorders. Psychopharmacology (Berl) 2016; 233:1623-36. [PMID: 26803500 DOI: 10.1007/s00213-016-4214-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 01/12/2016] [Indexed: 12/13/2022]
Abstract
Mood disorders, despite the widespread availability of monoamine-based antidepressant treatments, are associated with persistently high rates of disability, together with elevated rates of mortality due to suicide, cardiovascular disease, and other causes. The development of more effective treatments has been hindered by the lack of knowledge about the etiology and pathogenesis of mood disorders. An emerging area of science that promises novel pathways to antidepressant and mood stabilizing therapies surrounds evidence that immune cells and their signaling play a major role in the pathophysiology of major depressive disorder (MDD) and bipolar disorder (BD). Here, we review evidence that the release of neuroactive cytokines, particularly interleukins such as IL-1β, IL-6, and TNF-α, is altered in these disorders and discuss mechanisms such as the ATP-gated ion channel P2X7, through which cytokine signaling can influence neuro-glial interactions. Brain P2X7, an emerging target and antagonism of P2X7 holds promise as a novel mechanism for targeting treatment-resistant depression. We further discuss the role of microglia and astroglia in central neuroinflammation and their interaction with the peripheral immune system We present extant clinical evidence that bolsters the role of neuroinflammation and neuroactive cytokines in mood disorders. To that end, the role of clinical imaging by probing neuroinflammatory markers is also discussed briefly. Finally, we present data using preclinical neuroinflammation models that produce depression-like behaviors in experimental animals to identify neuroinflammatory mechanisms which may aid in novel neuroimmune target identification for the development of exciting pharmacological interventions in mood disorders.
Collapse
Affiliation(s)
| | - Noel C Derecki
- Neuroscience, Janssen Research and Development, LLC, San Diego, CA, 92121, USA
| | - Timothy W Lovenberg
- Neuroscience, Janssen Research and Development, LLC, San Diego, CA, 92121, USA
| | - Wayne C Drevets
- Neuroscience, Janssen Research and Development, LLC, Titusville, NJ, 08560, USA
| |
Collapse
|
10
|
Bhattacharya A, Drevets WC. Role of Neuro-Immunological Factors in the Pathophysiology of Mood Disorders: Implications for Novel Therapeutics for Treatment Resistant Depression. Curr Top Behav Neurosci 2016; 31:339-356. [PMID: 27677784 DOI: 10.1007/7854_2016_43] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mood disorders are associated with persistently high rates of morbidity and mortality, despite the widespread availability of antidepressant treatments. One limitation to extant therapeutic options has been that nearly all approved antidepressant pharmacotherapies exert a similar primary action of blocking monoamine transporters, and few options exist for transitioning treatment resistant patients to alternatives with distinct mechanisms. An emerging area of science that promises novel pathways to antidepressant and mood-stabilizing therapies has followed from evidence that immunological factors play major roles in the pathophysiology of at least some mood disorder subtypes. Here we review evidence that the compounds that reduce the release or signaling of neuroactive cytokines, particularly IL-1β, IL-6, and TNF-α, can exert antidepressant effects in subgroups of depressed patients who are identified by blood-based biomarkers associated with inflammation. Within this context we discuss the role of microglia in central neuroinflammation, and the interaction between the peripheral immune system and the central synaptic microenvironment during and after neuroinflammation. Finally we review data using preclinical neuroinflammation models that produce depression-like behaviors in experimental animals to guide the discovery of novel neuro-immune drug targets.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- Neuroscience Drug Discovery, Janssen Research & Development, LLC, Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| | - Wayne C Drevets
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
| |
Collapse
|
11
|
Blocking stroke-induced immunodeficiency increases CNS antigen-specific autoreactivity but does not worsen functional outcome after experimental stroke. J Neurosci 2015; 35:7777-94. [PMID: 25995466 DOI: 10.1523/jneurosci.1532-14.2015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Stroke-induced immunodepression (SIDS) is an essential cause of poststroke infections. Pharmacological inhibition of SIDS appears promising in preventing life-threatening infections in stroke patients. However, SIDS might represent an adaptive mechanism preventing autoreactive immune responses after stroke. To address this, we used myelin oligodendrocyte glycoprotein (MOG) T-cell receptor transgenic (2D2) mice where >80% of peripheral CD4(+) T cells express a functional receptor for MOG. We investigated in a murine model of middle cerebral artery occlusion the effect of blocking SIDS by inhibiting body's main stress axes, the sympathetic nervous system (SNS) with propranolol and the hypothalamic-pituitary-adrenal axis (HPA) with mifepristone. Blockade of both stress axes robustly reduced infarct volumes, decreased infection rate, and increased long-term survival of 2D2 and C57BL/6J wild-type mice. Despite these protective effects, blockade of SIDS increased CNS antigen-specific Type1 T helper cell (Th1) responses in the brains of 2D2 mice 14 d after middle cerebral artery occlusion. One month after experimental stroke, 2D2 mice developed signs of polyradiculitis, which were diminished by SIDS blockade. Adoptive transfer of CD4(+) T cells, isolated from 2D2 mice, into lymphocyte-deficient Rag-1KO mice did not reveal differences between SIDS blockade and vehicle treatment in functional long-term outcome after stroke. In conclusion, inhibiting SIDS by pharmacological blockade of body's stress axes increases autoreactive CNS antigen-specific T-cell responses in the brain but does not worsen functional long-term outcome after experimental stroke, even in a mouse model where CNS antigen-specific autoreactive T-cell responses are boosted.
Collapse
|
12
|
Immunization with a Myelin-Derived Antigen Activates the Brain's Choroid Plexus for Recruitment of Immunoregulatory Cells to the CNS and Attenuates Disease Progression in a Mouse Model of ALS. J Neurosci 2015; 35:6381-93. [PMID: 25904790 DOI: 10.1523/jneurosci.3644-14.2015] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating fatal motor neuron disease, for which there is currently no cure or effective treatment. In this disease, local neuroinflammation develops along the disease course and contributes to its rapid progression. In several models of CNS pathologies, circulating immune cells were shown to display an indispensable role in the resolution of the neuroinflammatory response. The recruitment of such cells to the CNS involves activation of the choroid plexus (CP) of the brain for leukocyte trafficking, through a mechanism that requires IFN-γ signaling. Here, we found that in the mutant SOD1(G93A) (mSOD1) mouse model of ALS, the CP does not support leukocyte trafficking during disease progression, due to a local reduction in IFN-γ levels. Therapeutic immunization of mSOD1 mice with a myelin-derived peptide led to CP activation, and was followed by the accumulation of immunoregulatory cells, including IL-10-producing monocyte-derived macrophages and Foxp3(+) regulatory T cells, and elevation of the neurotrophic factors IGF-1 and GDNF in the diseased spinal cord parenchyma. The immunization resulted in the attenuation of disease progression and an increased life expectancy of the mSOD1 mice. Collectively, our results demonstrate that recruitment of immunoregulatory cells to the diseased spinal cord in ALS, needed for fighting off the pathology, can be enhanced by transiently boosting peripheral immunity to myelin antigens.
Collapse
|
13
|
Breaking peripheral immune tolerance to CNS antigens in neurodegenerative diseases: Boosting autoimmunity to fight-off chronic neuroinflammation. J Autoimmun 2014; 54:8-14. [DOI: 10.1016/j.jaut.2014.08.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 08/12/2014] [Indexed: 12/14/2022]
|
14
|
Urra X, Miró F, Chamorro A, Planas AM. Antigen-specific immune reactions to ischemic stroke. Front Cell Neurosci 2014; 8:278. [PMID: 25309322 PMCID: PMC4162361 DOI: 10.3389/fncel.2014.00278] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/24/2022] Open
Abstract
Brain proteins are detected in the cerebrospinal fluid (CSF) and blood of stroke patients and their concentration is related to the extent of brain damage. Antibodies against brain antigens develop after stroke, suggesting a humoral immune response to the brain injury. Furthermore, induced immune tolerance is beneficial in animal models of cerebral ischemia. The presence of circulating T cells sensitized against brain antigens, and antigen presenting cells (APCs) carrying brain antigens in draining lymphoid tissue of stroke patients support the notion that stroke might induce antigen-specific immune responses. After stroke, brain proteins that are normally hidden from the periphery, inflammatory mediators, and danger signals can exit the brain through several efflux routes. They can reach the blood after leaking out of the damaged blood-brain barrier (BBB) or following the drainage of interstitial fluid to the dural venous sinus, or reach the cervical lymph nodes through the nasal lymphatics following CSF drainage along the arachnoid sheaths of nerves across the nasal submucosa. The route and mode of access of brain antigens to lymphoid tissue could influence the type of response. Central and peripheral tolerance prevents autoimmunity, but the actual mechanisms of tolerance to brain antigens released into the periphery in the presence of inflammation, danger signals, and APCs, are not fully characterized. Stroke does not systematically trigger autoimmunity, but under certain circumstances, such as pronounced systemic inflammation or infection, autoreactive T cells could escape the tolerance controls. Further investigation is needed to elucidate whether antigen-specific immune events could underlie neurological complications impairing recovery from stroke.
