1
|
Poletti S, Zanardi R, Mandelli A, Aggio V, Finardi A, Lorenzi C, Borsellino G, Carminati M, Manfredi E, Tomasi E, Spadini S, Colombo C, Drexhage HA, Furlan R, Benedetti F. Low-dose interleukin 2 antidepressant potentiation in unipolar and bipolar depression: Safety, efficacy, and immunological biomarkers. Brain Behav Immun 2024; 118:52-68. [PMID: 38367846 DOI: 10.1016/j.bbi.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024] Open
Abstract
Immune-inflammatory mechanisms are promising targets for antidepressant pharmacology. Immune cell abnormalities have been reported in mood disorders showing a partial T cell defect. Following this line of reasoning we defined an antidepressant potentiation treatment with add-on low-dose interleukin 2 (IL-2). IL-2 is a T-cell growth factor which has proven anti-inflammatory efficacy in autoimmune conditions, increasing thymic production of naïve CD4 + T cells, and possibly correcting the partial T cell defect observed in mood disorders. We performed a single-center, randomised, double-blind, placebo-controlled phase II trial evaluating the safety, clinical efficacy and biological responses of low-dose IL-2 in depressed patients with major depressive (MDD) or bipolar disorder (BD). 36 consecutively recruited inpatients at the Mood Disorder Unit were randomised in a 2:1 ratio to receive either aldesleukin (12 MDD and 12 BD) or placebo (6 MDD and 6 BD). Active treatment significantly potentiated antidepressant response to ongoing SSRI/SNRI treatment in both diagnostic groups, and expanded the population of T regulatory, T helper 2, and percentage of Naive CD4+/CD8 + immune cells. Changes in cell frequences were rapidly induced in the first five days of treatment, and predicted the later improvement of depression severity. No serious adverse effect was observed. This is the first randomised control trial (RCT) evidence supporting the hypothesis that treatment to strengthen the T cell system could be a successful way to correct the immuno-inflammatory abnormalities associated with mood disorders, and potentiate antidepressant response.
Collapse
Affiliation(s)
- Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy.
| | - Raffaella Zanardi
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Alessandra Mandelli
- Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Veronica Aggio
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | | | - Matteo Carminati
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Elena Manfredi
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Enrico Tomasi
- Hospital Pharmacy, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milano, Italy; Mood Disorder Unit, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Hemmo A Drexhage
- Coordinator EU consortium MoodStratification, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roberto Furlan
- Vita-Salute San Raffaele University, Milano, Italy; Clinical Neuroimmunology, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
2
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
3
|
Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol 2019; 320:112957. [PMID: 31108085 DOI: 10.1016/j.expneurol.2019.112957] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of both acute and long-term morbidity in the pediatric population, leading to a substantial, long-term socioeconomic burden. Despite the increase in the amount of pre-clinical and clinical research, treatment options for TBI rely heavily on supportive care with very limited targeted interventions that improve the acute and chronic sequelae of TBI. Other than injury prevention, not much can be done to limit the primary injury, which consists of tissue damage and cellular destruction. Secondary injury is the result of the ongoing complex inflammatory pathways that further exacerbate tissue damage, resulting in the devastating chronic outcomes of TBI. On the other hand, some level of inflammation is essential for neuronal regeneration and tissue repair. In this review article we discuss the various stages of the neuroimmune response in the immature, pediatric brain in the context of normal maturation and development of the immune system. The developing brain has unique features that distinguish it from the adult brain, and the immune system plays an integral role in CNS development. Those features could potentially make the developing brain more susceptible to worse outcomes, both acutely and in the long-term. The neuroinflammatory reaction which is triggered by TBI can be described as a highly intricate interaction between the cells of the innate and the adaptive immune systems. The innate immune system is triggered by non-specific danger signals that are released from damaged cells and tissues, which in turn leads to neutrophil infiltration, activation of microglia and astrocytes, complement release, as well as histamine release by mast cells. The adaptive immune response is subsequently activated leading to the more chronic effects of neuroinflammation. We will also discuss current attempts at modulating the TBI-induced neuroinflammatory response. A better understanding of the role of the immune system in normal brain development and how immune function changes with age is crucial for designing therapies to appropriately target the immune responses following TBI in order to enhance repair and plasticity.
Collapse
Affiliation(s)
- Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Young Chun
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States of America.
| |
Collapse
|
4
|
Kertser A, Baruch K, Deczkowska A, Weiner A, Croese T, Kenigsbuch M, Cooper I, Tsoory M, Ben-Hamo S, Amit I, Schwartz M. Corticosteroid signaling at the brain-immune interface impedes coping with severe psychological stress. SCIENCE ADVANCES 2019; 5:eaav4111. [PMID: 31149632 PMCID: PMC6541460 DOI: 10.1126/sciadv.aav4111] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/22/2019] [Indexed: 05/22/2023]
Abstract
The immune system supports brain plasticity and homeostasis, yet it is prone to changes following psychological stress. Thus, it remains unclear whether and how stress-induced immune alterations contribute to the development of mental pathologies. Here, we show that following severe stress in mice, leukocyte trafficking through the choroid plexus (CP), a compartment that mediates physiological immune-brain communication, is impaired. Blocking glucocorticoid receptor signaling, either systemically or locally through its genetic knockdown at the CP, facilitated the recruitment of Gata3- and Foxp3-expressing T cells to the brain and attenuated post-traumatic behavioral deficits. These findings functionally link post-traumatic stress behavior with elevated stress-related corticosteroid signaling at the brain-immune interface and suggest a novel therapeutic target to attenuate the consequences of severe psychological stress.
