1
|
Chiang W, Urban JM, Yanchik-Slade F, Stout A, Hammond JM, Nilsson BL, Gelbard HA, Krauss TD. Hybrid Amyloid Quantum Dot Nano-Bio Assemblies to Probe Neuroinflammatory Damage. ACS Chem Neurosci 2024; 15:3124-3135. [PMID: 39146244 PMCID: PMC11378299 DOI: 10.1021/acschemneuro.4c00183] [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: 03/27/2024] [Revised: 06/29/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
Various oligomeric species of amyloid-beta have been proposed to play different immunogenic roles in the cellular pathology of Alzheimer's Disease. The dynamic interconversion between various amyloid oligomers and fibrillar assemblies makes it difficult to elucidate the role each potential aggregation state may play in driving neuroinflammatory and neurodegenerative pathology. The ability to identify the amyloid species that are key and essential drivers of these pathological hallmarks of Alzheimer's Disease is of fundamental importance for also understanding downstream events including tauopathies that mediate neuroinflammation with neurologic deficits. Here, we report the design and construction of a quantum dot mimetic for larger spherical oligomeric amyloid species as an "endogenously" fluorescent proxy for this cytotoxic assembly of amyloid to investigate its role in inducing inflammatory and stress response states in neuronal and glial cell types. The design parameters and construction protocol developed here may be adapted for developing quantum dot nano-bio assemblies for other biological systems of interest, particularly neurodegenerative diseases involving other protein aggregates.
Collapse
Affiliation(s)
- Wesley Chiang
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
- Department
of Biochemistry and Biophysics, University
of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jennifer M. Urban
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Francine Yanchik-Slade
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Angela Stout
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jennetta M. Hammond
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Bradley L. Nilsson
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Harris A. Gelbard
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
- Departments
of Pediatrics, Neuroscience, and Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Todd D. Krauss
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
- The
Institute of Optics, University of Rochester
Medical Center, Rochester, New York 14627-0216, United States
| |
Collapse
|
2
|
Tan Y, Lai T, Li Y, Tang Q, Zhang W, Liu Q, Wu S, Peng X, Sui X, Reggiori F, Jiang X, Chen Q, Wang C. An oil-in-gel type of organohydrogel loaded with methylprednisolone for the treatment of secondary injuries following spinal cord traumas. J Control Release 2024; 374:505-524. [PMID: 39182693 DOI: 10.1016/j.jconrel.2024.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The secondary injuries following traumatic spinal cord injury (SCI) is a multiphasic and complex process that is difficult to treat. Although methylprednisolone (MP) is the only available pharmacological regime for SCI treatment, its efficacy remains controversial due to its very narrow therapeutic time window and safety concerns associated with high dosage. In this study, we have developed an oil-in-gel type of organohydrogel (OHG) in which the binary oleic-water phases coexist, for the local delivery of MP. This new OHG is fabricated by a glycol chitosan/oxidized hyaluronic acid hydrophilic network that is uniformly embedded with a biocompatible oil phase, and it can be effectively loaded with MP or other hydrophobic compounds. In addition to spatiotemporally control MP release, this biodegradable OHG also provides a brain tissue-mimicking scaffold that can promote tissue regeneration. OHG remarkably decreases the therapeutic dose of MP in animals and extends its treatment course over 21 d, thereby timely manipulating microglia/macrophages and their associated with signaling molecules to restore immune homeostasis, leading to a long-term functional improvement in a complete transection SCI rat model. Thus, this OHG represents a new type of gel for clinical treatment of secondary injuries in SCI.
Collapse
Affiliation(s)
- Yinqiu Tan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, PR China; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Ting Lai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Yuntao Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Qi Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Weijia Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Qi Liu
- The First Dongguan Affiliated Hospital Guangdong Medical University No. 42, Jiaoping Road Dongguan, Guangdong 523710, PR China
| | - Sihan Wu
- Center for Biomedical Optics and Photonics (CBOP)&College of Physics and Optoelectronic Engineering, Key Lab of Optoelectronics Devices and systems of Ministry of Education/Guangdong Province, Shenzhen University, Shenzhen 518060, PR China
| | - Xiao Peng
- Center for Biomedical Optics and Photonics (CBOP)&College of Physics and Optoelectronic Engineering, Key Lab of Optoelectronics Devices and systems of Ministry of Education/Guangdong Province, Shenzhen University, Shenzhen 518060, PR China
| | - Xiaofeng Sui
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, PR China
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus C, Denmark; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Høegh-Guldbergs Gade 6B, 8000 Aarhus C, Denmark.
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, PR China.
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.
| | - Cuifeng Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Department of Neurosurgery, JiuJiang Hospital of Traditional Chinese Medicine, Jiujiang, PR China.
| |
Collapse
|
3
|
Chiang W, Urban JM, Yanchik-Slade F, Stout A, Nilsson BL, Gelbard HA, Krauss TD. Hybrid Amyloid Quantum Dot Nanoassemblies to Probe Neuroinflammatory Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555592. [PMID: 37693630 PMCID: PMC10491264 DOI: 10.1101/2023.08.30.555592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Various oligomeric species of amyloid-beta have been proposed to play different immunogenic roles in the cellular pathology of Alzheimer's Disease. However, investigating the role of a homogenous single oligomeric species has been difficult due to highly dynamic oligomerization and fibril formation kinetics that convert between many species. Here we report the design and construction of a quantum dot mimetic for larger spherical oligomeric amyloid species as an "endogenously" fluorescent proxy for this cytotoxic species to investigate its role in inducing inflammatory and stress response states in neuronal and glial cell types.
Collapse
Affiliation(s)
- Wesley Chiang
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, 14642
| | - Jennifer M. Urban
- Department of Chemistry, Rochester, New York 14627-0216, United States
| | | | - Angela Stout
- Center for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642
| | | | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642
- Departments of Pediatrics, Neuroscience, and Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Todd D. Krauss
- Department of Chemistry, Rochester, New York 14627-0216, United States
- The Institute of Optics, Rochester, New York 14627-0216, United States
| |
Collapse
|
4
|
Juul SE, Voldal E, Comstock BA, Massaro AN, Bammler TK, Mayock DE, Heagerty PJ, Wu YW, Numis AL. Association of High-Dose Erythropoietin With Circulating Biomarkers and Neurodevelopmental Outcomes Among Neonates With Hypoxic Ischemic Encephalopathy: A Secondary Analysis of the HEAL Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2322131. [PMID: 37418263 PMCID: PMC10329214 DOI: 10.1001/jamanetworkopen.2023.22131] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/18/2023] [Indexed: 07/08/2023] Open
Abstract
Importance The ability to predict neurodevelopmental impairment (NDI) for infants diagnosed with hypoxic ischemic encephalopathy (HIE) is important for parental guidance and clinical treatment as well as for stratification of patients for future neurotherapeutic studies. Objectives To examine the effect of erythropoietin on plasma inflammatory mediators in infants with moderate or severe HIE and to develop a panel of circulating biomarkers that improves the projection of 2-year NDI over and above the clinical data available at the time of birth. Design, Setting, and Participants This study is a preplanned secondary analysis of prospectively collected data from infants enrolled in the High-Dose Erythropoietin for Asphyxia and Encephalopathy (HEAL) Trial, which tested the efficacy of erythropoietin as an adjunctive neuroprotective therapy to therapeutic hypothermia. The study was conducted at 17 academic sites comprising 23 neonatal intensive care units in the United States between January 25, 2017, and October 9, 2019, with follow-up through October 2022. Overall, 500 infants born at 36 weeks' gestation or later with moderate or severe HIE were included. Intervention Erythropoietin treatment 1000 U/kg/dose on days 1, 2, 3, 4 and 7. Main Outcomes and Measures Plasma erythropoietin was measured in 444 infants (89%) within 24 hours after birth. A subset of 180 infants who had plasma samples available at baseline (day 0/1), day 2, and day 4 after birth and either died or had 2-year Bayley Scales of Infant Development III assessments completed were included in the biomarker analysis. Results The 180 infants included in this substudy had a mean (SD) gestational age of 39.1 (1.5) weeks, and 83 (46%) were female. Infants who received erythropoietin had increased concentrations of erythropoietin at day 2 and day 4 compared with baseline. Erythropoietin treatment did not alter concentrations of other measured biomarkers (eg, difference in interleukin [IL] 6 between groups on day 4: -1.3 pg/mL; 95% CI, -4.8 to 2.0 pg/mL). After adjusting for multiple comparisons, we identified 6 plasma biomarkers (C5a, interleukin [IL] 6, and neuron-specific enolase at baseline; IL-8, tau, and ubiquitin carboxy-terminal hydrolase-L1 at day 4) that significantly improved estimations of death or NDI at 2 years compared with clinical data alone. However, the improvement was only modest, increasing the AUC from 0.73 (95% CI, 0.70-0.75) to 0.79 (95% CI, 0.77-0.81; P = .01), corresponding to a 16% (95% CI, 5%-44%) increase in correct classification of participant risk of death or NDI at 2 years. Conclusions and Relevance In this study, erythropoietin treatment did not reduce biomarkers of neuroinflammation or brain injury in infants with HIE. Circulating biomarkers modestly improved estimation of 2-year outcomes. Trial Registration ClinicalTrials.gov Identifier: NCT02811263.