Collapse
Affiliation(s)
- Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic Barcelona, Spain ; August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Francesc Miró
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Angel Chamorro
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic Barcelona, Spain ; August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Anna M Planas
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain ; Department of Brain Ischemia and Neurodegeneration, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC) Barcelona, Spain
| |
Collapse
|
15
|
Khailaie S, Bahrami F, Janahmadi M, Milanez-Almeida P, Huehn J, Meyer-Hermann M. A mathematical model of immune activation with a unified self-nonself concept. Front Immunol 2013; 4:474. [PMID: 24409179 PMCID: PMC3872974 DOI: 10.3389/fimmu.2013.00474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/06/2013] [Indexed: 12/14/2022] Open
Abstract
The adaptive immune system reacts against pathogenic nonself, whereas it normally remains tolerant to self. The initiation of an immune response requires a critical antigen(Ag)-stimulation and a critical number of Ag-specific T cells. Autoreactive T cells are not completely deleted by thymic selection and partially present in the periphery of healthy individuals that respond in certain physiological conditions. A number of experimental and theoretical models are based on the concept that structural differences discriminate self from nonself. In this article, we establish a mathematical model for immune activation in which self and nonself are not distinguished. The model considers the dynamic interplay of conventional T cells, regulatory T cells (Tregs), and IL-2 molecules and shows that the renewal rate ratio of resting Tregs to naïve T cells as well as the proliferation rate of activated T cells determine the probability of immune stimulation. The actual initiation of an immune response, however, relies on the absolute renewal rate of naïve T cells. This result suggests that thymic selection reduces the probability of autoimmunity by increasing the Ag-stimulation threshold of self reaction which is established by selection of a low number of low-avidity autoreactive T cells balanced with a proper number of Tregs. The stability analysis of the ordinary differential equation model reveals three different possible immune reactions depending on critical levels of Ag-stimulation: a subcritical stimulation, a threshold stimulation inducing a proper immune response, and an overcritical stimulation leading to chronic co-existence of Ag and immune activity. The model exhibits oscillatory solutions in the case of persistent but moderate Ag-stimulation, while the system returns to the homeostatic state upon Ag clearance. In this unifying concept, self and nonself appear as a result of shifted Ag-stimulation thresholds which delineate these three regimes of immune activation.
Collapse
Affiliation(s)
- Sahamoddin Khailaie
- Department of Systems Immunology, Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Fariba Bahrami
- CIPCE, School of Electrical and Computer Engineering, College of Engineering, University of Tehran , Tehran , Iran
| | - Mahyar Janahmadi
- Neuroscience Research Centre and Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Pedro Milanez-Almeida
- Department of Experimental Immunology, Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology, Helmholtz Centre for Infection Research , Braunschweig , Germany ; Bio Centre for Life Science, Technische Universität Braunschweig , Braunschweig , Germany
| |
Collapse
|
16
|
Schwartz M, Baruch K. The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus. EMBO J 2013; 33:7-22. [PMID: 24357543 DOI: 10.1002/embj.201386609] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inflammation is an integral part of the body's physiological repair mechanism, unless it remains unresolved and becomes pathological, as evident in the progressive nature of neurodegeneration. Based on studies from outside the central nervous system (CNS), it is now understood that the resolution of inflammation is an active process, which is dependent on well-orchestrated innate and adaptive immune responses. Due to the immunologically privileged status of the CNS, such resolution mechanism has been mostly ignored. Here, we discuss resolution of neuroinflammation as a process that depends on a network of immune cells operating in a tightly regulated sequence, involving the brain's choroid plexus (CP), a unique neuro-immunological interface, positioned to integrate signals it receives from the CNS parenchyma with signals coming from circulating immune cells, and to function as an on-alert gate for selective recruitment of inflammation-resolving leukocytes to the inflamed CNS parenchyma. Finally, we propose that functional dysregulation of the CP reflects a common underlying mechanism in the pathophysiology of neurodegenerative diseases, and can thus serve as a potential novel target for therapy.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
17
|
Baruch K, Schwartz M. CNS-specific T cells shape brain function via the choroid plexus. Brain Behav Immun 2013; 34:11-6. [PMID: 23597431 DOI: 10.1016/j.bbi.2013.04.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/07/2013] [Accepted: 04/08/2013] [Indexed: 12/17/2022] Open
Abstract
Adaptive immunity was repeatedly shown to play a role in maintaining lifelong brain function. Under physiological conditions, this activity was associated with CD4+ T cells specific for brain self-antigens. Nevertheless, direct interactions of T cells with the healthy neuronal parenchyma are hardly detectable. Recent studies have identified the brain's choroid plexus (CP) as an active neuro-immunological interface, enriched with CNS-specific CD4+ T cells. Strategically positioned for receiving signals from both the central nervous system (CNS) through the cerebrospinal fluid (CSF), and from the circulation through epithelium-immune cell interactions, the CP has recently been recognized as an important immunological compartment in maintaining and restoring brain homeostasis/allostasis. Here, we propose that CNS-specific T cells shape brain function via the CP, and suggest this immunological control to be lost as part of aging, in general, and immune senescence, in particular. Accordingly, the CP may serve as a novel target for immunomodulation to restore brain equilibrium.
Collapse
Affiliation(s)
- Kuti Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
18
|
Jiang Y, Zou Y, Chen S, Zhu C, Wu A, Liu Y, Ma L, Zhu D, Ma X, Liu M, Kang Z, Pi R, Peng F, Wang Q, Chen X. The anti-inflammatory effect of donepezil on experimental autoimmune encephalomyelitis in C57 BL/6 mice. Neuropharmacology 2013; 73:415-24. [DOI: 10.1016/j.neuropharm.2013.06.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 11/26/2022]
|
19
|
Ishii H, Tanabe S, Ueno M, Kubo T, Kayama H, Serada S, Fujimoto M, Takeda K, Naka T, Yamashita T. ifn-γ-dependent secretion of IL-10 from Th1 cells and microglia/macrophages contributes to functional recovery after spinal cord injury. Cell Death Dis 2013; 4:e710. [PMID: 23828573 PMCID: PMC3730408 DOI: 10.1038/cddis.2013.234] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 05/06/2013] [Accepted: 06/03/2013] [Indexed: 01/23/2023]
Abstract
Transfer of type-1 helper T-conditioned (Th1-conditioned) cells promotes functional recovery with enhanced axonal remodeling after spinal cord injury (SCI). This study explored the molecular mechanisms underlying the beneficial effects of pro-inflammatory Th1-conditioned cells after SCI. The effect of Th1-conditioned cells from interferon-γ (ifn-γ) knockout mice (ifn-γ−/− Th1 cells) on the recovery after SCI was reduced. Transfer of Th1-conditioned cells led to the activation of microglia (MG) and macrophages (MΦs), with interleukin 10 (IL-10) upregulation. This upregulation of IL-10 was reduced when ifn-γ−/− Th1 cells were transferred. Intrathecal neutralization of IL-10 in the spinal cord attenuated the effects of Th1-conditioned cells. Further, IL-10 is robustly secreted from Th1-conditioned cells in an ifn-γ-dependent manner. Th1-conditioned cells from interleukin 10 knockout (il-10−/−) mice had no effects on recovery from SCI. These findings demonstrate that ifn-γ-dependent secretion of IL-10 from Th1 cells, as well as native MG/MΦs, is required for the promotion of motor recovery after SCI.