Collapse
Affiliation(s)
- A. Kertser
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - K. Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - A. Deczkowska
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - A. Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - T. Croese
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - M. Kenigsbuch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - I. Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
| | - M. Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - S. Ben-Hamo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - I. Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - M. Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- Corresponding author.
| |
Collapse
|
5
|
Sun Y, Jing Y, Huang M, Ma J, Peng X, Wang J, Li G, Cheng X. The PD-1/PD-Ls pathway is up-regulated during the suppression of experimental autoimmune encephalomyelitis treated by Astragalus polysaccharides. J Neuroimmunol 2019; 332:78-90. [PMID: 30981049 DOI: 10.1016/j.jneuroim.2019.03.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of CNS. Astragalus polysaccharides (APS), the main active extract from astragalus membranaceus which is a kind of traditional Chinese medicinal herb, is associated with a variety of immunomodulatory activities. We have evaluated the therapeutic effects of APS in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). It was found that APS could effectively alleviate EAE through inhibiting MOG35-55-specific T cell proliferation and reducing the expression of proinflammatory cytokines, which is mediated by up-regulating the expression of PD-1/PD-Ls signaling pathway. Our results demonstrated that EAE could be suppressed significantly by APS administration. It indicated that APS might be a potential of developing innovative drug for the therapy of MS.
Collapse
Affiliation(s)
- Yu Sun
- Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yuanya Jing
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Mengwen Huang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jinyun Ma
- Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaoyan Peng
- Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jinying Wang
- Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Guoling Li
- Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaodong Cheng
- Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
6
|
Abstract
Spinal cord injury (SCI) is a common medical condition with a poor prognosis for recovery and catastrophic effects on a patient's quality of life. Available treatments for SCI are limited, and the evidence suggesting their harmful side effects is more consistent than any suggestion of clinical benefit. Developing novel safe and effective therapeutic options for SCI is crucial. Granulocyte colony-stimulating factor (G-CSF) is a hematopoietic cytokine with known multifaceted effects on the central nervous system. Herein, we review the accumulating preclinical evidence for the beneficial effects of G-CSF on functional and structural outcomes after SCI. Meanwhile we present and discuss multiple mechanisms for G-CSF's neuroprotective and neuroregenerative actions through the results of these studies. In addition, we present the available clinical evidence indicating the efficacy and safety of G-CSF administration for the treatment of acute and chronic traumatic SCI, compression myelopathy, and SCI-associated neuropathic pain. Our review indicates that although the quality of clinical evidence regarding the use of G-CSF in SCI is inadequate, the encouraging available preclinical and clinical data warrant its further clinical development, and bring new hope to the longstanding challenge that is treatment of SCI.
Collapse
|
7
|
Braun M, Vaibhav K, Saad N, Fatima S, Brann DW, Vender JR, Wang LP, Hoda MN, Baban B, Dhandapani KM. Activation of Myeloid TLR4 Mediates T Lymphocyte Polarization after Traumatic Brain Injury. THE JOURNAL OF IMMUNOLOGY 2017; 198:3615-3626. [PMID: 28341672 DOI: 10.4049/jimmunol.1601948] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/21/2017] [Indexed: 12/15/2022]
Abstract
Traumatic brain injury (TBI) is a major public health issue, producing significant patient mortality and poor long-term outcomes. Increasing evidence suggests an important, yet poorly defined, role for the immune system in the development of secondary neurologic injury over the days and weeks following a TBI. In this study, we tested the hypothesis that peripheral macrophage infiltration initiates long-lasting adaptive immune responses after TBI. Using a murine controlled cortical impact model, we used adoptive transfer, transgenic, and bone marrow chimera approaches to show increased infiltration and proinflammatory (classically activated [M1]) polarization of macrophages for up to 3 wk post-TBI. Monocytes purified from the injured brain stimulated the proliferation of naive T lymphocytes, enhanced the polarization of T effector cells (TH1/TH17), and decreased the production of regulatory T cells in an MLR. Similarly, elevated T effector cell polarization within blood and brain tissue was attenuated by myeloid cell depletion after TBI. Functionally, C3H/HeJ (TLR4 mutant) mice reversed M1 macrophage and TH1/TH17 polarization after TBI compared with C3H/OuJ (wild-type) mice. Moreover, brain monocytes isolated from C3H/HeJ mice were less potent stimulators of T lymphocyte proliferation and TH1/TH17 polarization compared with C3H/OuJ monocytes. Taken together, our data implicate TLR4-dependent, M1 macrophage trafficking/polarization into the CNS as a key mechanistic link between acute TBI and long-term, adaptive immune responses.
Collapse
Affiliation(s)
- Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA 30912
| | - Nancy Saad
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA 30912
| | - Sumbul Fatima
- Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Lei P Wang
- Department of Psychiatry, Medical College of Georgia, Augusta University, Augusta, GA 30912; and
| | - Md Nasrul Hoda
- Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912; .,Charlie Norwood VA Medical Center, Augusta, GA 30912
| |
Collapse
|
8
|
Tseng CH, Chen JH, Lin CL, Kao CH. Decreased risk of intracerebral hemorrhage among patients with milder allergic rhinitis. QJM 2016; 109:161-5. [PMID: 26025692 DOI: 10.1093/qjmed/hcv104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND This study investigated whether allergic rhinitis (AR) increases the risk of intracerebral hemorrhage (ICH). METHODS Using Taiwanese insurance outpatient claims data, 52,870 patients with new diagnoses of AR between 2000 and 2010 were identified, and 105,680 age- and sex-frequency matched people without AR were chosen randomly as controls. Risks of ICH were correlated with AR and comorbidities, such as diabetes, hypertension, coronary artery disease, hyperlipidemia, disease and chronic kidney disease, and were estimated by the end of 2011. RESULTS The risk of ICH was lower in the AR cohort than in the comparison cohort, with an adjusted hazard ratio (aHR) of 0.58 (95% confidence interval [CI] = 0.50-0.66), assessed using the multivariable Cox model. Age-specific analysis demonstrated that the aHR for ICH in the AR patients increased with age, with an aHR of 0.53 (95% CI = 0.39-0.71) in the ≤49-year age group, which increased to 0.72 (95% CI = 0.59-0.89) in the elderly age group. The risk of ICH increased with the severity of AR, from an aHR of 0.47 (95% CI = 0.40-0.56) in mild AR patients to 2.55 (95% CI = 1.88-3.47) in severe ones. For patients without comorbidities, the risk of ICH was 0.46 times (95% CI = 0.34-0.63) lower in the AR cohort than in the comparison cohort. CONCLUSION This study showed for the first time that milder AR is correlated with a reduction in the risk of ICH, particularly for elderly patients.