Collapse
|
5
|
Jin S, Eussen SJPM, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Plasma factor D is cross-sectionally associated with low-grade inflammation, endothelial dysfunction and cardiovascular disease: The Maastricht study. Atherosclerosis 2023; 377:60-67. [PMID: 37406499 DOI: 10.1016/j.atherosclerosis.2023.06.079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND AND AIMS The complement system, particularly the alternative complement pathway, may contribute to vascular damage and development of cardiovascular disease (CVD). We investigated the association of factor D, the rate-limiting protease in alternative pathway activation, with adverse cardiovascular outcomes. METHODS In 2947 participants (50.6% men, 59.9 ± 8.2 years, 26.5% type 2 diabetes [T2D], oversampled) we measured markers of low-grade inflammation (LGI, composite score, in SD) and, endothelial dysfunction (ED, composite score, in SD), carotid intima-media thickness (cIMT, μm), ankle-brachial index (ABI), CVD (yes/no) and plasma concentrations of factor D (in SD). Associations were estimated using multiple linear and logistic regression, adjusting for demographic, lifestyle, and dietary factors. RESULTS Factor D (per SD) significantly associated with LGI (0.171 SD [0.137; 0.205]), ED (0.158 SD [0.123; 0.194]) and CVD (OR 1.15 [1.04; 1.27]) but not significantly with cIMT (-6.62 μm [-13.51; 0.27]) or ABI (-0.003 [-0.007; 0.001]). Interaction analyses show that factor D more strongly associated with ED in non-diabetes (0.237 SD [0.189; 0.285] than in T2D (0.095 SD [0.034; 0.157]), pinteraction <0.05. These results were largely corroborated by additional analyses with C3 and C3a. In contrast, factor D inversely associated with cIMT in non-diabetes (-13.37 μm [-21.84; -4.90]), but not in T2D (4.49 [-7.91; 16.89]), pinteraction <0.05. CONCLUSIONS Plasma factor D is independently associated with LGI, ED, and prevalent CVD but not with ABI or cIMT. Hence, greater plasma factor D concentration in CVD may potentially induce complement activation which, in turn, might contribute to further disease progression via a process that may involve inflammation and endothelial dysfunction but was not directly related to atherosclerosis or arterial injury. The observation that, in participants without diabetes, factor D associated with worse ED but smaller cIMT warrants further investigation.
Collapse
Affiliation(s)
- Shunxin Jin
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, the Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM School for Cardiovascular Diseases, the Netherlands; CAPHRI School for Public Health and Primary Care, Maastricht University and Maastricht University Medical Centre, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, the Netherlands
| | | |
Collapse
|
6
|
Stennett A, Friston K, Harris CL, Wollman AJM, Bronowska AK, Madden KS. The case for complement component 5 as a target in neurodegenerative disease. Expert Opin Ther Targets 2023; 27:97-109. [PMID: 36786123 DOI: 10.1080/14728222.2023.2177532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Complement-based drug discovery is undergoing a renaissance, empowered by new advances in structural biology, complement biology and drug development. Certain components of the complement pathway, particularly C1q and C3, have been extensively studied in the context of neurodegenerative disease, and established as key therapeutic targets. C5 also has huge therapeutic potential in this arena, with its druggability clearly demonstrated by the success of C5-inhibitor eculizumab. AREAS COVERED We will discuss the evidence supporting C5 as a target in neurodegenerative disease, along with the current progress in developing different classes of C5 inhibitors and the gaps in knowledge that will help progress in the field. EXPERT OPINION Validation of C5 as a therapeutic target for neurodegenerative disease would represent a major step forward for complement therapeutics research and has the potential to furnish disease-modifying drugs for millions of patients suffering worldwide. Key hurdles that need to be overcome for this to be achieved are understanding how C5a and C5b should be targeted to bring therapeutic benefit and demonstrating the ability to target C5 without creating vulnerability to infection in patients. This requires greater biological elucidation of its precise role in disease pathogenesis, supported by better chemical/biological tools.
Collapse
Affiliation(s)
- Amelia Stennett
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK
| | - Kallie Friston
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK
| | - Claire L Harris
- Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle-Upon-Tyne, UK
| | - Adam J M Wollman
- Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle-Upon-Tyne, UK
| | - Agnieszka K Bronowska
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK
| | - Katrina S Madden
- School of Natural and Environmental Sciences, Newcastle University, NE1 7RU, Newcastle-Upon-Tyne, UK.,Faculty of Medical Sciences, Newcastle University, NE2 4HH, Newcastle-Upon-Tyne, UK
| |
Collapse
|
7
|
Wu M, Chen K, Jiang M, Xie F, Cao X, Chen L, Chen Z, Yin X. High plasma complement C4 levels as a novel predictor of clinical outcome in intracerebral hemorrhage. Front Aging Neurosci 2023; 15:1103278. [PMID: 36891553 PMCID: PMC9986541 DOI: 10.3389/fnagi.2023.1103278] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/20/2023] [Indexed: 02/22/2023] Open
Abstract
Objective The complement cascade is activated and contributes to the brain injury after intracerebral hemorrhage (ICH). Complement component 4 (C4), an important component of complement cascade, has been associated with severity of neurological impairment that occurs during ICH. However, the correlation of plasma complement C4 levels with hemorrhagic severity and clinical outcome in ICH patients has not been reported. Materials and methods This study is a monocentric, real-world, cohort study. In this study, we measured the plasma complement C4 levels of 83 ICH patients and 78 healthy controls. The hematoma volume, the National Institutes of Health Stroke Scale (NIHSS) score, the Glasgow Coma Scale (GCS) score, and the permeability surface (PS) were used to assess and quantify neurological deficit following ICH. Logistic regression analysis was configured to determine the independent relation of plasma complement C4 levels to hemorrhagic severity and clinical outcomes. The contribution of complement C4 to secondary brain injury (SBI) was assessed by changes in plasma C4 levels between admission and at day 7 after ICH. Results There was a significant elevation of plasma complement C4 levels in ICH patients than in healthy controls (40.48 ± 1.07 vs. 35.25 ± 0.60, p < 0.0001), and the plasma complement C4 levels were closely related to the hemorrhagic severity. Moreover, plasma complement C4 levels of patients were positively correlated with the hematoma volume (r = 0.501, p < 0.001), NIHSS score (r = 0.362, p < 0.001), the GCS score (r = -0.490, p < 0.001), and PS (r = 0.683, p = 0.045) following ICH. Logistic regression analysis also confirmed that patients with high plasma complement C4 levels show a poor clinical outcome after ICH (p < 0.001). Meanwhile, the elevated plasma levels at day 7 after ICH indicated the correlation of complement C4 with SBI (p < 0.01). Conclusion Plasma complement C4 levels are significantly elevated in ICH patients and positively correlated with the illness severity. Thus, these findings highlight the importance of complement C4 in brain injury after ICH and provide a novel predictor of clinical outcome for this disease.
Collapse
Affiliation(s)
- Moxin Wu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, China.,Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
| | - Kai Chen
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
| | - Fusheng Xie
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xianming Cao
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Liang Chen
- Department of Radiology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China.,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China.,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| |
Collapse
|
8
|
O'Shea TM, Ao Y, Wang S, Wollenberg AL, Kim JH, Ramos Espinoza RA, Czechanski A, Reinholdt LG, Deming TJ, Sofroniew MV. Lesion environments direct transplanted neural progenitors towards a wound repair astroglial phenotype in mice. Nat Commun 2022; 13:5702. [PMID: 36171203 PMCID: PMC9519954 DOI: 10.1038/s41467-022-33382-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 09/14/2022] [Indexed: 01/30/2023] Open
Abstract
Neural progenitor cells (NPC) represent potential cell transplantation therapies for CNS injuries. To understand how lesion environments influence transplanted NPC fate in vivo, we derived NPC expressing a ribosomal protein-hemagglutinin tag (RiboTag) for transcriptional profiling of transplanted NPC. Here, we show that NPC grafted into uninjured mouse CNS generate cells that are transcriptionally similar to healthy astrocytes and oligodendrocyte lineages. In striking contrast, NPC transplanted into subacute CNS lesions after stroke or spinal cord injury in mice generate cells that share transcriptional, morphological and functional features with newly proliferated host astroglia that restrict inflammation and fibrosis and isolate lesions from adjacent viable neural tissue. Our findings reveal overlapping differentiation potentials of grafted NPC and proliferating host astrocytes; and show that in the absence of other interventions, non-cell autonomous cues in subacute CNS lesions direct the differentiation of grafted NPC towards a naturally occurring wound repair astroglial phenotype.
Collapse
Affiliation(s)
- T M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA.
| | - Y Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA
| | - S Wang
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA
| | - A L Wollenberg
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - J H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA
| | - R A Ramos Espinoza
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215-2407, USA
| | - A Czechanski
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | - T J Deming
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - M V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1763, USA.
| |
Collapse
|
9
|
Brennan FH, Li Y, Wang C, Ma A, Guo Q, Li Y, Pukos N, Campbell WA, Witcher KG, Guan Z, Kigerl KA, Hall JCE, Godbout JP, Fischer AJ, McTigue DM, He Z, Ma Q, Popovich PG. Microglia coordinate cellular interactions during spinal cord repair in mice. Nat Commun 2022; 13:4096. [PMID: 35835751 PMCID: PMC9283484 DOI: 10.1038/s41467-022-31797-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/01/2022] [Indexed: 12/27/2022] Open
Abstract
Traumatic spinal cord injury (SCI) triggers a neuro-inflammatory response dominated by tissue-resident microglia and monocyte derived macrophages (MDMs). Since activated microglia and MDMs are morphologically identical and express similar phenotypic markers in vivo, identifying injury responses specifically coordinated by microglia has historically been challenging. Here, we pharmacologically depleted microglia and use anatomical, histopathological, tract tracing, bulk and single cell RNA sequencing to reveal the cellular and molecular responses to SCI controlled by microglia. We show that microglia are vital for SCI recovery and coordinate injury responses in CNS-resident glia and infiltrating leukocytes. Depleting microglia exacerbates tissue damage and worsens functional recovery. Conversely, restoring select microglia-dependent signaling axes, identified through sequencing data, in microglia depleted mice prevents secondary damage and promotes recovery. Additional bioinformatics analyses reveal that optimal repair after SCI might be achieved by co-opting key ligand-receptor interactions between microglia, astrocytes and MDMs.