Collapse
Affiliation(s)
- H Ishii
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Vaughn CN, Iafrate JL, Henley JB, Stevenson EK, Shlifer IG, Jones TB. Cellular Neuroinflammation in a Lateral Forceps Compression Model of Spinal Cord Injury. Anat Rec (Hoboken) 2013; 296:1229-46. [DOI: 10.1002/ar.22730] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/31/2013] [Accepted: 05/17/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Chloe N. Vaughn
- Biomedical Sciences Program; Midwestern University; Glendale Arizona
| | - Julia L. Iafrate
- College of Osteopathic Medicine; Midwestern University; Glendale Arizona
| | | | | | - Igor G. Shlifer
- College of Osteopathic Medicine; Midwestern University; Glendale Arizona
| | - T. Bucky Jones
- College of Osteopathic Medicine; Midwestern University; Glendale Arizona
- Department of Anatomy; Midwestern University; Glendale Arizona
| |
Collapse
|
21
|
Modulation of neural stem/progenitor cell proliferation during experimental Herpes Simplex encephalitis is mediated by differential FGF-2 expression in the adult brain. Neurobiol Dis 2013; 58:144-55. [PMID: 23748078 DOI: 10.1016/j.nbd.2013.05.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 05/07/2013] [Accepted: 05/22/2013] [Indexed: 11/20/2022] Open
Abstract
Neural stem cells (NSCs) respond to inflammatory cues induced during brain injury and are thought to be involved in recovery from brain damage. Little is known about NSC response during brain infections. The present study evaluated NSC proliferation during Herpes Simplex Virus-1 brain infection. Total numbers of nestin(+) NSCs increased significantly in infected brains at 6 days post infection (p.i.). However, by 15 days p.i. the nestin(+) population decreased significantly below levels observed in uninfected brains and remained depressed through 30 days p.i. This initial increase in NSC population occurred concurrently with increased brain cell proliferation, which peaked at 3 days p.i. On closer examination, we found that while actively proliferating Sox2(+) NSCs increased in number at 6 days p.i., proliferating DCX(+) neuroblasts contributed to the increased response at 3 days p.i. However, overall proliferation decreased steadily from 15 days p.i. to below control levels. To determine the mechanisms involved in altering NSC proliferation, neurotrophin and growth factor expression profiles were assessed. FGF-2 gene expression increased at 5 days p.i. and was robustly down-regulated at 15 days p.i. (>1000-fold), which was further confirmed by increased FGF-2 immunostaining around the lateral ventricles. Furthermore, supplementing infected animals with recombinant FGF-2, at 15 days p.i., significantly increased the number of proliferating brain cells. These findings demonstrate that the temporal changes in NSC proliferation are mediated through the regulation of FGF-2 and that the NSC niche may benefit from supplementation with FGF-2 during HSV-1 brain infection.
Collapse
|
22
|
Baruch K, Ron-Harel N, Gal H, Deczkowska A, Shifrut E, Ndifon W, Mirlas-Neisberg N, Cardon M, Vaknin I, Cahalon L, Berkutzki T, Mattson MP, Gomez-Pinilla F, Friedman N, Schwartz M. CNS-specific immunity at the choroid plexus shifts toward destructive Th2 inflammation in brain aging. Proc Natl Acad Sci U S A 2013; 110:2264-9. [PMID: 23335631 PMCID: PMC3568380 DOI: 10.1073/pnas.1211270110] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The adaptive arm of the immune system has been suggested as an important factor in brain function. However, given the fact that interactions of neurons or glial cells with T lymphocytes rarely occur within the healthy CNS parenchyma, the underlying mechanism is still a mystery. Here we found that at the interface between the brain and blood circulation, the epithelial layers of the choroid plexus (CP) are constitutively populated with CD4(+) effector memory cells with a T-cell receptor repertoire specific to CNS antigens. With age, whereas CNS specificity in this compartment was largely maintained, the cytokine balance shifted in favor of the T helper type 2 (Th2) response; the Th2-derived cytokine IL-4 was elevated in the CP of old mice, relative to IFN-γ, which decreased. We found this local cytokine shift to critically affect the CP epithelium, triggering it to produce the chemokine CCL11 shown to be associated with cognitive dysfunction. Partial restoration of cognitive ability in aged mice, by lymphopenia-induced homeostasis-driven proliferation of memory T cells, was correlated with restoration of the IL-4:IFN-γ ratio at the CP and modulated the expression of plasticity-related genes at the hippocampus. Our data indicate that the cytokine milieu at the CP epithelium is affected by peripheral immunosenescence, with detrimental consequences to the aged brain. Amenable to immunomodulation, this interface is a unique target for arresting age-related cognitive decline.
Collapse
Affiliation(s)
| | - Noga Ron-Harel
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Hilah Gal
- Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Eric Shifrut
- Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Wilfred Ndifon
- Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224; and
| | - Fernando Gomez-Pinilla
- Departments of Integrative Biology and Physiology and
- Neurosurgery, University of California, Los Angeles, CA 90095
| | - Nir Friedman
- Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
23
|
Ishii H, Jin X, Ueno M, Tanabe S, Kubo T, Serada S, Naka T, Yamashita T. Adoptive transfer of Th1-conditioned lymphocytes promotes axonal remodeling and functional recovery after spinal cord injury. Cell Death Dis 2012; 3:e363. [PMID: 22875000 PMCID: PMC3434665 DOI: 10.1038/cddis.2012.106] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The role of T lymphocytes in central nervous system (CNS) injuries is controversial, with inconsistent results reported concerning the effects of T-lymphocyte transfer on spinal cord injury (SCI). Here, we demonstrate that a specific T-lymphocyte subset enhances functional recovery after contusion SCI in mice. Intraperitoneal adoptive transfer of type 1 helper T (Th1)-conditioned cells 4 days after SCI promoted recovery of locomotor activity and tactile sensation and concomitantly induced regrowth of corticospinal tract and serotonergic fibers. However, neither type 2 helper T (Th2)- nor IL-17-producing helper T (Th17)-conditioned cells had such effects. Activation of microglia and macrophages were observed in the spinal cords of Th1-transfered mice after SCI. Specifically, M2 subtype of microglia/macrophages was upregulated after Th1 cell transfer. Neutralization of interleukin 10 secreted by Th1-conditioned cells significantly attenuated the beneficial effects by Th1-conditioned lymphocytes after SCI. We also found that Th1-conditioned lymphocytes secreted significantly higher levels of neurotrophic factor, neurotrophin 3 (NT-3), than Th2- or Th17-conditioned cells. Thus, adoptive transfer of pro-inflammatory Th1-conditioned cells has neuroprotective effects after SCI, with prospective implications in immunomodulatory treatment of CNS injury.
Collapse
Affiliation(s)
- H Ishii
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Kuerten S, Lehmann PV. The Immune Pathogenesis of Experimental Autoimmune Encephalomyelitis: Lessons Learned for Multiple Sclerosis? J Interferon Cytokine Res 2011; 31:907-16. [DOI: 10.1089/jir.2011.0072] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Stefanie Kuerten
- Department of Anatomy I, University Hospitals of Cologne, Cologne, Germany
| | - Paul V. Lehmann
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
- Cellular Technology Limited, Shaker Heights, Cleveland, Ohio
| |
Collapse
|
25
|
Tysseling VM, Mithal DS, Sahni V, Birch D, Jung H, Belmadani A, Miller RJ, Kessler JA. SDF1 in the dorsal corticospinal tract promotes CXCR4+ cell migration after spinal cord injury. J Neuroinflammation 2011; 8:16. [PMID: 21324162 PMCID: PMC3050722 DOI: 10.1186/1742-2094-8-16] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 02/16/2011] [Indexed: 11/24/2022] Open
Abstract
Background Stromal cell-derived factor-1 (SDF1) and its major signaling receptor, CXCR4, were initially described in the immune system; however, they are also expressed in the nervous system, including the spinal cord. After spinal cord injury, the blood brain barrier is compromised, opening the way for chemokine signaling between these two systems. These experiments clarified prior contradictory findings on normal expression of SDF1 and CXCR4 as well as examined the resulting spinal cord responses resulting from this signaling. Methods These experiments examined the expression and function of SDF1 and CXCR4 in the normal and injured adult mouse spinal cord primarily using CXCR4-EGFP and SDF1-EGFP transgenic reporter mice. Results In the uninjured spinal cord, SDF1 was expressed in the dorsal corticospinal tract (dCST) as well as the meninges, whereas CXCR4 was found only in ependymal cells surrounding the central canal. After spinal cord injury (SCI), the pattern of SDF1 expression did not change rostral to the lesion but it disappeared from the degenerating dCST caudally. By contrast, CXCR4 expression changed dramatically after SCI. In addition to the CXCR4+ cells in the ependymal layer, numerous CXCR4+ cells appeared in the peripheral white matter and in the dorsal white matter localized between the dorsal corticospinal tract and the gray matter rostral to the lesion site. The non-ependymal CXCR4+ cells were found to be NG2+ and CD11b+ macrophages that presumably infiltrated through the broken blood-brain barrier. One population of macrophages appeared to be migrating towards the dCST that contains SDF1 rostral to the injury but not towards the caudal dCST in which SDF1 is no longer present. A second population of the CXCR4+ macrophages was present near the SDF1-expressing meningeal cells. Conclusions These observations suggest that attraction of CXCR4+ macrophages is part of a programmed response to injury and that modulation of the SDF1 signaling system may be important for regulating the inflammatory response after SCI.