Collapse
Affiliation(s)
- C-H Tseng
- From the Department of Neurology, China Medical University Hospital, School of Medicine, China Medical University College of Medicine
| | - J-H Chen
- School of Medicine, China Medical University College of Medicine, Department of Internal Medicine, China Medical University Hospital
| | - C-L Lin
- School of Medicine, China Medical University College of Medicine, Management Office for Health Data, China Medical University Hospital
| | - C-H Kao
- Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine, China Medical University and Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
9
|
Tseng CH, Muo CH, Lin MC, Kao CH. Association Between Reduced Risk of Intracerebral Hemorrhage and Pelvic Inflammatory Disease. Medicine (Baltimore) 2016; 95:e2726. [PMID: 26844517 PMCID: PMC4748934 DOI: 10.1097/md.0000000000002726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This study examines whether pelvic inflammatory disease (PID) facilitates the development of intracerebral hemorrhage (ICH).By using outpatient claims data from the National Health Insurance Research Database (NHIRD) of Taiwan, we included the data of 25,508 patients who were newly diagnosed with PID between 1999 and 2004, and also from the Taiwan NHIRD, we randomly selected 102,032 women without PID, who were frequency-matched by age and entry-year and with 4 times the number of the PID patients, as the control cohort. We measured ICH risks associated with PID and comorbidities, including hyperlipidemia, diabetes, hypertension, ischemic heart disease, and atrial fibrillation, by the end of 2011.In comparison with the controls, the ICH hazard was less in the PID group with an adjusted hazard ratio (aHR) of 0.67 (95% confidence interval [CI]:0.50-0.90), which was noted by calculation with the Cox proportional regression model. The ICH risk in the PID patients reduced progressively with the advance of age, with aHRs of 0.75 (95% CI:0.41-1.39) and 0.50 (95% CI:0.29-0.88), respectively, in the age <35-year and age ≥50-year groups. ICH risk lowered gradually with the progress of PID severity, from mild PID with an aHR of 0.72 (95% CI:0.53-0.98) to severe PID with that of 0.30 (95% CI:0.10-0.92). PID patients without any comorbidites had lower ICH risk (aHR = 0.63, 95% CI:0.42-0.94) than the controls without any comorbidites did.Our findings revealed that PID is associated with reduced ICH development, especially for older patients.
Collapse
Affiliation(s)
- Chun-Hung Tseng
- From the Department of Neurology, China Medical University Hospital (C-HT); School of Medicine, China Medical University (C-HT, C-HM); Management Office for Health Data, China Medical University Hospital, Taichung (C-HM); Department of Nuclear Medicine, E-Da Hospital and I-Shou University, Kaohsiung (M-GL); Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine (C-HK); and Department of Nuclear Medicine and PET Center, China Medical University Hospital (C-HK), Taichung, Taiwan
| | | | | | | |
Collapse
|
10
|
Zhang Y, Li Q, Zou J, Zuo Z, Yao Z. Continuous vaccinations of 4Aβ1-15 induces specific fluctuation of inflammatory factors accompany with pathologic alterations alleviation in APP/PS1 mice. Hum Vaccin Immunother 2015; 11:2674-81. [PMID: 26295455 DOI: 10.1080/21645515.2015.1034922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The common pathological hallmark of Alzheimer's disease (AD) is β-amyloid plaques deposition. Immunotherapy is a revolutionary pharmacological treatment for AD, aiming at improving plaque clearance while concomitantly decreasing inflammation. Our previous study prepared antigen 4Aβ1-15 and found that it could alleviate pathologic alterations in APP/PS1 transgenic mice. The objective of our study was to research the changing processes induced by immunotherapy, including the inflammatory factor levels and microglial activation that is closely associated with Aβ burdens clearance. APP/PS1 mice were injected with 4Aβ1-15 6 times. Each time, the inflammatory factors in sera were detected, and a specific fluctuation that first increased and then decreased was found, in which there was a turning point after the third injection. It prompted us to further detect the indicators in the brains after the third injection and the sixth injection. The results showed that the therapeutic effects for Aβ burdens and behaviors were continuously improved during the whole immune processes, whereas the inflammatory factor levels and microglial activation experienced similar specific fluctuations. The novel discovery may provide convenient methods for further detection and evaluation of immunotherapy in disease courses.