Collapse
Affiliation(s)
- Faith H Brennan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yang Li
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Qi Guo
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yi Li
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nicole Pukos
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Warren A Campbell
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristina G Witcher
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Zhen Guan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristina A Kigerl
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Jodie C E Hall
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Dana M McTigue
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Lin W, Xin Z, Wang J, Ren X, Liu Y, Yang L, Guo S, Yang Y, Li Y, Cao J, Ning X, Liu M, Su Y, Sun L, Zhang F, Zhang W. Hypocomplementemia in primary Sjogren’s syndrome: association with serological, clinical features, and outcome. Clin Rheumatol 2022; 41:2091-2102. [PMID: 35348930 PMCID: PMC9187545 DOI: 10.1007/s10067-022-06135-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/07/2022] [Accepted: 03/13/2022] [Indexed: 11/03/2022]
Abstract
Abstract
Objective
The aim of the present study was to assess the clinical characteristic of hypocomplementemia (HC) in primary Sjogren’s syndrome (pSS), and to address possible risk factors and the prognosis associated with HC in pSS patients.
Methods
pSS patients with HC in Hebei General Hospital from September 2016 to March 2019 were retrospectively analyzed and compared to those with normocomplementemia (NC). Logistic regression analysis was used to detect risk factors.
Results
Of the 333 patients with pSS, 84 patients (25.23%) were presented with HC at diagnosis. The presence of hyper-IgG and anti-Ro52 antibodies was significantly more common in patients with HC. In addition to systemic involvement, pSS patients with HC had more hematological, renal, and nervous system involvement, and received more immunosuppressant treatments than NC group (p < 0.05). ESSDAI score was significantly higher in patients with HC (p < 0.05). Multivariate logistic analysis indicated that leukopenia (OR = 2.23) and hyper-IgG (OR = 2.13) were independent risk factors for pSS with HC. In addition, profound CD16/CD56+ NK-cell lymphopenia was found in pSS-HC patients. More pSS patients developed SLE in the HC group than NC group (4.76% vs. 0.80%, p = 0.04) during the follow-up.
Conclusion
HC was not an uncommon manifestation of pSS and had an independent association with the main clinical and immunological features. Patients with pSS-HC had an increased possibility to develop SLE that required more positive treatment with glucocorticoids and immunosuppressants.
Key Points:
• Hypocomplementemia had an independent association with the main clinical and immunological features in primary Sjogren’s syndrome patients.
• ESSDAI score was significantly higher in patients with hypocomplementemia.
• The pSS patients with hypocomplementemia had an increased possibility to develop SLE.
Collapse
|
11
|
Berkowitz S, Chapman J, Dori A, Gofrit SG, Maggio N, Shavit-Stein E. Complement and Coagulation System Crosstalk in Synaptic and Neural Conduction in the Central and Peripheral Nervous Systems. Biomedicines 2021; 9:biomedicines9121950. [PMID: 34944766 PMCID: PMC8698364 DOI: 10.3390/biomedicines9121950] [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: 11/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Complement and coagulation are both key systems that defend the body from harm. They share multiple features and are similarly activated. They each play individual roles in the systemic circulation in physiology and pathophysiology, with significant crosstalk between them. Components from both systems are mapped to important structures in the central nervous system (CNS) and peripheral nervous system (PNS). Complement and coagulation participate in critical functions in neuronal development and synaptic plasticity. During pathophysiological states, complement and coagulation factors are upregulated and can modulate synaptic transmission and neuronal conduction. This review summarizes the current evidence regarding the roles of the complement system and the coagulation cascade in the CNS and PNS. Possible crosstalk between the two systems regarding neuroinflammatory-related effects on synaptic transmission and neuronal conduction is explored. Novel treatment based on the modulation of crosstalk between complement and coagulation may perhaps help to alleviate neuroinflammatory effects in diseased states of the CNS and PNS.
Collapse
Affiliation(s)
- Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Robert and Martha Harden Chair in Mental and Neurological Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Amir Dori
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan 6997801, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-50-921-0400
| |
Collapse
|
12
|
Allinovi M, Bellinvia A, Pesce F, Milan Manani S, Razzolini L, Brezzi B, Protopapa P, Mantero V, Caroti L, Cirami CL, Amato MP, Del Vecchio L. Safety and Efficacy of Eculizumab Therapy in Multiple Sclerosis: A Case Series. Brain Sci 2021; 11:1341. [PMID: 34679405 PMCID: PMC8533854 DOI: 10.3390/brainsci11101341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Complement system activation has been proposed as one of the different factors that contribute to Multiple Sclerosis (MS) pathogenesis. In this study, we aimed to describe the potential effects of eculizumab, an anticomplement therapy, on MS disease activity in a cohort of relapsing-remitting (RR) MS patients who discontinued IFN-β therapy due to IFN-β-related thrombotic microangiopathy (TMA) onset. (2) Methods: In this retrospective observational multicentric study, we searched for all patients with MS treated by eculizumab with a survey of several nephrological and neurological centers (over 45 centers). (3) Results: Nine patients were included. The mean follow-up time under eculizumab was 3.72 ± 2.58 years. There were no significant differences in disease activity (EDSS, relapses, new T2, and/or Gd-enhancing lesions at MRI) considering the two years before and after eculizumab therapy. No adverse events potentially related to eculizumab therapy were reported during follow-up. (4) Conclusions: In this preliminary study, we described a good safety profile for eculizumab therapy in MS. However, the available data are not sufficient to make firm conclusions about the possible efficacy of eculizumab as a disease-modifying therapy for MS patients.
Collapse
Affiliation(s)
- Marco Allinovi
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, 50139 Florence, Italy; (L.C.); (C.L.C.)
| | - Angelo Bellinvia
- NEUROFARBA Department, University of Florence, 50139 Florence, Italy; (A.B.); (L.R.); (M.P.A.)
| | - Francesco Pesce
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari, 70121 Bari, Italy; (F.P.); (P.P.)
| | - Sabrina Milan Manani
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, 36100 Vicenza, Italy;
| | - Lorenzo Razzolini
- NEUROFARBA Department, University of Florence, 50139 Florence, Italy; (A.B.); (L.R.); (M.P.A.)
| | - Brigida Brezzi
- Division of Nephrology and Dialysis, SS Antonio e Biagio e C. Arrigo Hospital, 15121 Alessandria, Italy;
| | - Paolo Protopapa
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari, 70121 Bari, Italy; (F.P.); (P.P.)
| | - Vittorio Mantero
- MS Center, Department of Neurology, ASST Lecco, 23900 Lecco, Italy;
| | - Leonardo Caroti
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, 50139 Florence, Italy; (L.C.); (C.L.C.)
| | - Calogero Lino Cirami
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, 50139 Florence, Italy; (L.C.); (C.L.C.)
| | - Maria Pia Amato
- NEUROFARBA Department, University of Florence, 50139 Florence, Italy; (A.B.); (L.R.); (M.P.A.)
- IRCCS Don Carlo Gnocchi, 50143 Florence, Italy
| | | |
Collapse
|
13
|
Gomez-Arboledas A, Acharya MM, Tenner AJ. The Role of Complement in Synaptic Pruning and Neurodegeneration. Immunotargets Ther 2021; 10:373-386. [PMID: 34595138 PMCID: PMC8478425 DOI: 10.2147/itt.s305420] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
The complement system, an essential part of the innate immune system, is composed of a group of secreted and membrane proteins that collectively participate in maintaining the function of the healthy and diseased brain. However, an inappropriate activation of the complement system has been related to an inflammatory response in multiple diseases, such as stroke, traumatic brain injury, multiple sclerosis, and Alzheimer's disease, as well as Zika infection and radiotherapy. In addition, C1q and C3 (initial activation components of the complement cascade) have been shown to play a key beneficial role in the refinement of synaptic circuits during developmental stages and adult plasticity. Nevertheless, excessive synaptic pruning in the adult brain can be detrimental and has been associated with synaptic loss in several pathological conditions. In this brief review, we will discuss the role of the complement system in synaptic pruning as well as its contribution to neurodegeneration and cognitive deficits. We also mention potential therapeutic approaches to target the complement system to treat several neuroinflammatory diseases and unintended consequences of radiotherapy.
Collapse
Affiliation(s)
- Angela Gomez-Arboledas
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
14
|
The complement cascade in the regulation of neuroinflammation, nociceptive sensitization, and pain. J Biol Chem 2021; 297:101085. [PMID: 34411562 PMCID: PMC8446806 DOI: 10.1016/j.jbc.2021.101085] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 01/13/2023] Open
Abstract
The complement cascade is a key component of the innate immune system that is rapidly recruited through a cascade of enzymatic reactions to enable the recognition and clearance of pathogens and promote tissue repair. Despite its well-understood role in immunology, recent studies have highlighted new and unexpected roles of the complement cascade in neuroimmune interaction and in the regulation of neuronal processes during development, aging, and in disease states. Complement signaling is particularly important in directing neuronal responses to tissue injury, neurotrauma, and nerve lesions. Under physiological conditions, complement-dependent changes in neuronal excitability, synaptic strength, and neurite remodeling promote nerve regeneration, tissue repair, and healing. However, in a variety of pathologies, dysregulation of the complement cascade leads to chronic inflammation, persistent pain, and neural dysfunction. This review describes recent advances in our understanding of the multifaceted cross-communication that takes place between the complement system and neurons. In particular, we focus on the molecular and cellular mechanisms through which complement signaling regulates neuronal excitability and synaptic plasticity in the nociceptive pathways involved in pain processing in both health and disease. Finally, we discuss the future of this rapidly growing field and what we believe to be the significant knowledge gaps that need to be addressed.