Collapse
Affiliation(s)
- Vicki M Tysseling
- Northwestern University's Feinberg School of Medicine, Department of Neurology, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Yong J, Lacan G, Dang H, Hsieh T, Middleton B, Wasserfall C, Tian J, Melega WP, Kaufman DL. BCG vaccine-induced neuroprotection in a mouse model of Parkinson's disease. PLoS One 2011; 6:e16610. [PMID: 21304945 PMCID: PMC3031604 DOI: 10.1371/journal.pone.0016610] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 12/28/2010] [Indexed: 12/14/2022] Open
Abstract
There is a growing interest in using vaccination with CNS antigens to induce autoreactive T cell responses that home to damaged areas in the CNS and ameliorate neurodegenerative disease. Neuroprotective vaccine studies have focused on administering oligodendrocyte antigens or Copaxone® in complete Freund's adjuvant (CFA). Theoretical considerations, however, suggest that vaccination with a neuronal antigen may induce more robust neuroprotective immune responses. We assessed the neuroprotective potential of vaccines containing tyrosine hydroxylase (a neuronal protein involved in dopamine synthesis) or Copaxone® in CFA in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. Surprisingly, we observed that the main beneficial factor in these vaccines was the CFA. Since the major immunogenic component in CFA is Mycobacterium tuberculosis, which closely related to the bacille Calmette-Guérin (BCG) that is used in human vaccines, we tested BCG vaccination in the MPTP mouse model. We observed that BCG vaccination partially preserved markers of striatal dopamine system integrity and prevented an increase in activated microglia in the substantia nigra of MPTP-treated mice. These results support a new neuroprotective vaccine paradigm in which general (nonself-reactive) immune stimulation in the periphery can limit potentially deleterious microglial responses to a neuronal insult and exert a neurorestorative effect in the CNS. Accordingly, BCG vaccination may provide a new strategy to augment current treatments for a wide range of neuropathological conditions.
Collapse
Affiliation(s)
- Jing Yong
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Goran Lacan
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Terry Hsieh
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Blake Middleton
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Clive Wasserfall
- Department of Pathology, University of Florida, Gainesville, Florida, United States of America
| | - Jide Tian
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - William P. Melega
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Daniel L. Kaufman
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ishii H, Kubo T, Kumanogoh A, Yamashita T. Th1 cells promote neurite outgrowth from cortical neurons via a mechanism dependent on semaphorins. Biochem Biophys Res Commun 2010; 402:168-72. [PMID: 20946887 DOI: 10.1016/j.bbrc.2010.10.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 10/06/2010] [Indexed: 12/11/2022]
Abstract
The roles of T lymphocytes in the central nervous system (CNS) are diverse; their roles in the injured CNS have been reported to be both detrimental and advantageous. Hence, an investigation of the effects of specific subsets of T cells on neurons may provide an insight into the interaction between the nervous system and the immune system. In the present study, we demonstrate that a specific subset of T lymphocytes enhanced neurite outgrowth in vitro. When cultured T helper type 1 (Th1) cells were co-cultured with cortical neurons, neurite outgrowth from neurons was enhanced; however, the same was not observed when Th2 or naïve T cells were used. We observed that the promotion of neurite outgrowth by Th1 cells was completely inhibited by anti-interferon γ (IFN-γ) neutralizing antibody, but that IFN-γ did not directly promote neurite growth. Furthermore, experiments using knockout mice revealed that semaphorin 4A (Sema4A) but not Sema7A was required for the effect produced by Th1 cells. These results demonstrate that Sema4A and IFN-γ expressed in Th1 cells play a critical role in enhancing neurite outgrowth from cortical neurons.
Collapse
Affiliation(s)
- Hiroshi Ishii
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
28
|
Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Soloski MJ, Crow MT, Rabb H. Mycophenolate mofetil modifies kidney tubular injury and Foxp3+ regulatory T cell trafficking during recovery from experimental ischemia-reperfusion. Transpl Immunol 2010; 23:45-52. [PMID: 20412855 DOI: 10.1016/j.trim.2010.04.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 03/02/2010] [Accepted: 04/05/2010] [Indexed: 01/25/2023]
Abstract
Lymphocytes participate in the early pathogenesis of ischemia-reperfusion injury (IRI) in kidney; however, their role during repair is largely unknown. Recent data have shown that Foxp3(+) regulatory T cells (Tregs) traffic into kidney during healing from IRI and directly participate in repair. Since lymphocyte-targeting therapy is currently administered to prevent rejection during recovery from IRI in renal transplants, we hypothesized that mycophenolate mofetil (MMF) would alter Treg trafficking and kidney repair. C57BL/6J and T cell deficient mice underwent unilateral clamping of renal pedicle for 45 min, followed by reperfusion, and were sacrificed at day 10. Mice were treated with saline (C) or MMF (100mg/kg) i.p. daily starting at day 2 until sacrifice (n=5-12/group). MMF worsened kidney tubular damage compared to C at 10 days (cortex and outer medulla: p<0.05) in wild-type mice; tubular apoptotic index was increased in cortex in MMF group as well (p=0.01). MMF reduced the total number of kidney-infiltrating mononuclear cells (p<0.001 versus C) and the percentages of TCRbeta(+)CD4(+) and TCRbeta(+)CD8(+) T cells (p<0.01), but not natural killer (NK), NKT or B lymphocytes. MMF specifically reduced kidney Foxp3(+) Tregs (0.82+/-0.11% versus 1.75+/-0.17%, p<0.05). Tubular proliferative index and tissue levels of basic FGF were increased in MMF group (p<0.05), IL-10 and IL-6 were decreased (p<0.05). To evaluate if MMF effect occurred through non-lymphocytic cells, T cell deficient mice were treated with MMF. Tubular injury in T cell deficient mice was not affected by MMF treatment, though MMF-treated animals had increased VEGF and decreased PDGF-BB protein tissue levels compared to controls (p<0.05). Thus, MMF modifies the structural, epithelial proliferative and inflammatory response during healing, likely through effects on T cells and possibly Tregs. Kidney repair after IRI can be altered by agents that target lymphocytes.
Collapse
Affiliation(s)
- Maria Teresa Gandolfo
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Derecki NC, Privman E, Kipnis J. Rett syndrome and other autism spectrum disorders--brain diseases of immune malfunction? Mol Psychiatry 2010; 15:355-63. [PMID: 20177406 PMCID: PMC3368984 DOI: 10.1038/mp.2010.21] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuroimmunology was once referred to in terms of its pathological connotation only and was generally understood as covering the deleterious involvement of the immune system in various diseases and disorders of the central nervous system (CNS). However, our conception of the function of the immune system in the structure, function, and plasticity of the CNS has undergone a sea change after relevant discoveries over the past two decades, and continues to be challenged by more recent studies of neurodevelopment and cognition. This review summarizes the recent advances in understanding of immune-system participation in the development and functioning of the CNS under physiological conditions. Considering as an example Rett syndrome a devastating neurodevelopmental disease, we offer a hypothesis that might help to explain the part played by immune cells in its etiology, and hence suggests that the immune system might be a feasible therapeutic target for alleviation of some of the symptoms of this and other autism spectrum disorders.
Collapse
Affiliation(s)
- NC Derecki
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
,Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - E Privman
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
,Medical Scientist Training Program, University of Virginia, Charlottesville, VA, USA
| | - J Kipnis
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
,Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
,Medical Scientist Training Program, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
30
|
Stem/Precursor Cell-Based CNS Therapy: The Importance of Circumventing Immune Suppression by Transplanting Autologous Cells. Stem Cell Rev Rep 2010; 6:405-10. [DOI: 10.1007/s12015-010-9141-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med 2009; 6:e1000113. [PMID: 19636355 PMCID: PMC2707628 DOI: 10.1371/journal.pmed.1000113] [Citation(s) in RCA: 587] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 06/16/2009] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Although macrophages (MPhi) are known as essential players in wound healing, their contribution to recovery from spinal cord injury (SCI) is a subject of debate. The difficulties in distinguishing between different MPhi subpopulations at the lesion site have further contributed to the controversy and led to the common view of MPhi as functionally homogenous. Given the massive accumulation in the injured spinal cord of activated resident microglia, which are the native immune occupants of the central nervous system (CNS), the recruitment of additional infiltrating monocytes from the peripheral blood seems puzzling. A key question that remains is whether the infiltrating monocyte-derived MPhi contribute to repair, or represent an unavoidable detrimental response. The hypothesis of the current study is that a specific population of infiltrating monocyte-derived MPhi is functionally distinct from the inflammatory resident microglia and is essential for recovery from SCI. METHODS AND FINDINGS We inflicted SCI in adult mice, and tested the effect of infiltrating monocyte-derived MPhi on the recovery process. Adoptive transfer experiments and bone marrow chimeras were used to functionally distinguish between the resident microglia and the infiltrating monocyte-derived MPhi. We followed the infiltration of the monocyte-derived MPhi to the injured site and characterized their spatial distribution and phenotype. Increasing the naïve monocyte pool by either adoptive transfer or CNS-specific vaccination resulted in a higher number of spontaneously recruited cells and improved recovery. Selective ablation of infiltrating monocyte-derived MPhi following SCI while sparing the resident microglia, using either antibody-mediated depletion or conditional ablation by diphtheria toxin, impaired recovery. Reconstitution of the peripheral blood with monocytes resistant to ablation restored the lost motor functions. Importantly, the infiltrating monocyte-derived MPhi displayed a local anti-inflammatory beneficial role, which was critically dependent upon their expression of interleukin 10. CONCLUSIONS The results of this study attribute a novel anti-inflammatory role to a unique subset of infiltrating monocyte-derived MPhi in SCI recovery, which cannot be provided by the activated resident microglia. According to our results, limited recovery following SCI can be attributed in part to the inadequate, untimely, spontaneous recruitment of monocytes. This process is amenable to boosting either by active vaccination with a myelin-derived altered peptide ligand, which indicates involvement of adaptive immunity in monocyte recruitment, or by augmenting the naïve monocyte pool in the peripheral blood. Thus, our study sheds new light on the long-held debate regarding the contribution of MPhi to recovery from CNS injuries, and has potentially far-reaching therapeutic implications.