Collapse
Affiliation(s)
- Yuwei Zhang
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Qingqing Li
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Juntao Zou
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Zejie Zuo
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| | - Zhibin Yao
- a Department of Anatomy and Neurobiology ; Zhongshan School of Medicine; Sun Yat-sen University ; Guangzhou , PR China
| |
Collapse
|
11
|
Vidal PM, Lemmens E, Dooley D, Hendrix S. The role of “anti-inflammatory” cytokines in axon regeneration. Cytokine Growth Factor Rev 2013; 24:1-12. [DOI: 10.1016/j.cytogfr.2012.08.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 08/20/2012] [Indexed: 11/25/2022]
|
12
|
Ayer RE, Ostrowski RP, Sugawara T, Ma Q, Jafarian N, Tang J, Zhang JH. Statin-induced T-lymphocyte modulation and neuroprotection following experimental subarachnoid hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 115:259-66. [PMID: 22890678 DOI: 10.1007/978-3-7091-1192-5_46] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Statins influence immune system activities through mechanisms independent of their lipid-lowering properties. T cells can be subdivided based on cytokine secretion patterns into two subsets: T-helper cells type 1 (Th1) and type 2 (Th2). Independent laboratory studies have shown statins to be potent inducers of a Th2 switch in immune cell response and be neuroprotective in several models of central nervous system (CNS) disease. This study was the first to evaluate the immune modulating effects of statins in subarachnoid hemorrhage (SAH). METHODS Simvastatin was administered to rats intraperitoneally in two dosages (1 and 20 mg/kg) 30 min after the induction of SAH using endovascular perforation. Neurological scores were assessed 24 h later. Animals were then sacrificed, and samples of cortex and brain stem were tested for expression of the T-regulatory cell cytokine transforming growth factor (TGF) β1, as well as interleukin (IL) 1β, a proinflammatory cytokine associated with Th1 immune responses. The presence of TGF-β1 secreting T cells was evaluated with the use of brain slices. RESULTS SAH significantly impaired neurological function in all SAH groups (treated and untreated) versus sham. Animals treated with high-dose simvastatin had less neurological impairment than both untreated and low-dose groups. Cortical and brain-stem levels of TGF-β1 were significantly elevated following SAH in the high-dose group. IL-1β was significantly elevated following the induction of SAH but was inhibited by high-dose simvastatin. Double-labeled fluorescent immunohistochemical data demonstrated the presence of lymphocytes in the subarachnoid and perivascular spaces following SAH. Expression of TGF-β1 by lymphocytes was markedly increased following treatment with high-dose simvastatin. CONCLUSION The present study elucidated the potential role of a Th2 immune switch in statin provided neuroprotection following SAH.
Collapse
Affiliation(s)
- Robert E Ayer
- Department of Neurosurgery, Loma Linda University Medical Center, Loma Linda, CA, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Garg SK, Kipnis J, Banerjee R. IFN-gamma and IL-4 differentially shape metabolic responses and neuroprotective phenotype of astrocytes. J Neurochem 2009; 108:1155-66. [PMID: 19141080 DOI: 10.1111/j.1471-4159.2009.05872.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Astrocytes can either exacerbate or ameliorate secondary degeneration at sites of injury in the CNS but the contextual basis for eliciting these opposing phenotypes is poorly understood. In this study, we demonstrate that the two major cytokines produced by Th1 and Th2 cells, interferon-gamma (IFN-gamma), and interleukin-4 (IL-4), respectively, contribute differentially to shaping a neuroprotective response in astrocytes. While IFN-gamma protects the ability of oxidatively stressed murine astrocytes to clear extracellular glutamate in culture, IL-4 has no effect at any concentration that was tested (10-100 ng/mL). The enhanced release of neuroprotective thiols and lactate by astrocytes in response to T cell stimulation is mimicked by both IL-4 and IFN-gamma. When co-administered, IL-4 abrogated the protective effect of low IFN-gamma on the glutamate clearance function of oxidatively stressed astrocytes in a dose-dependent manner. Astrocyte-conditioned media obtained from cells cultured in the presence of IL-4 (10 or 100 ng/mL) or IFN-gamma (10 ng/mL) decreased by approximately 2-fold, neuronal apoptosis induced by oxidative stress in vitro. However, unlike IL-4, IFN-gamma at high concentrations (100 ng/mL) was not neuroprotective. Our studies with IFN-gamma and IL-4 suggest that a balanced Th1 and Th2 cytokine response might be needed for protecting two key astrocytic functions, glutamate clearance and thiol secretion and might be pertinent to neuroprotective approaches that are aimed at inhibition of an initial pro-inflammatory response to injury or its sustained boosting.
Collapse
Affiliation(s)
- Sanjay K Garg
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, 48109-0606, USA
| | | | | |
Collapse
|
14
|
Androgen depletion increases the efficacy of bone marrow transplantation in ameliorating experimental autoimmune encephalomyelitis. Blood 2008; 113:204-13. [PMID: 18824597 DOI: 10.1182/blood-2008-05-156042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bone marrow transplantation (BMT) potentially represents a novel therapy for the amelioration and even cure for multiple sclerosis (MS). It has important advantages over immunosuppressive drug treatments because, while effecting broad-based ablation of the immune system and autoreactive cells, it provides an important means for overcoming the resultant immunodeficiency, while possibly restoring self-tolerance. However, both of these benefits are predicated on a functional thymus that undergoes profound age-induced atrophy from puberty. Reversal of thymic atrophy has been achieved by several procedures, including removal of sex steroids by surgical or chemical (LHRH agonist) castration. Using a murine model of MS, experimental autoimmune encephalomyelitis (EAE), we combined BMT with androgen depletion to induce immune regeneration, and investigated the kinetics of increased thymic function on immune reconstitution and disease reduction. We show that androgen depletion significantly increased the efficacy of BMT to ameliorate the clinical signs of EAE while concurrently restoring the periphery with increased naive and regulatory lymphocytic populations. Upon rechallenge, mice with a regenerated thymus had a slower onset of clinical symptoms compared with mice undergoing BMT only. These results suggest that thymic regeneration strategies may be used as a complement to conventional BMT protocols for the treatment of MS.
Collapse
|
15
|
Gorantla S, Liu J, Sneller H, Dou H, Holguin A, Smith L, Ikezu T, Volsky DJ, Poluektova L, Gendelman HE. Copolymer-1 induces adaptive immune anti-inflammatory glial and neuroprotective responses in a murine model of HIV-1 encephalitis. THE JOURNAL OF IMMUNOLOGY 2007; 179:4345-56. [PMID: 17878329 DOI: 10.4049/jimmunol.179.7.4345] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Copolymer-1 (COP-1) elicits neuroprotective activities in a wide range of neurodegenerative disorders. This occurs, in part, by adaptive immune-mediated suppression of microglial inflammatory responses. Because HIV infection and immune activation of perivascular macrophages and microglia drive a metabolic encephalopathy, we reasoned that COP-1 could be developed as an adjunctive therapy for disease. To test this, we developed a novel animal model system that reflects HIV-1 encephalitis in rodents with both innate and adaptive arms of the immune system. Bone marrow-derived macrophages were infected with HIV-1/vesicular stomatitis-pseudotyped virus and stereotactically injected into the basal ganglia of syngeneic mice. HIV-1 pseudotyped with vesicular stomatitis virus envelope-infected bone marrow-derived macrophages induced significant neuroinflammation, including astrogliosis and microglial activation with subsequent neuronal damage. Importantly, COP-1 immunization reduced astro- and microgliosis while diminishing neurodegeneration. Hippocampal neurogenesis was, in part, restored. This paralleled reductions in proinflammatory cytokines, including TNF-alpha and IL-1beta, and inducible NO synthase, and increases in brain-derived neurotrophic factor. Ingress of Foxp3- and IL-4-expressing lymphocytes into brains of COP-1-immunized animals was observed. We conclude that COP-1 may warrant therapeutic consideration for HIV-1-associated cognitive impairments.