Collapse
|
15
|
Chen M, Edwards SR, Reutens DC. Complement in the Development of Post-Traumatic Epilepsy: Prospects for Drug Repurposing. J Neurotrauma 2021; 37:692-705. [PMID: 32000582 DOI: 10.1089/neu.2019.6942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeting neuroinflammation is a novel frontier in the prevention and treatment of epilepsy. A substantial body of evidence supports a key role for neuroinflammation in epileptogenesis, the pathological process that leads to the development and progression of spontaneous recurrent epileptic seizures. It is also well recognized that traumatic brain injury (TBI) induces a vigorous neuroinflammatory response and that a significant proportion of patients with TBI suffer from debilitating post-traumatic epilepsy. The complement system is a potent effector of innate immunity and a significant contributor to secondary tissue damage and to epileptogenesis following central nervous system injury. Several therapeutic agents targeting the complement system are already on the market to treat other central nervous system disorders or are well advanced in their development. The purpose of this review is to summarize findings on complement activation in experimental TBI and epilepsy models, highlighting the potential of drug repurposing in the development of therapeutics to ameliorate post-traumatic epileptogenesis.
Collapse
Affiliation(s)
- Min Chen
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| | - Stephen R Edwards
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| | - David C Reutens
- Center for Advanced Imaging, University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
16
|
Traylor M, Malik R, Gesierich B, Dichgans M. The BS variant of C4 protects against age-related loss of white matter microstructural integrity. Brain 2021; 145:295-304. [PMID: 34358307 DOI: 10.1093/brain/awab261] [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: 02/23/2021] [Revised: 05/12/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Age-related loss of white matter microstructural integrity is a major determinant of cognitive decline, dementia, and gait disorders. However, the mechanisms and molecular pathways that contribute to this loss of integrity remain elusive. We performed a GWAS of white matter microstructural integrity as quantified by diffusion MRI metrics (mean diffusivity, MD; and fractional anisotropy, FA) in up to 31,128 individuals from UK Biobank (age 45-81 years) based on a 2 degrees of freedom (2df) test of single nucleotide polymorphism (SNP) and SNP x age effects. We identified 18 loci that were associated at genome-wide significance with either MD (N = 16) or FA (N = 6). Among the top loci was a region on chromosome 6 encoding the human major histocompatibility complex (MHC). Variants in the MHC region were strongly associated with both MD (best SNP: 6:28866209_TTTTG_T, beta(SE)=-0.069(0.009); 2df p = 6.5x10-15) and FA (best SNP: rs3129787, beta(SE)=-0.056(0.008); 2df p = 3.5x10-12). Of the imputed HLA alleles and complement component 4 (C4) structural haplotype variants in the human MHC, the strongest association was with the C4-BS variant (for MD: beta(SE)=-0.070(0.010); p = 2.7x10-11; for FA: beta(SE)=-0.054(0.011); p = 1.6x10-7). After conditioning on C4-BS no associations with HLA alleles remained significant. The protective influence of C4-BS was stronger in older subjects (age ≥ 65; interaction p = 0.0019 (MD), p = 0.015 (FA)) and in subjects without a history of smoking (interaction p = 0.00093 (MD), p = 0.021 (FA)). Taken together, our findings demonstrate a role of the complement system and of gene-environment interactions in age-related loss of white matter microstructural integrity.
Collapse
Affiliation(s)
- Matthew Traylor
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK.,The Barts Heart Centre and NIHR Barts Biomedical Research Centre-Barts Health NHS Trust, The William Harvey Research Institute, Queen Mary University London, London, UK
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.,German Centre for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
17
|
Stokum JA, Cannarsa GJ, Wessell AP, Shea P, Wenger N, Simard JM. When the Blood Hits Your Brain: The Neurotoxicity of Extravasated Blood. Int J Mol Sci 2021; 22:5132. [PMID: 34066240 PMCID: PMC8151992 DOI: 10.3390/ijms22105132] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022] Open
Abstract
Hemorrhage in the central nervous system (CNS), including intracerebral hemorrhage (ICH), intraventricular hemorrhage (IVH), and aneurysmal subarachnoid hemorrhage (aSAH), remains highly morbid. Trials of medical management for these conditions over recent decades have been largely unsuccessful in improving outcome and reducing mortality. Beyond its role in creating mass effect, the presence of extravasated blood in patients with CNS hemorrhage is generally overlooked. Since trials of surgical intervention to remove CNS hemorrhage have been generally unsuccessful, the potent neurotoxicity of blood is generally viewed as a basic scientific curiosity rather than a clinically meaningful factor. In this review, we evaluate the direct role of blood as a neurotoxin and its subsequent clinical relevance. We first describe the molecular mechanisms of blood neurotoxicity. We then evaluate the clinical literature that directly relates to the evacuation of CNS hemorrhage. We posit that the efficacy of clot removal is a critical factor in outcome following surgical intervention. Future interventions for CNS hemorrhage should be guided by the principle that blood is exquisitely toxic to the brain.
Collapse
Affiliation(s)
- Jesse A. Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Gregory J. Cannarsa
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Aaron P. Wessell
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Phelan Shea
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Nicole Wenger
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
- Departments of Pathology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
18
|
Brennan FH, Coulthard LG, Alawieh AM, Reiner O, Pekna M. Editorial: Complement in the Development and Regeneration of the Nervous System. Front Immunol 2021; 12:694810. [PMID: 34040618 PMCID: PMC8141916 DOI: 10.3389/fimmu.2021.694810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- Faith H Brennan
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Liam G Coulthard
- Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,Faculty of Medicine, University of Queensland, Herston, QLD, Australia
| | - Ali M Alawieh
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Marcela Pekna
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
19
|
Wang K, Li J, Zhang Y, Huang Y, Chen D, Shi Z, Smith AD, Li W, Gao Y. Central nervous system diseases related to pathological microglial phagocytosis. CNS Neurosci Ther 2021; 27:528-539. [PMID: 33650762 PMCID: PMC8025646 DOI: 10.1111/cns.13619] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/18/2022] Open
Abstract
Microglia are important phagocytes of the central nervous system (CNS). They play an important role in protecting the CNS by clearing necrotic tissue and apoptotic cells in many CNS diseases. However, recent studies have found that microglia can phagocytose parts of neurons excessively, such as the neuronal cell body, synapse, or myelin sheaths, before or after the onset of CNS diseases, leading to aggravated injury and impaired tissue repair. Meanwhile, reduced phagocytosis of synapses and myelin results in abnormal circuit connections and inhibition of remyelination, respectively. Previous studies focused primarily on the positive effects of microglia phagocytosis, whereas only a few studies have focused on the negative effects. In this review, we use the term "pathological microglial phagocytosis" to refer to excessive or reduced phagocytosis by microglia that leads to structural or functional abnormalities in target cells and brain tissue. The classification of pathological microglial phagocytosis, the composition, and activation of related signaling pathways, as well as the process of pathological phagocytosis in various kinds of CNS diseases, are described in this review. We hypothesize that pathological microglial phagocytosis leads to aggravation of tissue damage and negative functional outcome. For example, excessive microglial phagocytosis of synapses can be observed in Alzheimer's disease and schizophrenia, leading to significant synapse loss and memory impairment. In Parkinson's disease, ischemic stroke, and traumatic brain injury, excessive microglial phagocytosis of neuronal cell bodies causes impaired gray matter recovery and sensory dysfunction. We therefore believe that more studies should focus on the mechanism of pathological microglial phagocytosis and activation to uncover potential targets of therapeutic intervention.
Collapse
Affiliation(s)
- Ke Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yue Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Di Chen
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ziyu Shi
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Amanda D Smith
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Wei Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Shah TA, Pallera HK, Kaszowski CL, Bass WT, Lattanzio FA. Therapeutic Hypothermia Inhibits the Classical Complement Pathway in a Rat Model of Neonatal Hypoxic-Ischemic Encephalopathy. Front Neurosci 2021; 15:616734. [PMID: 33642979 PMCID: PMC7907466 DOI: 10.3389/fnins.2021.616734] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/25/2021] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE Complement activation is instrumental in the pathogenesis of Hypoxic-ischemic encephalopathy (HIE), a significant cause of neonatal mortality and disability worldwide. Therapeutic hypothermia (HT), the only available treatment for HIE, only modestly improves outcomes. Complement modulation as a therapeutic adjunct to HT has been considered, but is challenging due to the wide-ranging role of the complement system in neuroinflammation, homeostasis and neurogenesis in the developing brain. We sought to identify potential therapeutic targets by measuring the impact of treatment with HT on complement effector expression in neurons and glia in neonatal HIE, with particular emphasis on the interactions between microglia and C1q. METHODS The Vannucci model was used to induce HIE in term-equivalent rat pups. At P10-12, pups were randomly assigned to three different treatment groups: Sham (control), normothermia (NT), and hypothermia (HT) treatment. Local and systemic complement expression and neuronal apoptosis were measured by ELISA, TUNEL and immunofluorescence labeling, and differences compared between groups. RESULTS Treatment with HT is associated with decreased systemic and microglial expression of C1q, decreased systemic C5a levels, and decreased microglial and neuronal deposition of C3 and C9. The effect of HT on cytokines was variable with decreased expression of pro and anti-inflammatory effectors. HT treatment was associated with decreased C1q binding on cells undergoing apoptosis. CONCLUSION Our data demonstrate the extreme complexity of the immune response in neonatal HIE. We propose modulation of downstream effectors C3a and C5a as a therapeutic adjunct to HT to enhance neuroprotection in the developing brain.