Collapse
|
32
|
Schwartz M, London A, Shechter R. Boosting T-cell immunity as a therapeutic approach for neurodegenerative conditions: The role of innate immunity. Neuroscience 2009; 158:1133-42. [DOI: 10.1016/j.neuroscience.2008.12.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 12/09/2008] [Accepted: 12/10/2008] [Indexed: 12/14/2022]
|
33
|
Abstract
Inflammatory and immune responses play important roles following ischaemic stroke. Inflammatory responses contribute to damage and also contribute to repair. Injury to tissue triggers an immune response. This is initiated through activation of the innate immune system. In stroke there is microglial activation. This is followed by an influx of lymphocytes and macrophages into the brain, triggered by production of pro-inflammatory cytokines. This inflammatory response contributes to further tissue injury. There is also a systemic immune response to stroke, and there is a degree of immunosuppression that may contribute to the stroke patient's risk of infection. This immunosuppressive response may also be protective, with regulatory lymphocytes producing cytokines and growth factors that are neuroprotective. The specific targets of the immune response after stroke are not known, and the details of the immune and inflammatory responses are only partly understood. The role of inflammation and immune responses after stroke is twofold. The immune system may contribute to damage after stroke, but may also contribute to repair processes. The possibility that some of the immune response after stroke may be neuroprotective is exciting and suggests that deliberate enhancement of these responses may be a therapeutic option.
Collapse
Affiliation(s)
- P A McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital and Neuroimmunology Research Unit, Central Clinical School, University of Queensland, Brisbane, Australia.
| | | |
Collapse
|
34
|
Schwartz M, Bukshpan S, Kunis G. Application of glatiramer acetate to neurodegenerative diseases beyond multiple sclerosis: the need for disease-specific approaches. BioDrugs 2008; 22:293-9. [PMID: 18778111 DOI: 10.2165/00063030-200822050-00002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Adaptive and innate immunity, if well controlled, contribute to the maintenance of the CNS, as well as to downregulation of adverse acute and chronic neurological conditions. T cells that recognize CNS antigens are needed to activate resident immune cells and to recruit blood-borne monocytes, which act to restore homeostasis and facilitate repair. However, boosting such a T-cell response in a risk-free way requires a careful choice of the antigen, carrier, and regimen. A single vaccination with CNS-derived peptides or their weak agonists reduces neuronal loss in animal models of acute neurodegeneration. Repeated injections are needed to maintain a long-lasting effect in chronic neurodegenerative conditions, yet the frequency of the injections seems to have a critical effect on the outcome. An example is glatiramer acetate, a compound that is administered in a daily regimen to patients with multiple sclerosis. A single injection of glatiramer acetate, with or without an adjuvant, is neuroprotective in some animal models of acute CNS injuries. However, in an animal model of amyotrophic lateral sclerosis, a single injection of adjuvant-free glatiramer acetate is insufficient, while daily injections are not only ineffective but can carry an increased risk of mortality in female mice.Thus, considering immune-based therapies as a single therapy, rather than as a family of therapies that are regimen dependent, may be misleading. Moreover, the vaccination regimen and administration of a compound, even one shown to be safe in humans for the treatment of a particular neurodegenerative disease, must be studied in preclinical experiments before it is tested in a clinical trial for a novel indication; otherwise, an effective drug in a certain regimen for one disease may be ineffective or even carry risks when used for another disorder.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel.
| | | | | |
Collapse
|
35
|
Kipnis J, Derecki NC, Yang C, Scrable H. Immunity and cognition: what do age-related dementia, HIV-dementia and 'chemo-brain' have in common? Trends Immunol 2008; 29:455-63. [PMID: 18789764 DOI: 10.1016/j.it.2008.07.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Revised: 07/01/2008] [Accepted: 07/03/2008] [Indexed: 11/24/2022]
Abstract
Until recently, dogma dictated that the immune system and the central nervous system (CNS) live mostly separate, parallel lives, and any interactions between the two were assumed to be limited to extreme cases of pathological insult. It was only a decade ago that T cells in the injured brain were shown to play a protective rather than a destructive role. In this article, we explore the role of the immune system in the healthy brain, focusing on the key function that T lymphocytes have in the regulation of cognition. We discuss candidate mechanisms underlying T cell-mediated control of cognitive function in human cognitive diseases associated with immune decline, such as age- and HIV-related dementias, 'chemo-brain' and others.
Collapse
Affiliation(s)
- Jonathan Kipnis
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | |
Collapse
|
36
|
Rolls A, Shechter R, London A, Segev Y, Jacob-Hirsch J, Amariglio N, Rechavi G, Schwartz M. Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation. PLoS Med 2008; 5:e171. [PMID: 18715114 PMCID: PMC2517615 DOI: 10.1371/journal.pmed.0050171] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 07/07/2008] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chondroitin sulfate proteoglycan (CSPG) is a major component of the glial scar. It is considered to be a major obstacle for central nervous system (CNS) recovery after injury, especially in light of its well-known activity in limiting axonal growth. Therefore, its degradation has become a key therapeutic goal in the field of CNS regeneration. Yet, the abundant de novo synthesis of CSPG in response to CNS injury is puzzling. This apparent dichotomy led us to hypothesize that CSPG plays a beneficial role in the repair process, which might have been previously overlooked because of nonoptimal regulation of its levels. This hypothesis is tested in the present study. METHODS AND FINDINGS We inflicted spinal cord injury in adult mice and examined the effects of CSPG on the recovery process. We used xyloside to inhibit CSPG formation at different time points after the injury and analyzed the phenotype acquired by the microglia/macrophages in the lesion site. To distinguish between the resident microglia and infiltrating monocytes, we used chimeric mice whose bone marrow-derived myeloid cells expressed GFP. We found that CSPG plays a key role during the acute recovery stage after spinal cord injury in mice. Inhibition of CSPG synthesis immediately after injury impaired functional motor recovery and increased tissue loss. Using the chimeric mice we found that the immediate inhibition of CSPG production caused a dramatic effect on the spatial organization of the infiltrating myeloid cells around the lesion site, decreased insulin-like growth factor 1 (IGF-1) production by microglia/macrophages, and increased tumor necrosis factor alpha (TNF-alpha) levels. In contrast, delayed inhibition, allowing CSPG synthesis during the first 2 d following injury, with subsequent inhibition, improved recovery. Using in vitro studies, we showed that CSPG directly activated microglia/macrophages via the CD44 receptor and modulated neurotrophic factor secretion by these cells. CONCLUSIONS Our results show that CSPG plays a pivotal role in the repair of injured spinal cord and in the recovery of motor function during the acute phase after the injury; CSPG spatially and temporally controls activity of infiltrating blood-borne monocytes and resident microglia. The distinction made in this study between the beneficial role of CSPG during the acute stage and its deleterious effect at later stages emphasizes the need to retain the endogenous potential of this molecule in repair by controlling its levels at different stages of post-injury repair.