Collapse
Affiliation(s)
- Santhi Gorantla
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Smorodchenko A, Wuerfel J, Pohl EE, Vogt J, Tysiak E, Glumm R, Hendrix S, Nitsch R, Zipp F, Infante-Duarte C. CNS-irrelevant T-cells enter the brain, cause blood-brain barrier disruption but no glial pathology. Eur J Neurosci 2007; 26:1387-98. [PMID: 17880383 DOI: 10.1111/j.1460-9568.2007.05792.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Invasion of autoreactive T-cells and alterations of the blood-brain barrier (BBB) represent early pathological manifestations of multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). Non-CNS-specific T-cells are also capable of entering the CNS. However, studies investigating the spatial pattern of BBB alterations as well as the exact localization and neuropathological consequences of transferred non-CNS-specific cells have been thus far lacking. Here, we used magnetic resonance imaging and multiphoton microscopy, as well as histochemical and high-precision unbiased stereological analyses to compare T-cell transmigration, localization, persistence, relation to BBB disruption and subsequent effects on CNS tissue in a model of T-cell transfer of ovalbumin (OVA)- and proteolipid protein (PLP)-specific T-cells. BBB alterations were present in both EAE-mice and mice transferred with OVA-specific T-cells. In the latter case, BBB alterations were less pronounced, but the pattern of initial cell migration into the CNS was similar for both PLP- and OVA-specific cells [mean (SEM), 95 x 10(3) (7.6 x 10(3)) and 88 x 10(3) (18 x 10(3)), respectively]. Increased microglial cell density, astrogliosis and demyelination were, however, observed exclusively in the brain of EAE-mice. While mice transferred with non-neural-specific cells showed similar levels of rhodamine-dextran extravasation in susceptible brain regions, EAE-mice presented huge BBB disruption in brainstem and moderate leakage in cerebellum. This suggests that antigen specificity and not the absolute number of infiltrating cells determine the magnitude of BBB disruption and glial pathology.
Collapse
Affiliation(s)
- Alina Smorodchenko
- Cecilie-Vogt-Clinic for Molecular Neurology, Charité-Universitaetsmedizin Berlin and Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
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: 1.5] [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
|
18
|
Ramos SJ, Hardison JL, Stiles LN, Lane TE, Walsh CM. Anti-viral effector T cell responses and trafficking are not dependent upon DRAK2 signaling following viral infection of the central nervous system. Autoimmunity 2007; 40:54-65. [PMID: 17364498 DOI: 10.1080/08916930600996700] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The signaling events involved in T cell trafficking into the central nervous system (CNS) following viral infection are not fully understood. Intracerebral infection of mice with mouse hepatitis virus (MHV) results in an acute encephalomyelitis followed by an immune-mediated demyelinating disease. Although chemokine signaling is critical in promoting T cell infiltration into the CNS and control of viral replication, additional signaling pathways have not been completely explored. DRAK2, a lymphoid-restricted serine/threonine kinase, prevents spurious T cell activation. Yet Drak2- / - mice are resistant to MOG-induced experimental autoimmune encephalomyelitis (EAE), suggesting that DRAK2 may influence T cell trafficking into the CNS. In order to further characterize the molecular mechanisms governing T cell activation and accumulation within the CNS in response to viral infection, MHV was instilled into the CNS of Drak2- / - mice. Drak2-deficient T cells possessed no obvious defects in trafficking into the CNS following MHV infection. Moreover, Drak2-deficient T cell activation, expansion and cytokine production were unimpaired in response to acute MHV infection. These results demonstrate that DRAK2 signaling is dispensable for T cell recruitment into the CNS following viral infection, suggesting that the resistance of Drak2- / - mice to EAE is not due to overt T cell trafficking defects.
Collapse
Affiliation(s)
- Stephanie J Ramos
- Center for Immunology and Department of Molecular Biology & Biochemistry, University of California, Irvine, CA, 92697-3900, USA
| | | | | | | | | |
Collapse
|
19
|
Quirico-Santos T, Suppiah V, Heggarty S, Caetano R, Alves-Leon S, Vandenbroeck K. Study of polymorphisms in the interleukin-4 and IL-4 receptor genes in a population of Brazilian patients with multiple sclerosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2007; 65:15-9. [PMID: 17420820 DOI: 10.1590/s0004-282x2007000100005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Accepted: 10/05/2006] [Indexed: 11/22/2022]
Abstract
This study aimed to investigate in a population of Brazilian patients with multiple sclerosis (MS) single-nucleotide polymorphisms (SNP) in the promoter region of IL4 (*33C-T) and receptor IL4R (*Q551R A-G) genes proposed to interfere with disease progression. No significant differences were observed in either of the SNPs investigated between healthy controls (n=135) and MS patients (n=129). However, the IL4+33 TT genotype was significantly (p=0.039) higher in African descendants MS (AF-MS= 9.09%) than in Caucasian MS (CA-MS= 1.35%). It was also observed a significant (p=0.016) increase for the IL4R* Q551R CC genotype in AF-MS compared to those of Caucasian ethnicity (AF-MS= 21.62%; CA-MS= 4.35%). These results suggest that IL4+33 and IL4R*Q551 polymorphisms may have a disease-promoting role of TH2 mediators in African MS descendants. Additionally neither IL4 nor IL4R genes are susceptibility factors for Brazilian MS but may be able to modify ethnicity-dependent disease risk and penetrance of susceptibility factors.