Collapse
Affiliation(s)
- Tushar A. Shah
- Department of Pediatrics, Eastern Virginia Medical School, Norfolk, VA, United States
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
- Children’s Specialty Group, Norfolk, VA, United States
- Children’s Hospital of The King’s Daughters, Norfolk, VA, United States
| | - Haree K. Pallera
- Department of Pediatrics, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | - William Thomas Bass
- Department of Pediatrics, Eastern Virginia Medical School, Norfolk, VA, United States
- Children’s Specialty Group, Norfolk, VA, United States
- Children’s Hospital of The King’s Daughters, Norfolk, VA, United States
| | - Frank A. Lattanzio
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
21
|
Bennett C, Álvarez-Ciara A, Franklin M, Dietrich WD, Prasad A. The complement cascade at the Utah microelectrode-tissue interface. Biomaterials 2021; 268:120583. [PMID: 33310540 PMCID: PMC7856077 DOI: 10.1016/j.biomaterials.2020.120583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/16/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023]
Abstract
Devices implanted within the central nervous system (CNS) are subjected to tissue reactivity due to the lack of biocompatibility between implanted material and the cells' microenvironment. Studies have attributed blood-brain barrier disruption, inflammation, and oxidative stress as main contributing factors that lead to electrode recording failure. The complement cascade is a part of the innate immunity that focuses on recognizing and targeting foreign objects; however, its role in the context of neural implants is substantially unknown. In this study, we implanted a non-functional 4x4 Utah microelectrode array (UEA) into the somatosensory cortex and studied the complement cascade via combined gene and immunohistochemistry quantification at acute (48-h), sub-acute (1-week), and early chronic (4-weeks) time points. The results of this study demonstrate the activation and continuation of the complement cascade at the electrode-tissue interface, illustrating the therapeutic potential of modulating the foreign body response via the complement cascade.
Collapse
Affiliation(s)
- Cassie Bennett
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | - Melissa Franklin
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, FL, USA.
| |
Collapse
|
22
|
Popiolek-Barczyk K, Ciechanowska A, Ciapała K, Pawlik K, Oggioni M, Mercurio D, De Simoni MG, Mika J. The CCL2/CCL7/CCL12/CCR2 pathway is substantially and persistently upregulated in mice after traumatic brain injury, and CCL2 modulates the complement system in microglia. Mol Cell Probes 2020; 54:101671. [PMID: 33160071 DOI: 10.1016/j.mcp.2020.101671] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/15/2020] [Accepted: 11/01/2020] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of death in the global population. Disturbed inflammatory processes after TBI exacerbate secondary brain injury and contribute to unfavorable outcomes. Multiple inflammatory events that accompany brain trauma, such as glial activation, chemokine release, or the initiation of the complement system cascade, have been identified as potential targets for TBI treatment. However, the participation of chemokines in the complement activation remains unknown. Our studies sought to determine the changes in the expression of the molecules involved in the CCL2/CCL7/CCL12/CCR2 pathway in the injured brain and the effect of CCL2, CCL7, and CCL12 (10, 100, and 500 ng/mL) on the classic and lectin complement pathways and inflammatory factors in microglial cell cultures. Brain injury in mice was modeled by controlled cortical impact (CCI). Our findings indicate a time-dependent upregulation of CCL2, CCL7, and CCL12 at the mRNA and protein levels within the cortex, striatum, and/or thalamus beginning 24 h after the trauma. The analysis of the expression of the receptor of the tested chemokines, CCR2, revealed its substantial upregulation within the injured brain areas mainly on the mRNA level. Using primary cortical microglial cell cultures, we observed a substantial increase in the expression of CCL2, CCL7, and CCL12 after 24 h of LPS (100 ng/mL) treatment. CCL2 stimulation of microglia increased the level of IL-1β mRNA but did not influence the expression of IL-18, IL-6, and IL-10. Moreover, CCL2 significantly increased the expression of Iba1, a marker of microglia activation. CCL2 and CCL12 upregulated the expression of C1qa but did not influence the expression of C1ra and C1s1 (classical pathway); moreover, CCL2 increased ficolin A expression and reduced collectin 11 expression (lectin pathway). Additionally, we observed the downregulation of pentraxin 3, a modulator of the complement cascade, after CCL2 and CCL12 treatment. We did not detect the expression of ficolin B, Mbl1, and Mbl2 in microglial cells. Our data identify CCL2 as a modulator of the classical and lectin complement pathways suggesting that CCL2 may be a promising target for pharmacological intervention after brain injury. Moreover, our study provides evidence that CCL2 and two other CCR2 ligands may play a role in the development of changes in TBI.
Collapse
Affiliation(s)
- Katarzyna Popiolek-Barczyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Agata Ciechanowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Katarzyna Ciapała
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Katarzyna Pawlik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Marco Oggioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Domenico Mercurio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Joanna Mika
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland.
| |
Collapse
|
23
|
Asano S, Hayashi Y, Iwata K, Okada-Ogawa A, Hitomi S, Shibuta I, Imamura Y, Shinoda M. Microglia-Astrocyte Communication via C1q Contributes to Orofacial Neuropathic Pain Associated with Infraorbital Nerve Injury. Int J Mol Sci 2020; 21:ijms21186834. [PMID: 32957694 PMCID: PMC7560139 DOI: 10.3390/ijms21186834] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/29/2022] Open
Abstract
Trigeminal nerve injury causes a distinct time window of glial activation in the trigeminal spinal subnucleus caudalis (Vc), which are involved in the initiation and maintenance phases of orofacial neuropathic pain. Microglia-derived factors enable the activation of astrocytes. The complement component C1q, which promotes the activation of astrocytes, is known to be synthesized in microglia. However, it is unclear whether microglia–astrocyte communication via C1q is involved in orofacial neuropathic pain. Here, we analyzed microglia-astrocyte communication in a rat model with infraorbital nerve injury (IONI). The orofacial mechanical hypersensitivity induced by IONI was significantly attenuated by preemptive treatment with minocycline. Immunohistochemical analyses revealed that minocycline inhibited the increase in c-Fos immune-reactive (IR) cells and the fluorescence intensity of both Iba1 and glial fibrillary acidic protein (GFAP) in the Vc following IONI. Intracisternal administration of C1q caused orofacial mechanical hypersensitivity and an increase in the number of c-Fos-IR cells and fluorescence intensity of GFAP. C1q-induced orofacial mechanical hypersensitivity was completely abrogated by intracisternal administration of fluorocitrate. The present findings suggest that the enhancement in the excitability of Vc nociceptive neurons is produced by astrocytic activation via the signaling of C1q released from activated microglia in the Vc following IONI, resulting in persistent orofacial neuropathic pain.
Collapse
Affiliation(s)
- Sayaka Asano
- Department of Oral Diagnostic Science, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (S.A.); (A.O.-O.); (Y.I.)
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (K.I.); (S.H.); (I.S.); (M.S.)
- Correspondence: ; Tel.: +81-3-3219-8122
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (K.I.); (S.H.); (I.S.); (M.S.)
| | - Akiko Okada-Ogawa
- Department of Oral Diagnostic Science, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (S.A.); (A.O.-O.); (Y.I.)
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (K.I.); (S.H.); (I.S.); (M.S.)
| | - Ikuko Shibuta
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (K.I.); (S.H.); (I.S.); (M.S.)
| | - Yoshiki Imamura
- Department of Oral Diagnostic Science, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (S.A.); (A.O.-O.); (Y.I.)
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan; (K.I.); (S.H.); (I.S.); (M.S.)
| |
Collapse
|
24
|
Mallah K, Couch C, Borucki DM, Toutonji A, Alshareef M, Tomlinson S. Anti-inflammatory and Neuroprotective Agents in Clinical Trials for CNS Disease and Injury: Where Do We Go From Here? Front Immunol 2020; 11:2021. [PMID: 33013859 PMCID: PMC7513624 DOI: 10.3389/fimmu.2020.02021] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Neurological disorders are major contributors to death and disability worldwide. The pathology of injuries and disease processes includes a cascade of events that often involve molecular and cellular components of the immune system and their interaction with cells and structures within the central nervous system. Because of this, there has been great interest in developing neuroprotective therapeutic approaches that target neuroinflammatory pathways. Several neuroprotective anti-inflammatory agents have been investigated in clinical trials for a variety of neurological diseases and injuries, but to date the results from the great majority of these trials has been disappointing. There nevertheless remains great interest in the development of neuroprotective strategies in this arena. With this in mind, the complement system is being increasingly discussed as an attractive therapeutic target for treating brain injury and neurodegenerative conditions, due to emerging data supporting a pivotal role for complement in promoting multiple downstream activities that promote neuroinflammation and degeneration. As we move forward in testing additional neuroprotective and immune-modulating agents, we believe it will be useful to review past trials and discuss potential factors that may have contributed to failure, which will assist with future agent selection and trial design, including for complement inhibitors. In this context, we also discuss inhibition of the complement system as a potential neuroprotective strategy for neuropathologies of the central nervous system.
Collapse
Affiliation(s)
- Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Davis M. Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Mohammed Alshareef
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurological Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Ralph Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
25
|
Slomnicki LP, Myers SA, Saraswat Ohri S, Parsh MV, Andres KR, Chariker JH, Rouchka EC, Whittemore SR, Hetman M. Improved locomotor recovery after contusive spinal cord injury in Bmal1 -/- mice is associated with protection of the blood spinal cord barrier. Sci Rep 2020; 10:14212. [PMID: 32848194 PMCID: PMC7450087 DOI: 10.1038/s41598-020-71131-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/10/2020] [Indexed: 11/09/2022] Open
Abstract
The transcription factor BMAL1/ARNTL is a non-redundant component of the clock pathway that regulates circadian oscillations of gene expression. Loss of BMAL1 perturbs organismal homeostasis and usually exacerbates pathological responses to many types of insults by enhancing oxidative stress and inflammation. Surprisingly, we observed improved locomotor recovery and spinal cord white matter sparing in Bmal1-/- mice after T9 contusive spinal cord injury (SCI). While acute loss of neurons and oligodendrocytes was unaffected, Bmal1 deficiency reduced the chronic loss of oligodendrocytes at the injury epicenter 6 weeks post SCI. At 3 days post-injury (dpi), decreased expression of genes associated with cell proliferation, neuroinflammation and disruption of the blood spinal cord barrier (BSCB) was also observed. Moreover, intraspinal extravasation of fibrinogen and immunoglobulins was decreased acutely at dpi 1 and subacutely at dpi 7. Subacute decrease of hemoglobin deposition was also observed. Finally, subacutely reduced levels of the leukocyte marker CD45 and even greater reduction of the pro-inflammatory macrophage receptor CD36 suggest not only lower numbers of those cells but also their reduced inflammatory potential. These data indicate that Bmal1 deficiency improves SCI outcome, in part by reducing BSCB disruption and hemorrhage decreasing cytotoxic neuroinflammation and attenuating the chronic loss of oligodendrocytes.