Collapse
Affiliation(s)
- Asya Rolls
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ravid Shechter
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | - Anat London
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | - Yifat Segev
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | - Jasmin Jacob-Hirsch
- Cancer Research Center, Sheba Medical Center and Sackler School of Medicine Tel-Aviv University, Ramat Aviv, Israel
| | - Ninette Amariglio
- Cancer Research Center, Sheba Medical Center and Sackler School of Medicine Tel-Aviv University, Ramat Aviv, Israel
| | - Gidon Rechavi
- Cancer Research Center, Sheba Medical Center and Sackler School of Medicine Tel-Aviv University, Ramat Aviv, Israel
| | - Michal Schwartz
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
37
|
Koehler NKU, Roebbert M, Dehghani K, Ballmaier M, Claus P, von Hoersten S, Shing M, Odin P, Strehlau J, Heidenreich F. Up-regulation of platelet-derived growth factor by peripheral-blood leukocytes during experimental allergic encephalomyelitis. J Neurosci Res 2008; 86:392-402. [PMID: 17893914 DOI: 10.1002/jnr.21497] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In multiple sclerosis (MS) and its animal model, experimental allergic encephalomyelitis (EAE), clinical disease is associated with infiltration of the central nervous system (CNS) by immune cells. Subsequent remission with remyelination has been linked to an increased occurrence of oligodendrocyte progenitor (O2A) cells. Platelet-derived growth factor (PDGF) and fibroblast growth factor-2 (FGF-2) are key growth factors for O2A cells, yet little is known about their relevance in EAE and MS. We analyzed the expression of PDGF, FGF-2, and their receptors by peripheral-blood leukocytes (PBLs) and lymphocyte subsets during MBP-induced EAE. Strong up-regulation of PDGF, but not FGF-2, was observed in PBLs, with the highest expression after the disease maximum. T, NK, and NKT cells expressed PDGF, which is a novel observation because thus far only monocytes/macrophages have been reported to express PDGF. These results extend the idea that growth factors may contribute to improved CNS tissue repair, including PDGF, which is secreted by lesion-homing immune cells. The production of PDGF by lymphocytes may have potential therapeutic value when activating or modulating T-cell responses in demyelinating diseases.
Collapse
|
38
|
Martiñon S, García E, Flores N, Gonzalez I, Ortega T, Buenrostro M, Reyes R, Fernandez-Presas AM, Guizar-Sahagún G, Correa D, Ibarra A. Vaccination with a neural-derived peptide plus administration of glutathione improves the performance of paraplegic rats. Eur J Neurosci 2007; 26:403-12. [PMID: 17623024 DOI: 10.1111/j.1460-9568.2007.05650.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
After damage to the central nervous system (CNS) the body is protected by an adaptive immune response which is directed against myelin-associated proteins. Active immunization with nonpathogenic derivatives of CNS-associated peptides (DCAP) reduces the degeneration of neurons and promotes motor recovery after spinal cord injury (SCI) in rats. In order to improve even more the neurological outcome obtained with this therapy, either a combination of DCAP immunization plus glutathione monoethyl ester (GSHE) or a double DCAP immunization were performed. GSHE is a cell-permeant derivative of glutathione, a potent antioxidant agent that significantly inhibits lipid peroxidation after SCI. After a contusive or compressive SCI, the combination of GSHE + DCAP immunization, induced better motor recovery, a higher number of myelinated axons and better rubrospinal neuron survival than immunization alone. On the other hand, double-DCAP immunization counteracted the protective effect of DCAP therapy. Motor recovery and neuronal survival of double-immunized rats were similar to those observed in control animals (PBS-treated). Further studies revealed that double immunization was not encephalitogenic but inhibited the proliferative response of T-cells specific to the DCAP-immunized peptide. This clonal dysfunction was probably secondary to anergy. GSHE improves the protective effect induced by DCAP immunization while double immunization, reverts it.
Collapse
Affiliation(s)
- S Martiñon
- Unidad de Investigación Médica en Enfermedades Neurológicas, HE, CMN Siglo XXI, IMSS, Avenida Cuauhtemoc no. 330, Col. Doctores, C.P. 06720, México D.F., México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Denkinger CM, Denkinger MD, Forsthuber TG. Pertussis toxin-induced cytokine differentiation and clonal expansion of T cells is mediated predominantly via costimulation. Cell Immunol 2007; 246:46-54. [PMID: 17601518 PMCID: PMC2724065 DOI: 10.1016/j.cellimm.2007.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 05/10/2007] [Accepted: 05/11/2007] [Indexed: 02/03/2023]
Abstract
Pertussis toxin (PTX) has potent immunologic adjuvant activity in vivo and concomitantly enhances both T helper type (Th1) and Th2 cytokine responses. The PTX-induced enhancement of Th1 and Th2 immunity is mediated via the activation of antigen presenting cells (APCs), but the underlying mechanism is not known. Here we asked whether the adjuvant activity of PTX on T cell immunity was mediated by cytokines and/or costimulatory signals. The results show that in vivo blockade of CD28-CD80/86 costimulation essentially abrogated PTX-mediated enhancement of antigen-specific Th1 and Th2 responses. Blockade of CD40L-CD40 interactions was less efficient in inhibiting PTX-mediated enhancement of Th1 and Th2 responses. In contrast, the adjuvant activity of PTX was not mediated via cytokines, because neither Th1 nor Th2 responses were substantially impaired in mice deficient for IL-12, IFN-gamma, IL-4, IL-5, or IL-6. Collectively, the data suggest that PTX mediates its adjuvant effects on T cell cytokine differentiation and clonal expansion via the modulation of costimulatory molecules on APCs. Understanding the costimulatory pathways targeted by PTX could lead to the design of novel adjuvants that selectively induce Th1 or Th2 immunity.
Collapse
Affiliation(s)
- Claudia M. Denkinger
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Michael D. Denkinger
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH
- Bethesda Geriatrische Klinik Ulm, Germany
| | - Thomas G. Forsthuber
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
40
|
Ankeny DP, Popovich PG. Central nervous system and non-central nervous system antigen vaccines exacerbate neuropathology caused by nerve injury. Eur J Neurosci 2007; 25:2053-64. [PMID: 17439492 DOI: 10.1111/j.1460-9568.2007.05458.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previously, we showed that autoimmune (central nervous system myelin-reactive) T cells exacerbate tissue damage and impair neurological recovery after spinal cord injury. Conversely, independent studies have shown T cell-mediated neuroprotection after spinal cord injury or facial nerve axotomy (FNAx). The antigen specificity of the neuroprotective T cells has not been investigated after FNAx. Here, we compared the neuroprotective capacity of autoimmune and non-autoimmune lymphocytes after FNAx. Prior to axotomy, C57BL/6 mice were immunized with myelin basic protein, myelin oligodendrocyte glycoprotein (MOG) or ovalbumin (a non-self antigen) emulsified in complete Freund's adjuvant (CFA). FNAx mice receiving injections of phosphate-buffered saline (PBS) only (unimmunized) or PBS/CFA emulsions served as controls. At 4 weeks after axotomy, bilateral facial motor neuron counts were obtained throughout the facial motor nucleus using unbiased stereology (optical fractionator). The data show that neuroantigen immunizations and 'generic' lymphocyte activation (e.g. PBS/CFA or ovalbumin/CFA immunizations) exacerbated neuron loss above that caused by FNAx alone. We also found that nerve injury potentiated the effector potential of autoimmune lymphocytes. Indeed, prominent forelimb and hindlimb motor deficits were accompanied by disseminated neuroinflammation and demyelination in FNAx mice receiving subencephalitogenic immunization with MOG. FNAx or neuroantigen (MOG or myelin basic protein) immunization alone did not cause these pathological changes. Thus, irrespective of the antigens used to trigger an immune response, neuropathology was enhanced when the immune system was primed in parallel with nerve injury. These data have important implications for therapeutic vaccination in clinical neurotrauma and neurodegeneration.