Collapse
Affiliation(s)
- Thereza Quirico-Santos
- Applied Genomics Research Group, School of Pharmacy, Queens University Belfast, Belfast, UK.
| | | | | | | | | | | |
Collapse
|
20
|
Bullard DC, Hu X, Schoeb TR, Collins RG, Beaudet AL, Barnum SR. Intercellular adhesion molecule-1 expression is required on multiple cell types for the development of experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2007; 178:851-7. [PMID: 17202346 DOI: 10.4049/jimmunol.178.2.851] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Many members of the Ig superfamily of adhesion molecules, such as ICAM-1 and VCAM-1, have been implicated in the pathogenesis of multiple sclerosis. Although it is well-established that VCAM-1/VLA-4 interactions can play important roles in mediating CNS inflammatory events in multiple sclerosis patients and during the development of experimental allergic encephalomyelitis (EAE), the contributions of ICAM-1 are poorly understood. This is due in large part to conflicting results from Ab inhibition studies and the observation of exacerbated EAE in ICAM-1 mutant mice that express a restricted set of ICAM-1 isoforms. To determine ICAM-1-mediated mechanisms in EAE, we analyzed ICAM-1 null mutant mice (ICAM-1(null)), which express no ICAM-1 isoforms. ICAM-1(null) mice had significantly attenuated EAE characterized by markedly reduced spinal cord T cell infiltration and IFN-gamma production by these cells. Adoptive transfer of Ag-restimulated T cells from wild-type to ICAM-1(null) mice or transfer of ICAM-1(null) Ag-restimulated T cells to control mice failed to induce EAE. ICAM-1(null) T cells also showed reduced proliferative capacity and substantially reduced levels of IFN-gamma, TNF-alpha, IL-4, IL-10, and IL-12 compared with that of control T cells following myelin oligodendrocyte glycoprotein 35-55 restimulation in vitro. Our results indicate that ICAM-1 expression is critical on T cells and other cell types for the development of demyelinating disease and suggest that expression of VCAM-1 and other adhesion molecules cannot fully compensate for the loss of ICAM-1 during EAE development.
Collapse
Affiliation(s)
- Daniel C Bullard
- Department of Genetics, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
21
|
Elovitz MA, Mrinalini C, Sammel MD. Elucidating the early signal transduction pathways leading to fetal brain injury in preterm birth. Pediatr Res 2006; 59:50-5. [PMID: 16327009 DOI: 10.1203/01.pdr.0000191141.21932.b6] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Adverse neurologic outcome, including cerebral palsy, is a significant contributor to long-term morbidity in preterm neonates. However, the mechanisms leading to brain injury in the setting of a preterm birth are poorly understood. In the last decade, there has been a growing body of evidence correlating infection or inflammation with preterm birth. The presence of intrauterine inflammation significantly increases the risk for adverse neurologic outcome in the neonate. These studies were performed to elucidate the early signal transduction pathways activated in the fetal brain that may result in long-term neurologic injury. Using our mouse model of localized intrauterine inflammation, the activation of TH1/TH2 pathways in the placenta, fetus corpus, fetal liver, and fetal brain was investigated. Additional studies determined whether activation of TH1/TH2 pathways could promote cell death and alter glial development. Real-time PCR studies demonstrated that a robust TH1/TH2 response occurs rapidly in the fetal brain after exposure to intrauterine inflammation. The cytokine response in the fetus and placenta was not significantly correlated with the response in the fetal brain. Along with an immune response, cell death pathways were activated early in the fetal brain in response to intrauterine LPS. Implicating TH1/TH2 and cell death pathways in permanent brain injury are our findings of an increase in GFAP mRNA and protein as well as a loss of pro-oligodendrocytes. With increased understanding of the mechanisms by which inflammation promotes brain injury in the preterm neonate, identification of potential targets to limit adverse neonatal outcomes becomes possible.
Collapse
Affiliation(s)
- Michal A Elovitz
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia 19104-6142, USA.
| | | | | |
Collapse
|
22
|
Calvo CF, Amigou E, Desaymard C, Glowinski J. A pro- and an anti-inflammatory cytokine are synthesised in distinct brain macrophage cells during innate activation. J Neuroimmunol 2005; 170:21-30. [PMID: 16185773 DOI: 10.1016/j.jneuroim.2005.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 08/05/2005] [Indexed: 12/25/2022]
Abstract
Brain macrophages are known to exert dual and opposing functions on neuronal survival, which can be either beneficial or detrimental. The rationale of our study is that this duality could arise from an exclusive secretion of either pro- or anti-inflammatory cytokine by distinct cell subsets, cytokines that could respectively mediate neurotoxic or neurotrophic effects. Innate immune response was induced in macrophage cultures prepared from embryonic-day-16 to postnatal-day-8 mouse brains. By immunofluorescent detection of intracellular cytokines, we have assessed the occurrence of TNFalpha or IL10 synthesis at single cell level and observed distinct secretory patterns that include cells producing exclusively TNFalpha or IL10, cells producing both cytokines and non-producer cells. These secretory patterns are differentially regulated by MAP-kinase inhibitors. Altogether, these results demonstrate that synthesis of either a pro- or an anti-inflammatory cytokine can segregate distinct brain macrophages and suggests a functional cell-subset-specialisation.