Collapse
Affiliation(s)
- Lukasz P Slomnicki
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Scott A Myers
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA.
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA.
| | - Molly V Parsh
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Julia H Chariker
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY, 40202, USA
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
| | - Eric C Rouchka
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY, 40202, USA
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, 40202, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville, Louisville, KY, 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA.
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, 40202, USA.
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 S. Floyd St., MDR616, Louisville, KY, 40292, USA.
| |
Collapse
|
26
|
Girardi G, Lingo JJ, Fleming SD, Regal JF. Essential Role of Complement in Pregnancy: From Implantation to Parturition and Beyond. Front Immunol 2020; 11:1681. [PMID: 32849586 PMCID: PMC7411130 DOI: 10.3389/fimmu.2020.01681] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
The complement cascade was identified over 100 years ago, yet investigation of its role in pregnancy remains an area of intense research. Complement inhibitors at the maternal-fetal interface prevent inappropriate complement activation to protect the fetus. However, this versatile proteolytic cascade also favorably influences numerous stages of pregnancy, including implantation, fetal development, and labor. Inappropriate complement activation in pregnancy can have adverse lifelong sequelae for both mother and child. This review summarizes the current understanding of complement activation during all stages of pregnancy. In addition, consequences of complement dysregulation during adverse pregnancy outcomes from miscarriage, preeclampsia, and pre-term birth are examined. Finally, future research directions into complement activation during pregnancy are considered.
Collapse
Affiliation(s)
- Guillermina Girardi
- Department of Basic Medical Sciences, College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Joshua J Lingo
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, United States
| |
Collapse
|
27
|
Ma WJ, Shi YH, Chen J. Ayu (Plecoglossus altivelis) CD46 isoforms protect the cells from autologous complement attack. FISH & SHELLFISH IMMUNOLOGY 2020; 102:267-275. [PMID: 32360277 DOI: 10.1016/j.fsi.2020.04.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/19/2020] [Accepted: 04/23/2020] [Indexed: 06/11/2023]
Abstract
CD46 is an important immune regulatory receptor with multiple functions. However, studies on the function of teleost CD46, especially the different CD46 isoforms are limited. In this study, we identified three membrane cofactor protein (MCP, CD46) gene isoforms from ayu (Plecoglossus altivelis) and tentatively named as PaCD46 isoforms. PaCD46 isoforms were generated by alternative splicing and all consisted of four conserved short consensus repeats (SCRs), and the variable serine-threonine-proline-rich domain, transmembrane hydrophobic domain, and cytoplasmic tail. Phylogenetic analysis showed that the isoforms clustered together with other fish CD46 and then with higher animal CD46. Western blotting analysis of peripheral blood mononuclear cells (PBMC) revealed three bands, all of which had much larger molecular weights than the theoretical values of the three PaCD46 isoforms. Moreover, three PaCD46 isoforms were individually expressed on HEK293 cells, and Western blotting showed the similar band profile to that of PBMC. The recombinant extracellular domain of the PaCD46 isoforms, obtained by expression in Pichia pastoris, significantly reduced hemolysis activity of ayu sera. Furthermore, each of the three PaCD46 isoforms respectively protected the HEK293 cells expressing the isoform. The isoforms were also identified for their protection of autologous PBMC from complement activation. These results provided the first evidence that PaCD46 isoforms may be complement regulatory proteins to prevent complement-induced damage to self-tissue.
Collapse
Affiliation(s)
- Wen-Jing Ma
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Yu-Hong Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
28
|
Tan N, Hu S, Hu Z, Wu Z, Wang B. Quantitative proteomic characterization of microvesicles/exosomes from the cerebrospinal fluid of patients with acute bilirubin encephalopathy. Mol Med Rep 2020; 22:1257-1268. [PMID: 32468033 PMCID: PMC7339682 DOI: 10.3892/mmr.2020.11194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/06/2020] [Indexed: 01/10/2023] Open
Abstract
Severe hyperbilirubinemia causes neurotoxicity and may lead to acute bilirubin encephalopathy (ABE) during the critical period of central nervous system development. The aim of the present study was to identify differentially expressed proteins (DEPs) in microvesicles/exosomes (MV/E) isolated from the cerebrospinal fluid (CSF) of patients with ABE. Co-precipitation was used to isolate the MV/E from the CSF of patients with ABE and age-matched controls. Isobaric tagging for relative and absolute quantification-based proteomic technology combined with liquid chromatography/tandem mass spectrometry was used to identify DEPs in the MV/E. Bioinformatics analysis was subsequently performed to investigate Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes enriched signaling pathways of these DEPs. A total of four proteins were selected for further validation via western blotting. A total of 291 dysregulated proteins were identified by comparing the patients with ABE with the controls. Bioinformatics analysis indicated the involvement of immune-inflammation-associated cellular processes and signaling pathways in the pathophysiology of ABE. In conclusion, the present study identified the proteomic profile of MV/E isolated from the CSF of patients with ABE. These results may provide an improved understanding of the pathogenesis of ABE and may help to identify early diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ning Tan
- Department of Pediatrics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Shuiwang Hu
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhen Hu
- National and Local Joint Engineering Laboratory for High‑through Molecular Diagnosis Technology, Translational Medicine Institute, Collaborative Research Center for Post‑doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, Southern Medical University, University of South China, Chenzhou, Hunan 423000, P.R. China
| | - Zhouli Wu
- Department of Neonatology, Affiliated The First People's Hospital of Chenzhou, Southern Medical University, University of South China, Chenzhou, Hunan 423000, P.R. China
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
29
|
Magdalon J, Mansur F, Teles E Silva AL, de Goes VA, Reiner O, Sertié AL. Complement System in Brain Architecture and Neurodevelopmental Disorders. Front Neurosci 2020; 14:23. [PMID: 32116493 PMCID: PMC7015047 DOI: 10.3389/fnins.2020.00023] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/10/2020] [Indexed: 01/18/2023] Open
Abstract
Current evidence indicates that certain immune molecules such as components of the complement system are directly involved in neurobiological processes related to brain development, including neurogenesis, neuronal migration, synaptic remodeling, and response to prenatal or early postnatal brain insults. Consequently, complement system dysfunction has been increasingly implicated in disorders of neurodevelopmental origin, such as schizophrenia, autism spectrum disorder (ASD) and Rett syndrome. However, the mechanistic evidence for a causal relationship between impaired complement regulation and these disorders varies depending on the disease involved. Also, it is still unclear to what extent altered complement expression plays a role in these disorders through inflammation-independent or -dependent mechanisms. Furthermore, pathogenic mutations in specific complement components have been implicated in the etiology of 3MC syndrome, a rare autosomal recessive developmental disorder. The aims of this review are to discuss the current knowledge on the roles of the complement system in sculpting brain architecture and function during normal development as well as after specific inflammatory insults, such as maternal immune activation (MIA) during pregnancy, and to evaluate the existing evidence associating aberrant complement with developmental brain disorders.
Collapse
Affiliation(s)
- Juliana Magdalon
- Center for Experimental Research, Hospital Israelita Albert Einstein, São Paulo, Brazil.,School of Medicine, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
| | - Fernanda Mansur
- Center for Experimental Research, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - André Luiz Teles E Silva
- Center for Experimental Research, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, Brazil
| | - Vitor Abreu de Goes
- Center for Experimental Research, Hospital Israelita Albert Einstein, São Paulo, Brazil.,School of Medicine, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Andréa Laurato Sertié
- Center for Experimental Research, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
30
|
Muk T, Stensballe A, Pankratova S, Nguyen DN, Brunse A, Sangild PT, Jiang PP. Rapid Proteome Changes in Plasma and Cerebrospinal Fluid Following Bacterial Infection in Preterm Newborn Pigs. Front Immunol 2019; 10:2651. [PMID: 31803186 PMCID: PMC6873289 DOI: 10.3389/fimmu.2019.02651] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/25/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Neonatal infection and sepsis are common for preterm infants due to their immature immune system. Early diagnosis is important for effective treatment, but few early markers of systemic and neuro-inflammatory responses in neonates are known. We hypothesised that systemic infection with Staphylococcus epidermidis (SE), a Gram-positive bacteria, induces acute changes to proteins in the plasma and cerebrospinal fluid (CSF), potentially affecting the immature brain of preterm neonates. Methods: Using preterm pigs as a model for preterm infants, plasma and CSF samples were collected up to 24 h after SE infection and investigated by untargeted mass spectrometry (MS)-based proteomics. Multiple differentially expressed proteins were further studied in vitro. Results: The clinical signs of sepsis and neuroinflammation in SE-infected piglets were associated with changes of multiple CSF and plasma proteins. Eight plasma proteins, including APOA4, haptoglobin, MBL1, vWF, LBP, and sCD14, were affected 6 h after infection. Acute phase reactants, including complement components, showed a time-dependent activation pattern after infection. Feeding bovine colostrum reduced the sepsis-related changes in clinical indices and plasma proteins. Neuroinflammation-related neuropeptide Y (NPY), IL-18, and MMP-14 showed distinct changes in the CSF and several brain regions (the prefrontal cortex, PVWM, and hippocampus) 24 h after infection. These changes were verified in TLR2 agonist-challenged primary microglia cells, where exogenous NPY suppressed the inflammatory response. Conclusion: Systemic infection with SE induces inflammation with rapid proteome changes in the plasma and CSF in preterm newborn pigs. The observed early markers of sepsis and neuroinflammation in preterm pigs may serve as novel biomarkers for sepsis in preterm infants.