Collapse
Affiliation(s)
- Daniel P Ankeny
- Department of Molecular Virology, Immunology & Medical Genetics, The Center for Brain and Spinal Cord Repair and The Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA
| | | |
Collapse
|
41
|
Hofstetter HH, Kovalovsky A, Shive CL, Lehmann PV, Forsthuber TG. Neonatal induction of myelin-specific Th1/Th17 immunity does not result in experimental autoimmune encephalomyelitis and can protect against the disease in adulthood. J Neuroimmunol 2007; 187:20-30. [PMID: 17482277 PMCID: PMC3204791 DOI: 10.1016/j.jneuroim.2007.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 03/02/2007] [Accepted: 04/02/2007] [Indexed: 10/23/2022]
Abstract
The neonatal immune system is believed to be biased towards T helper type 2 (Th2) immunity, but under certain conditions neonates can also develop Th1 immune responses. Neonatal Th2 immunity to myelin antigens is not pathogenic and can prevent induction of experimental autoimmune encephalomyelitis (EAE) in adulthood, but the consequences of neonatally induced Th1 immunity to self-antigens have remained unresolved. Here, we show that neonatal injection of mice with myelin antigens emulsified in complete Freund's adjuvant (CFA) induced vigorous production of IFN-gamma and IL-17, but not IL-5, consistent with myelin-specific Th1/Th17 immunity. Importantly, the myelin-specific Th1/Th17 cells persisted in the mice until adulthood without causing symptoms of EAE. Intraperitoneal, but not subcutaneous injection of neonates with myelin antigens protected against induction of EAE as adults. Intraperitoneally injected neonates showed a substantial decrease of the number and avidity of myelin-reactive Th17 cells, suggesting a decrease in IL-17 producing precursor cells as the mechanism of protection from EAE upon re-injection with myelin antigens as adults. The results could provide a rationale for the presence of autoreactive T cells found in healthy human individuals without autoimmune disease.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Age Factors
- Animals
- Animals, Newborn
- CD4 Antigens/metabolism
- Central Nervous System/metabolism
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Enzyme-Linked Immunosorbent Assay
- Guinea Pigs
- Image Processing, Computer-Assisted
- Infusions, Parenteral/methods
- Mice
- Mice, Inbred Strains
- Myelin Proteolipid Protein/immunology
- Myelin Proteolipid Protein/therapeutic use
- Peptide Fragments/immunology
- Peptide Fragments/therapeutic use
- T-Lymphocytes/immunology
- Th1 Cells/immunology
- Time Factors
Collapse
Affiliation(s)
- Harald H. Hofstetter
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106
| | - Andra Kovalovsky
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106
| | - Carey L. Shive
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106
| | - Paul V. Lehmann
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106
| | - Thomas G. Forsthuber
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106
- Dept. Biology, University of Texas at San Antonio, San Antonio, TX, 78249
- Corresponding author. Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249 United States., (T.G. Forsthuber)
| |
Collapse
|
42
|
Yu P, Huang L, Zou J, Zhu H, Wang X, Yu Z, Xu XM, Lu PH. DNA vaccine against NgR promotes functional recovery after spinal cord injury in adult rats. Brain Res 2007; 1147:66-76. [PMID: 17362886 DOI: 10.1016/j.brainres.2007.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 01/30/2007] [Accepted: 02/06/2007] [Indexed: 10/23/2022]
Abstract
NgR is a common receptor for three myelin-associated inhibitors and mediates their inhibitory activities on neurite outgrowth. In the present study, we investigated whether a DNA vaccine targeting NgR could play a beneficial role in improving recovery from spinal cord injury (SCI). We demonstrated that a DNA vaccine against NgR was successfully constructed and expressed efficiently in vitro and in vivo. After immunization with anti-NgR DNA vaccine, a low level of antibody response and a T cell-mediated immune response were induced in the vaccinated rats. And the antisera taken from the anti-NgR DNA vaccinated rats could partly reverse the inhibition of MAG on neurite outgrowth. When the rats were subjected to a contusive SCI, the vaccinated rats showed much better functional recovery than the controls. In those vaccinated rats that induced a T cell response and generated antibodies against NgR, functional improvements were even better. Histological assessments by three-dimensional reconstruction further demonstrated that the total lesion volume in the vaccinated rats was reduced by 30.8% compared to the controls. These results collectively suggest that DNA vaccine against NgR can significantly improve functional recovery in rats that received contusive SCI and that the vaccination approach may provide a promising strategy for promoting SCI repair.
Collapse
Affiliation(s)
- Panpan Yu
- Department of Neurobiology, School of Medicine, Shanghai Jiaotong University, 280 South Chong Qing Road, Shanghai 200025, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Central nervous system (CNS) immune privilege is an experimentally defined phenomenon. Tissues that are rapidly rejected by the immune system when grafted in sites, such as the skin, show prolonged survival when grafted into the CNS. Initially, CNS immune privilege was construed as CNS isolation from the immune system by the blood-brain barrier (BBB), the lack of draining lymphatics, and the apparent immunoincompetence of microglia, the resident CNS macrophage. CNS autoimmunity and neurodegeneration were presumed automatic consequences of immune cell encounter with CNS antigens. Recent data have dramatically altered this viewpoint by revealing that the CNS is neither isolated nor passive in its interactions with the immune system. Peripheral immune cells can cross the intact BBB, CNS neurons and glia actively regulate macrophage and lymphocyte responses, and microglia are immunocompetent but differ from other macrophage/dendritic cells in their ability to direct neuroprotective lymphocyte responses. This newer view of CNS immune privilege is opening the door for therapies designed to harness autoreactive lymphocyte responses and also implies (i) that CNS autoimmune diseases (i.e. multiple sclerosis) may result as much from neuronal and/or glial dysfunction as from immune system dysfunctions and (ii) that the severe neuronal and glial dysfunction associated with neurodegenerative disorders (i.e. Alzheimer's disease) likely alters CNS-specific regulation of lymphocyte responses affecting the utility of immune-based therapies (i.e. vaccines).
Collapse
Affiliation(s)
- Monica J Carson
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA.
| | | | | | | | | |
Collapse
|
44
|
Mutlu L, Brandt C, Kwidzinski E, Sawitzki B, Gimsa U, Mahlo J, Aktas O, Nitsch R, van Zwam M, Laman JD, Bechmann I. Tolerogenic effect of fiber tract injury: reduced EAE severity following entorhinal cortex lesion. Exp Brain Res 2006; 178:542-53. [PMID: 17091291 DOI: 10.1007/s00221-006-0758-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 10/10/2006] [Indexed: 12/25/2022]
Abstract
Despite transient, myelin-directed adaptive immune responses in regions of fiber tract degeneration, none of the current models of fiber tract injuries evokes disseminated demyelination, implying effective mechanisms maintaining or re-establishing immune tolerance. In fact, we have recently detected CD95L upregulation accompanied by apoptosis of leukocytes in zones of axonal degeneration induced by entorhinal cortex lesion (ECL), a model of layer-specific axonal degeneration. Moreover, infiltrating monocytes readily transformed into ramified microglia exhibiting a phenotype of immature (CD86+/CD80-) antigen-presenting cells. We now report the appearance of the axonal antigen neurofilament-light along with increased T cell apoptosis and enhanced expression of the pro-apoptotic gene Bad in cervical lymph nodes after ECL. In order to test the functional significance of such local and systemic depletory/regulatory mechanisms on subsequent immunity to central nervous system antigens, experimental autoimmune encephalomyelitis was induced by proteolipid protein immunization 30 days after ECL. In three independent experiments, we found significantly diminished disease scores and infiltrates in lesioned compared to sham-operated SJL mice. This is consistent with a previous meta-statistical analysis (Goodin et al. in Neurology 52:1737-1745, 1999) rejecting the O-hypothesis that brain trauma causes or exacerbates multiple sclerosis. Conversely, brain injuries may involve long-term tolerogenic effects towards brain antigens.
Collapse
Affiliation(s)
- Leman Mutlu
- Institute of Cell Biology and Neurobiology, Charité, 10098, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Deboy CA, Xin J, Byram SC, Serpe CJ, Sanders VM, Jones KJ. Immune-mediated neuroprotection of axotomized mouse facial motoneurons is dependent on the IL-4/STAT6 signaling pathway in CD4+ T cells. Exp Neurol 2006; 201:212-24. [PMID: 16806176 DOI: 10.1016/j.expneurol.2006.04.028] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2005] [Revised: 03/13/2006] [Accepted: 04/05/2006] [Indexed: 12/12/2022]
Abstract
The CD4(+) T lymphocyte has recently been found to promote facial motoneuron (FMN) survival after nerve injury. Signal Transducer and Activator of Transcription (STAT)4 and STAT6 are key proteins involved in the CD4(+) T cell differentiation pathways leading to T helper type (Th)1 and Th2 cell development, respectively. To determine which CD4(+) T cell subset mediates FMN survival, the facial nerve axotomy paradigm was applied to STAT4-deficient (-/-) and STAT6-/- mice. A significant decrease in FMN survival 4 weeks after axotomy was observed in STAT6-/- mice compared to wild-type (WT) or STAT4-/- mice. Reconstituting STAT6-/- mice with CD4(+) T cells obtained from WT mice promoted WT levels of FMN survival after injury. Furthermore, rescue of FMN from axotomy-induced cell death in recombination activating gene (RAG)-2-/- mice (lacking T and B cells) could be achieved only by reconstitution with CD4(+) T cells expressing functional STAT6 protein. To determine if either the Th1 cytokine, interferon-gamma (IFN-gamma) or the Th2 cytokine IL-4 is involved in mediating FMN survival, facial nerve axotomy was applied to IFN-gamma-/- and IL-4-/- mice. A significant decrease in FMN survival after axotomy occurred in IL-4-/- but not in IFN-gamma-/- mice compared to WT mice, indicating that IL-4 but not IFN-gamma is important for FMN survival after nerve injury. In WT mice, intracellular IFN-gamma vs. IL-4 expression was examined in CD4(+) T cells from draining cervical lymph nodes 14 days after axotomy, and substantial increase in the production of both CD4(+) effector T cell subsets was found. Collectively, these data suggest that STAT6-mediated CD4(+) T cell differentiation into the Th2 subset is necessary for FMN survival. A hypothesis relevant to motoneuron disease progression is presented.