Collapse
Affiliation(s)
- Charles-Félix Calvo
- Chaire de Neuropharmacologie, INSERM U114, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France.
| | | | | | | |
Collapse
|
23
|
Seiffert E, Dreier JP, Ivens S, Bechmann I, Tomkins O, Heinemann U, Friedman A. Lasting blood-brain barrier disruption induces epileptic focus in the rat somatosensory cortex. J Neurosci 2005; 24:7829-36. [PMID: 15356194 PMCID: PMC6729929 DOI: 10.1523/jneurosci.1751-04.2004] [Citation(s) in RCA: 372] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Perturbations in the integrity of the blood-brain barrier have been reported in both humans and animals under numerous pathological conditions. Although the blood-brain barrier prevents the penetration of many blood constituents into the brain extracellular space, the effect of such perturbations on the brain function and their roles in the pathogenesis of cortical diseases are unknown. In this study we established a model for focal disruption of the blood-brain barrier in the rat cortex by direct application of bile salts. Exposure of the cerebral cortex in vivo to bile salts resulted in long-lasting extravasation of serum albumin to the brain extracellular space and was associated with a prominent activation of astrocytes with no inflammatory response or marked cell loss. Using electrophysiological recordings in brain slices we found that a focus of epileptiform discharges developed within 4-7 d after treatment and could be recorded up to 49 d postoperatively in >60% of slices from treated animals but only rarely (10%) in sham-operated controls. Epileptiform activity involved both glutamatergic and GABAergic neurotransmission. Epileptiform activity was also induced by direct cortical application of native serum, denatured serum, or albumin-containing solution. In contrast, perfusion with serum-adapted electrolyte solution did not induce abnormal activity, thereby suggesting that the exposure of the serum-devoid brain environment to serum proteins underlies epileptogenesis in the blood-brain barrier-disrupted cortex. Although many neuropathologies entail a compromised blood-brain barrier, this is the first direct evidence that it may have a role in the pathogenesis of focal cortical epilepsy, a common neurological disease.
Collapse
Affiliation(s)
- Ernst Seiffert
- Johannes-Müller-Institute of Physiology, Charité, University Medicine, 10117 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
24
|
Fontoura P, Ho PP, DeVoss J, Zheng B, Lee BJ, Kidd BA, Garren H, Sobel RA, Robinson WH, Tessier-Lavigne M, Steinman L. Immunity to the extracellular domain of Nogo-A modulates experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2005; 173:6981-92. [PMID: 15557195 DOI: 10.4049/jimmunol.173.11.6981] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nogo-66, the extracellular 66 aa loop of the Nogo-A protein found in CNS myelin, interacts with the Nogo receptor and has been proposed to mediate inhibition of axonal regrowth. It has been shown that immunization with Nogo-A promotes recovery in animal models of spinal cord injury through induction of Ab production. In this report, studies were performed to characterize the immune response to Nogo-66 and to determine the role of Nogo in experimental autoimmune encephalomyelitis (EAE). Immunization of EAE-susceptible mouse strains with peptides derived from Nogo-66 induced a CNS immune response with clinical and pathological similarities to EAE. The Nogo-66 peptides elicited strong T cell responses that were not cross-reactive to other encephalitogenic myelin Ags. Using a large scale spotted microarray containing proteins and peptides derived from a wide spectrum of myelin components, we demonstrated that Nogo-66 peptides also generated a specific Ab response that spreads to several other encephalitogenic myelin Ags following immunization. Nogo-66-specific T cell lines ameliorated established EAE, via Nogo-66-specific Th2 cells that entered the CNS. These results indicate that some T cell and B cell immune responses to Nogo-66 are associated with suppression of ongoing EAE, whereas other Nogo-66 epitopes can be encephalitogenic.
Collapse
MESH Headings
- Adoptive Transfer
- Amino Acid Sequence
- Animals
- B-Lymphocytes/immunology
- Cell Line
- Disease Susceptibility
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/immunology
- Extracellular Space/immunology
- Female
- Immunity, Innate/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Myelin Proteins/administration & dosage
- Myelin Proteins/deficiency
- Myelin Proteins/genetics
- Myelin Proteins/immunology
- Nogo Proteins
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Protein Isoforms/administration & dosage
- Protein Isoforms/deficiency
- Protein Isoforms/genetics
- Protein Isoforms/immunology
- Protein Structure, Tertiary
- Species Specificity
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Paulo Fontoura
- Department of Neurology and Neurological Sciences, School of Medicine, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vrbić M, Vrbić S, Skorić S, Mihailović V, Ranković Z, Konstantinović L, Kostić V, Krstić M, Jovanović M. [Soluble CD4 antigens in the cerebrospinal fluid as a marker of lymphocytic infiltration of the central nervous system]. VOJNOSANIT PREGL 2004; 61:247-53. [PMID: 15330296 DOI: 10.2298/vsp0403247v] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Early diagnosis of the central nervous system (CNS) infections is a precondition of their successful treatment. However, the essential standard examination of the cerebrospinal fluid (CSF) is sometimes neither specific enough to define their basic nature, nor sufficient to differentiate them from processes of non-infectious origin. Supposing that the released surface molecules of activated immunocompetent cells could better define the character of inflammatory reaction, the levels of soluble CD4 antigens (sCD4) were determined with enzyme-immunosorbent test in the CSF of the patients with various CNS diseases. In contrast to cerebrovascular insults, toxic-metabolic, and other conditions in control group, detectable sCD4 concentrations in acute encephalitis (24 +/- 11 U/ml) were verified at the beginning of the disease, being also present in cytologically diagnosed normal CSF findings. They were significantly higher (p<0.05) compared to acute serous meningitis (13.5 +/- 8 U/ml), while in purulent meningitis they were measurable only after the disease progression--in correlation with the disturbed brain system function. The obtained results suggested the significance of CD4 antigen levels in CSF as a sensitive and specific marker of lymphocytic infiltration of the brain parenchyma, the measurement of which could contribute to early identification of the CNS infections, better understanding of their pathogenesis, and the assessment of the actual level of the destruction of neurons.