Collapse
Affiliation(s)
- Tik Muk
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Stanislava Pankratova
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Department of Neonatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Brunse
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per Torp Sangild
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Neonatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Paediatrics, Odense University Hospital, Odense, Denmark
| | - Ping-Ping Jiang
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.,School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
Wei L, Zhang J, Zhang B, Geng J, Tan Q, Wang L, Chen Z, Feng H, Zhu G. Complement C3 participates in the function and mechanism of traumatic brain injury at simulated high altitude. Brain Res 2019; 1726:146423. [PMID: 31654641 DOI: 10.1016/j.brainres.2019.146423] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) leads to severe mortality and disability, in which secondary injury induced by complement activation plays an important role. TBI tends to be associated with more severe cerebral edema and worse neurological functional recovery if it occurs in high-altitude areas than in low-altitude areas. However, the underlying mechanism of this difference is unknown. Thus, we used cobra venom factor (CVF) to deplete complement C3 in simulated high-altitude areas to explore whether the differences in outcome at different altitudes are related to secondary injury caused by complement C3. METHODS The weight-drop model was adopted to induce TBI in rats. Rats were randomly divided into the following groups: sham + saline (sham), high altitude + TBI + saline (HAT), high altitude + TBI + CVF (H-CVF), low altitude + TBI + saline (LAT), and low altitude + TBI + CVF (L-CVF). Brain contusion and edema volumes, brain water content, myelin basic protein (MBP) expression, tumor necrosis factor alpha (TNF-a) expression, interleukin 1 beta (IL1B) expression, mortality rate, neurological function, and complement component 3 (C3) mRNA expression were measured by techniques such as Evans blue fluorescence, Perls staining, TUNEL staining, ELISA, immunohistochemistry and Western blotting to evaluate correlations between complement activation and secondary injury. RESULTS The activation of complement after TBI was significantly higher at high altitude than at low altitude. High-altitude TBI resulted in a leakier blood-brain barrier, more severe cerebral edema and higher mortality than low-altitude TBI did. In addition, high-altitude TBI tended to be associated with more MBP degradation, ferric iron deposition, neuronal apoptosis, and inflammatory factor deposition than low-altitude TBI. All of these effects of TBI were partially reversed by inhibiting complement activation using CVF. CONCLUSION Our study provided evidence that TBI at high altitude leads to severe edema and high mortality and disability rates. Complement C3 activation is one of the important factors contributing to secondary brain injury.
Collapse
Affiliation(s)
- Linjie Wei
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Jianbo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Bo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Junjun Geng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Ling Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
32
|
Alawieh A, Langley EF, Tomlinson S. Targeted complement inhibition salvages stressed neurons and inhibits neuroinflammation after stroke in mice. Sci Transl Med 2019; 10:10/441/eaao6459. [PMID: 29769288 DOI: 10.1126/scitranslmed.aao6459] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/22/2017] [Accepted: 04/27/2018] [Indexed: 12/20/2022]
Abstract
Ischemic stroke results from the interruption of blood flow to the brain resulting in long-term motor and cognitive neurological deficits, and it is a leading cause of death and disability. Current interventions focus on the restoration of blood flow to limit neuronal death, but these treatments have a therapeutic window of only a few hours and do not address post-stroke cerebral inflammation. The complement system, a component of the innate immune system, is activated by natural immunoglobulin M (IgM) antibodies that recognize neoepitopes expressed in the brain after ischemic stroke. We took advantage of this recognition system to inhibit complement activation locally in the ischemic area in mice. A single chain antibody recognizing a post-ischemic neoepitope linked to a complement inhibitor (termed B4Crry) was administered systemically as a single dose after stroke and shown to specifically target the ischemic hemisphere and improve long-term motor and cognitive recovery. We show that complement opsonins guide microglial phagocytosis of stressed but salvageable neurons, and that by locally and transiently inhibiting complement deposition, B4Crry prevented phagocytosis of penumbral neurons and inhibited pathologic complement and microglial activation that otherwise persisted for several weeks after stroke. B4Crry was protective in adult, aged, male and female mice and had a therapeutic window of at least 24 hours after stroke. Furthermore, the epitope recognized by B4Crry in mice is overexpressed in the ischemic penumbra of acute stroke patients, but not in the contralateral tissue, highlighting the translational potential of this approach.
Collapse
Affiliation(s)
- Ali Alawieh
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA.,Medical Scientist Training Program, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - E Farris Langley
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA. .,Ralph H. Johnson VA Medical Center, Charleston, SC 29425, USA
| |
Collapse
|
33
|
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
|
34
|
De Blasio D, Fumagalli S, Orsini F, Neglia L, Perego C, Ortolano F, Zanier ER, Picetti E, Locatelli M, Stocchetti N, Longhi L, Garred P, De Simoni MG. Human brain trauma severity is associated with lectin complement pathway activation. J Cereb Blood Flow Metab 2019; 39:794-807. [PMID: 29425056 PMCID: PMC6501516 DOI: 10.1177/0271678x18758881] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/03/2018] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
We explored the involvement of the lectin pathway of complement in post-traumatic brain injury (TBI) pathophysiology in humans. Brain samples were obtained from 28 patients who had undergone therapeutic contusion removal, within 12 h (early) or from >12 h until five days (late) from injury, and from five non-TBI patients. Imaging analysis indicated that lectin pathway initiator molecules (MBL, ficolin-1, ficolin-2 and ficolin-3), the key enzymes MASP-2 and MASP-3, and the downstream complement components (C3 fragments and TCC) were present inside and outside brain vessels in all contusions. Only ficolin-1 was found in the parenchyma of non-TBI tissues. Immunoassays in brain homogenates showed that MBL, ficolin-2 and ficolin-3 increased in TBI compared to non-TBI (2.0, 2.2 and 6.0-times) samples. MASP-2 increased with subarachnoid hemorrhage and abnormal pupil reactivity, two indicators of structural and functional damage. C3 fragments and TCC increased, respectively, by 3.5 - and 4.0-fold in TBI compared to non-TBI tissue and significantly correlated with MBL, ficolin-2, ficolin-3, MASP-2 and MASP-3 levels in the homogenates. In conclusion, we show for the first time the direct presence of lectin pathway components in human cerebral contusions and their association with injury severity, suggesting a central role for the lectin pathway in the post-traumatic pathophysiology of human TBI.
Collapse
Affiliation(s)
- Daiana De Blasio
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Stefano Fumagalli
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Franca Orsini
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Laura Neglia
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Carlo Perego
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Fabrizio Ortolano
- Department of Anesthesia and Critical
Care Medicine, Fondazione IRCCS Ca' Granda- Ospedale Maggiore Policlinico, Milano,
Italy
| | - Elisa R Zanier
- IRCCS-Istituto di Ricerche
Farmacologiche Mario Negri, Milano, Italy
| | - Edoardo Picetti
- Division of Anesthesia and Intensive
Care, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Marco Locatelli
- Department of Neurosurgery, Fondazione
IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milano, Italy
| | - Nino Stocchetti
- Department of Anesthesia and Critical
Care Medicine, Fondazione IRCCS Ca' Granda- Ospedale Maggiore Policlinico, Milano,
Italy
- Department of Physiopathology and
Transplantation, Milan University, Milan, Italy
| | - Luca Longhi
- Department of Anesthesia and Critical
Care Medicine, Neurosurgical Intensive Care Unit, Azienda Socio Sanitaria
Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Peter Garred
- Laboratory of Molecular Medicine,
Department of Clinical Immunology, Rigshospitalet Faculty of Medical and Health
Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
35
|
Brennan FH, Popovich PG. Emerging targets for reprograming the immune response to promote repair and recovery of function after spinal cord injury. Curr Opin Neurol 2019; 31:334-344. [PMID: 29465433 DOI: 10.1097/wco.0000000000000550] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW In adult mammals, a traumatic spinal cord injury (SCI) elicits a chronic unregulated neuroinflammatory response accompanied by seemingly paradoxical suppression of systemic immunity. These SCI-induced changes in immune function contribute to poor neurological outcomes and enhanced morbidity or mortality. Nonspecific anti-inflammatory or proinflammatory therapies are ineffective and can even worsen outcomes. Therefore, recent experimental SCI research has advanced the understanding of how neuroimmune cross-talk contributes to spinal cord and systemic pathology. RECENT FINDINGS It is now appreciated that the immune response caused by injury to the brain or spinal cord encompasses heterogeneous elements that can drive events on the spectrum between exacerbating pathology and promoting tissue repair, within the spinal cord and throughout the body. Recent novel discoveries regarding the role and regulation of soluble factors, monocytes/macrophages, microRNAs, lymphocytes and systemic immune function are highlighted in this review. SUMMARY A more nuanced understanding of how the immune system responds and reacts to nervous system injury will present an array of novel therapeutic opportunities for clinical SCI and other forms of neurotrauma.
Collapse
Affiliation(s)
- Faith H Brennan
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | |
Collapse
|
36
|
Warwick CA, Shutov LP, Shepherd AJ, Mohapatra DP, Usachev YM. Mechanisms underlying mechanical sensitization induced by complement C5a: the roles of macrophages, TRPV1, and calcitonin gene-related peptide receptors. Pain 2019; 160:702-711. [PMID: 30507785 PMCID: PMC6377341 DOI: 10.1097/j.pain.0000000000001449] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The complement system significantly contributes to the development of inflammatory and neuropathic pain, but the underlying mechanisms are poorly understood. Recently, we identified the signaling pathway responsible for thermal hypersensitivity induced by the complement system component C5a. Here, we examine the mechanisms of another important action of C5a, induction of mechanical hypersensitivity. We found that intraplantar injection of C5a produced a dose-dependent mechanical sensitization and that this effect was blocked by chemogenetic ablation of macrophages in both male and female mice. Knockout of TRPV1 or pretreatment with the TRPV1 antagonists, AMG9810 or 5'-iodoresiniferatoxin (5'-IRTX), significantly reduced C5a-induced mechanical sensitization. Notably, local administration of 5'-IRTX 90 minutes after C5a injection resulted in a slow, but complete, reversal of mechanical sensitization, indicating that TRPV1 activity was required for maintaining C5a-induced mechanical hypersensitivity. This slow reversal suggests that neurogenic inflammation and neuropeptide release may be involved. Indeed, pretreatment with a calcitonin gene-related peptide (CGRP) receptor antagonist (but not an antagonist of the neurokinin 1 receptor) prevented C5a-induced mechanical sensitization. Furthermore, intraplantar injection of CGRP produced significant mechanical sensitization in both wild-type and TRPV1 knockout mice. Taken together, these findings suggest that C5a produces mechanical sensitization by initiating macrophage-to-sensory-neuron signaling cascade that involves activation of TRPV1 and CGRP receptor as critical steps in this process.