Collapse
Affiliation(s)
- Cynthia A Deboy
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago, 2160 S. 1st Avenue, Maywood, IL 60153, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Ziv Y, Avidan H, Pluchino S, Martino G, Schwartz M. Synergy between immune cells and adult neural stem/progenitor cells promotes functional recovery from spinal cord injury. Proc Natl Acad Sci U S A 2006; 103:13174-9. [PMID: 16938843 PMCID: PMC1559772 DOI: 10.1073/pnas.0603747103] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The well regulated activities of microglia and T cells specific to central nervous system (CNS) antigens can contribute to the protection of CNS neural cells and their renewal from adult neural stem/progenitor cells (aNPCs). Here we report that T cell-based vaccination of mice with a myelin-derived peptide, when combined with transplantation of aNPCs into the cerebrospinal fluid (CSF), synergistically promoted functional recovery after spinal cord injury. The synergistic effect was correlated with modulation of the nature and intensity of the local T cell and microglial response, expression of brain-derived neurotrophic factor and noggin protein, and appearance of newly formed neurons from endogenous precursor-cell pools. These results substantiate the contention that the local immune response plays a crucial role in recruitment of aNPCs to the lesion site, and suggest that similar immunological manipulations might also serve as a therapeutic means for controlled migration of stem/progenitor cells to other acutely injured CNS sites.
Collapse
Affiliation(s)
- Yaniv Ziv
- *Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel; and
| | - Hila Avidan
- *Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel; and
| | - Stefano Pluchino
- Neuroimmunology Unit (DIBIT), Department of Neurology and Neurophysiology, San Raffaele Hospital, Via Olgettina 58, 20132 Milano, Italy
| | - Gianvito Martino
- Neuroimmunology Unit (DIBIT), Department of Neurology and Neurophysiology, San Raffaele Hospital, Via Olgettina 58, 20132 Milano, Italy
| | - Michal Schwartz
- *Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
47
|
DeBoy CA, Byram SC, Serpe CJ, Wisuri D, Sanders VM, Jones KJ. CD4+CD25+ regulatory T cells and CD1-restricted NKT cells do not mediate facial motoneuron survival after axotomy. J Neuroimmunol 2006; 176:34-8. [PMID: 16766044 DOI: 10.1016/j.jneuroim.2006.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2006] [Revised: 04/05/2006] [Accepted: 04/05/2006] [Indexed: 01/14/2023]
Abstract
CD4+ T cells rescue facial motoneurons (FMN) from axotomy-induced cell death. The objective of this study is to determine if the CD4+ T regulatory subsets, CD4+CD25+ T or CD1d-restricted NKT cells are critical for FMN survival after facial nerve axotomy. Surviving FMN within facial motor nuclei from axotomized and control sides 4 weeks after axotomy were counted to determine percent FMN survival. Data generated by applying this paradigm to recombination activating gene-2-deficient mice reconstituted with CD4+ T cells depleted of CD4+CD25+ T cells and to CD1-/- mice, deficient in CD1d-restricted NKT cells, suggest that neither regulatory CD4+ T subset is critical for FMN survival.
Collapse
Affiliation(s)
- Cynthia A DeBoy
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago, Maywood, IL 60153, and Research and Development Service, Hines VA Hospital 60141, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Bechmann I. Failed central nervous system regeneration: a downside of immune privilege? Neuromolecular Med 2006; 7:217-28. [PMID: 16247182 DOI: 10.1385/nmm:7:3:217] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 07/07/2005] [Indexed: 12/25/2022]
Abstract
Immunity is required to eliminate dangerous or degenerated material and to support regeneration, but also causes significant parenchymal damage. In the eye and the brain, in which cornea and lens poorly regenerate and neurons are hardly replaceable, early transplantation experiments demonstrated remarkable tolerance to various grafts. This "immunologically privileged status" (Billingham and Boswell, 1953) may reflect evolutionary pressure to downmodulate certain actions of immune cells within particularly vulnerable tissues. As an example, tolerating certain "neurotrophic" viruses may often be a more successful strategy for survival than the elimination of all infected neurons. While several constitutive and inducible signals maintaining or re-establishing immune tolerance within the brain have been identified, it has also become evident that the resulting anti-inflammatory environment limits certain beneficial effects of neuroinflammation such as neurotrophin secretion or glutamate buffering by T-cells and the clearance of growth-inhibiting myelin or amyloid. Following spinal cord injury, the costs and benefits of neuroinflammation seem to come close because enhancing as well as suppressing innate or adaptive immunity caused amelioration and aggravation of functional regeneration in similar experiments. Evaluating such balances has also begun in (animal models of) Alzheimer's disease, central nervous system trauma, and stroke, and the appreciation of the beneficial side of neuroinflammation has caused a rethinking of the ill-defined use of immune suppressants. As dual roles for individual molecules have been recognized (Merrill and Benveniste, 1996), we are uncovering an already fine-tuned system, but the challenge remains to further support beneficial immune cascades without causing additional damage, and vice versa.
Collapse
Affiliation(s)
- Ingo Bechmann
- Center for Anatomy, Institute of Cell Biology and Neurobiology, Department of Experimental Neuroimmunology, Charité-Universitätsmedizin Berlin, Schumannstr, 20/21 10098 Berlin, Germany.
| |
Collapse
|
49
|
Ling C, Sandor M, Suresh M, Fabry Z. Traumatic injury and the presence of antigen differentially contribute to T-cell recruitment in the CNS. J Neurosci 2006; 26:731-41. [PMID: 16421293 PMCID: PMC6675378 DOI: 10.1523/jneurosci.3502-05.2006] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
T-cell recruitment into the brain is critical in inflammatory and autoimmune diseases of the CNS. We use intracerebral antigen microinjection and tetramer technology to track antigen-specific CD8+ T-cells in the CNS and to clarify the contribution of antigen deposition or traumatic injury to the accumulation of T-cells in the brain. We demonstrate that, after intracerebral microinjection of ovalbumin, ovalbumin-specific CD8+ T-cells expand systemically and then migrate into the brain where they complete additional proliferation cycles. T-cells in the brain are activated and respond to in vitro secondary antigen challenge. CD8+ T-cells accumulate and persist in sites of antigen in the brain without replenishment from the periphery. Persistent survival of CD8+ T-cells at sites of cognate antigen is significantly reduced by blocking CD154 molecules. A small traumatic injury itself does not lead to recruitment of CD8+ T-cells into the brain but attracts activated antigen-specific CD8+ T-cells from cognate antigen injection sites. This process is presumably antigen independent and cannot be inhibited by blocking CD154 molecules. These data show that activated antigen-specific CD8+ T-cells accumulate in the CNS at both cognate antigen-containing and traumatic injury sites after intracerebral antigen delivery. The accumulation of activated antigen-specific T-cells at traumatic injury sites, in addition to antigen-containing areas, could amplify local inflammatory processes in the CNS. Combination therapies in neuroinflammatory diseases to block both of these processes should be considered.
Collapse
Affiliation(s)
- Changying Ling
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison Medical School, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|
50
|
Ponomarenko NA, Durova OM, Vorobiev II, Belogurov AA, Kurkova IN, Petrenko AG, Telegin GB, Suchkov SV, Kiselev SL, Lagarkova MA, Govorun VM, Serebryakova MV, Avalle B, Tornatore P, Karavanov A, Morse HC, Thomas D, Friboulet A, Gabibov AG. Autoantibodies to myelin basic protein catalyze site-specific degradation of their antigen. Proc Natl Acad Sci U S A 2006; 103:281-6. [PMID: 16387849 PMCID: PMC1324791 DOI: 10.1073/pnas.0509849103] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Autoantibody-mediated tissue destruction is among the main features of organ-specific autoimmunity. This report describes "an antibody enzyme" (abzyme) contribution to the site-specific degradation of a neural antigen. We detected proteolytic activity toward myelin basic protein (MBP) in the fraction of antibodies purified from the sera of humans with multiple sclerosis (MS) and mice with induced experimental allergic encephalomyelitis. Chromatography and zymography data demonstrated that the proteolytic activity of this preparation was exclusively associated with the antibodies. No activity was found in the IgG fraction of healthy donors. The human and murine abzymes efficiently cleaved MBP but not other protein substrates tested. The sites of MBP cleavage determined by mass spectrometry were localized within immunodominant regions of MBP. The abzymes could also cleave recombinant substrates containing encephalytogenic MBP(85-101) peptide. An established MS therapeutic Copaxone appeared to be a specific abzyme inhibitor. Thus, the discovered epitope-specific antibody-mediated degradation of MBP suggests a mechanistic explanation of the slow development of neurodegeneration associated with MS.
Collapse
Affiliation(s)
- Natalia A Ponomarenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya Street, Moscow 117997, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|