Collapse
Affiliation(s)
- Miodrag Vrbić
- Klinicki centar, Klinika za infektivne bolesti, Nis.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Innate responses in the CNS are critical to first line defense against infection and injury. Leukocytes migrate to inflammatory sites in response to chemokines. We studied leukocyte migration and glial chemokine expression within the denervated hippocampus in response to axonal injury caused by entorhinodentate lesions. A population of Mac1/CD11b+ CD45high macrophages (distinct from CD45low microglia) was specifically detected within the lesion-reactive hippocampus by 12 hr after injury. Significant infiltration by CD3+ T cells did not occur in the denervated hippocampus until 24 hr after axotomy. A broad spectrum of chemokines [RANTES/CCL5, monocyte chemoattractant protein (MCP)-1/CCL2, interferon gamma inducible protein (IP)-10/CXCL10, macrophage inflammatory protein (MIP)-1alpha/CCL3, MIP-1beta/CCL4, and MIP-2/CXCL2] was induced at this time. RANTES/CCL5 was not significantly elevated until 24 hr after axotomy, whereas MCP-1/CCL2 was significantly induced before leukocyte infiltration occurred. Neither T cells nor macrophages infiltrated the denervated hippocampus of CCR2-deficient mice, arguing for a critical role for the CCR2 ligand MCP-1/CCL2 in leukocyte migration. Both T cells and macrophages infiltrated CCR5-deficient hippocampi, showing that CCR5 ligands (including RANTES/CCL5) are not critical to this response. In situ hybridization combined with immunohistochemistry for ionized binding calcium adapter molecule (iba)1 or glial fibrillary acidic protein (GFAP) identified iba1+ microglia and GFAP+ astrocytes as major sources of MCP-1/CCL2 within the lesion-reactive hippocampus. We conclude that leukocyte responses to CNS axonal injury are directed via innate glial production of chemokines.
Collapse
|
27
|
Séguin R, Biernacki K, Prat A, Wosik K, Kim HJ, Blain M, McCrea E, Bar-Or A, Antel JP. Differential effects of Th1 and Th2 lymphocyte supernatants on human microglia. Glia 2003; 42:36-45. [PMID: 12594735 DOI: 10.1002/glia.10201] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We assessed the effects of soluble molecules (supernatants) produced by pro- (Th1) and anti- (Th2) inflammatory T-cell lines on the capacity of adult human CNS-derived microglia to express or produce selected cell surface and soluble molecules that regulate immune reactivity or impact on tissue protection/repair within the CNS. Treatment of microglia with supernatants from allo-antigen and myelin basic protein-specific Th1 cell lines augmented expression of cell surface molecules MHC class II, CD80, CD86, CD40, and CD54, enhanced the functional antigen-presenting cell capacity of microglia in a mixed lymphocyte reaction, and increased cytokine/chemokine secretion (TNFalpha, IL-6, and CXCL10/IP-10). These Th1-induced effects were not reproduced by interferon-gamma (IFNgamma) alone and were only incompletely blocked by anti-IFNgamma antibody. Th2 cell supernatant treatments did not alter costimulatory/adhesion molecule expression or induce cytokine/chemokine production by microglia. Th2 treatment, furthermore, failed to reduce the induction observed in response to Th1 supernatants. Neither Th1 nor Th2 supernatants induced production of the neurotrophin molecules, nerve growth factor, or brain-derived neurotrophic factor. Our results suggest that soluble molecules released by Th1 and not Th2 cells that infiltrate the CNS can stimulate resident microglia to acquire enhanced effector and accessory cell functions; the Th1-induced effects were not downregulated by Th2 supernatant-mediated bystander suppression.
Collapse
Affiliation(s)
- Rosanne Séguin
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Quebec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Emes RD, Wang WZ, Lanary K, Blackshaw SE. HmCRIP, a cysteine-rich intestinal protein, is expressed by an identified regenerating nerve cell. FEBS Lett 2003; 533:124-8. [PMID: 12505171 DOI: 10.1016/s0014-5793(02)03741-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A Hirudo medicinalis cDNA isolated from regenerating CNS tissue at 24 h post-axotomy was identified as a leech homologue of the mammalian cysteine-rich intestinal proteins (CRIPs) and named HmCRIP. HmCRIP is up-regulated within 6 h of axotomy, peaking at 24 h. This is the first demonstration of a CRIP homologue in regenerating CNS and in a serotonergic neurone. In rodents CRIP is an important factor in the regulation of the inflammatory immune response through control of Th1/Th2 differentiation. The role of HmCRIP in the regeneration competent environment of the annelid central nervous system is discussed.
Collapse
Affiliation(s)
- R D Emes
- MRC Functional Genetics Unit, Department of Human Anatomy and Genetics, University of Oxford, South Parks Road, OX1 3QX, Oxford, UK.
| | | | | | | |
Collapse
|
29
|
Wolf SA, Fisher J, Bechmann I, Steiner B, Kwidzinski E, Nitsch R. Neuroprotection by T-cells depends on their subtype and activation state. J Neuroimmunol 2002; 133:72-80. [PMID: 12446010 DOI: 10.1016/s0165-5728(02)00367-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This study analyzes how the antigen specificity, the subtype, and the activation state of T cells modulate their recently discovered neuroprotective potential. We assessed the prevention from neuronal damage in organotypic entorhinal-hippocampal slice cultures after co-culture with Th1 and Th2 cells either specific for myelin basic protein (MBP) or ovalbumin (OVA). We found that MBP-specific Th2 cells were the most effective in preventing central nervous system (CNS) tissue from secondary injury. This neuroprotective T cell effect appears to be mediated by soluble factors. After stimulation with phorbol myristate acetate and ionomycin, all T cells were most effective in preventing neuronal death. Our data show that the T cell subtype and activation state are important features in determining the neuroprotective potential of these cells.
Collapse
Affiliation(s)
- Susanne A Wolf
- Department of Cell and Neurobiology, Institute of Anatomy, Humboldt-University Hospital Charitè, 10098, Berlin, Germany
| | | | | | | | | | | |
Collapse
|