Collapse
Affiliation(s)
- Charles A. Warwick
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| | - Leonid P. Shutov
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| | - Andrew J. Shepherd
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Durga P. Mohapatra
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Yuriy M. Usachev
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
37
|
Chen H, Zheng J, Ma J. Vanillin ameliorates changes in HIF-1α expression and neuronal apoptosis in a rat model of spinal cord injury. Restor Neurol Neurosci 2019; 37:21-29. [PMID: 30741707 DOI: 10.3233/rnn-180879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In the search for treating neurological dysfunctions after spinal cord injury (SCI), methods of neuroprotection are of interest to intervene with the caspase pathway. OBJECTIVE To evaluate the neuroprotective effects of vanillin in a rat model of spinal cord injury (SCI). METHODS Rats were randomly assigned to one of three groups: a sham-operated group, and two groups where SCI was produced by ischemia/reperfusion which received either saline or vanillin (286 mg/kg, intraperitoneal [i.p.] 30 min prior to surgery). Neurological function was estimated by the Tarlov scale at 1, 12, and 24 h after surgery. Additionally, we estimated the levels of oxidative stress, inflammatory cytokines, and mitochondrial proteins in the homogenates of spinal tissues and terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) and immunohistochemical assays of spinal tissues. RESULTS Motor dysfunction was found to be significantly improved in the vanillin treated group compared to SCI rats. This was accompanied by altered levels of oxidative stress, inflammatory cytokines, and expressions of mitochondrial proteins in the SCI rats which were ameliorated by the vanillin treatment. Vanillin also significantly reduced the number of TUNEL-positive cells in spinal cord tissues compared to the sham group (p < 0.01) and decreased the number of hypoxia-inducible factor (HIF)-1α-positive cells. CONCLUSIONS In the SCI rat model vanillin exerted neuroprotective effects of reducing apoptosis and attenuating the expression of HIF-1α in spinal tissues.
Collapse
Affiliation(s)
- Hailong Chen
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Luoyang, Henan, China
| | - Jiuqin Zheng
- Department of Central Sterile Supply, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Luoyang, Henan, China
| | - Junjie Ma
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Luoyang, Henan, China
| |
Collapse
|
38
|
Younger D, Murugan M, Rama Rao KV, Wu LJ, Chandra N. Microglia Receptors in Animal Models of Traumatic Brain Injury. Mol Neurobiol 2018; 56:5202-5228. [PMID: 30554385 DOI: 10.1007/s12035-018-1428-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
39
|
Li Y, Zhu ZY, Huang TT, Zhou YX, Wang X, Yang LQ, Chen ZA, Yu WF, Li PY. The peripheral immune response after stroke-A double edge sword for blood-brain barrier integrity. CNS Neurosci Ther 2018; 24:1115-1128. [PMID: 30387323 PMCID: PMC6490160 DOI: 10.1111/cns.13081] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023] Open
Abstract
The blood‐brain barrier (BBB) is a highly regulated interface that separates the peripheral circulation and the brain. It plays a vital role in regulating the trafficking of solutes, fluid, and cells at the blood‐brain interface and maintaining the homeostasis of brain microenvironment for normal neuronal activity. Growing evidence has led to the realization that ischemic stroke elicits profound immune responses in the circulation and the activation of multiple subsets of immune cells, which in turn affect both the early disruption and the later repair of the BBB after stroke. Distinct phenotypes or subsets of peripheral immune cells along with diverse intracellular mechanisms contribute to the dynamic changes of BBB integrity after stroke. This review focuses on the interaction between the peripheral immune cells and the BBB after ischemic stroke. Understanding their reciprocal interaction may generate new directions for stroke research and may also drive the innovation of easy accessible immune modulatory treatment strategies targeting BBB in the pursuit of better stroke recovery.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ting-Ting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu-Xi Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zeng-Ai Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
40
|
Habib A, Sawmiller D, Hou H, Kanithi M, Tian J, Zeng J, Zi D, He ZX, Sanberg PR, Tan J. Human Cord Blood Serum-Derived APP α-Secretase Cleavage Activity is Mediated by C1 Complement. Cell Transplant 2018; 27:666-676. [PMID: 29871524 PMCID: PMC7020233 DOI: 10.1177/0963689718775941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's Disease (AD) is the leading cause of dementia in the elderly. In healthy individuals, amyloid precursor protein (APP) is cleaved by α-secretase, generating soluble α-amyloid precursor protein (sAPPα), which contributes neuroprotective functions in the neuronal environment. In contrast, in the neurodegenerative environment of AD patients, amyloid-β-peptide (Aβ) of either 40 or 42 residues are generated by increased activity of β- and γ-secretase. These proteins amalgamate in specific regions of the brain, which disrupts neuronal functions and leads to cognitive impairment. Human umbilical cord blood cells (HUCBC) have proven useful as potential immunomodulatory therapies in various models of neurodegenerative diseases, including AD. Our most recent work studied the impact of umbilical cord blood serum (CBS) on modulation of sAPPα production. Heat-sensitive CBS significantly promoted sAPPα production, indicating that heat-sensitive factor(s) play(s) a role in this process. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to determine the molecular source of α-secretase in purified CBS and aged blood serum (AgBS) fraction. Of the proteins identified, the subunits of C1 complex (C1q, C1r, and C1s) and alpha-2-macroglobulin showed significantly greater levels in purified α-CBS fraction (α-CBSF) compared with the AgBS fraction (AgBSF). Specifically, C1 markedly increased sAPPα and alpha-carboxyl-terminal fragment (α-CTF) production in a dose-dependent fashion, whereas C1q alone only minimally increased and C3 did not increase sAPPα production in the absence of sera. Furthermore, C1q markedly increased sAPPα and α-CTF, while decreasing Aβ, in CHO/APPwt cells cultured in the presence of whole sera. These results confirm our initial assumption that APP α-secretase activity in human blood serum is mediated by complement C1, opening a potential therapeutic modality for the future of AD.
Collapse
Affiliation(s)
- Ahsan Habib
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Darrell Sawmiller
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Huayan Hou
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Manasa Kanithi
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Tian
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jin Zeng
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Dan Zi
- 2 Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center & Sino-US Joint Laboratory for Medical Sciences, Laboratory Animal Center, Guizhou Medical University Guiyang, China
| | - Zhi-Xu He
- 2 Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center & Sino-US Joint Laboratory for Medical Sciences, Laboratory Animal Center, Guizhou Medical University Guiyang, China
| | - Paul R Sanberg
- 3 Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Tan
- 1 Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
41
|
Bao W, He F, Yu L, Gao J, Meng F, Ding Y, Zou H, Luo B. Complement cascade on severe traumatic brain injury patients at the chronic unconscious stage: implication for pathogenesis. Expert Rev Mol Diagn 2018; 18:761-766. [PMID: 29718755 DOI: 10.1080/14737159.2018.1471985] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Patients who awake from severely traumatic brain injury (TBI) may remain unconscious for many years. Although behavioral assessment and functional imaging are currently used as diagnostic tools, the molecular basis underlying chronic condition has yet to be explored. METHOD Plasma samples were obtained at 3 time points (1, 3 and 6 months) from 18 patients with chronic disorders of consciousness who survived severe TBI, and 6 healthy volunteers. A coupled isobaric tag for relative and absolute quantitation (iTRAQ)-based proteomics approach was used to screen differentially expressed proteins (DEPs) between patients and controls. Potential molecular mechanisms were further discussed through bioinformatics analyses. RESULT In total, 300 plasma proteins <1% false discovery rates were identified and 32 proteins were consistently altered between patients and controls. Biological pathway analysis revealed that the DEPs were predominantly involved in complement cascade. CONCLUSIONS This study discussed potential mechanisms of complement cascade underlying chronic stage in severe TBI.
Collapse
Affiliation(s)
- Wangxiao Bao
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| | - Fangping He
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| | - Lihua Yu
- b Department of Neurology , Zhejiang Provincial People's Hospital , Hangzhou , China.,e People's Hospital of Hangzhou Medical College , Hangzhou Zhejiang Province , China
| | - Jian Gao
- c Department of Rehabilitation , Hangzhou Hospital of Zhejiang CAPR , Hangzhou , China
| | - Fanxia Meng
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| | - Yahui Ding
- d Department of Neurology, First Affiliated Hospital , Zhejiang Provincial People's Hospital , Hangzhou , China.,e People's Hospital of Hangzhou Medical College , Hangzhou Zhejiang Province , China
| | - Hai Zou
- d Department of Neurology, First Affiliated Hospital , Zhejiang Provincial People's Hospital , Hangzhou , China.,e People's Hospital of Hangzhou Medical College , Hangzhou Zhejiang Province , China
| | - Benyan Luo
- a Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science , Zhejiang University School of Medicine , Hangzhou , China
| |
Collapse
|
42
|
Ye W, Chen S, Huang X, Qin W, Zhang T, Zhu X, Zhu X, Lin C, Wang X. Clinical features and risk factors of neurological involvement in Sjögren's syndrome. BMC Neurosci 2018; 19:26. [PMID: 29703151 PMCID: PMC5924492 DOI: 10.1186/s12868-018-0427-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 04/19/2018] [Indexed: 12/14/2022] Open
Abstract
Background To investigated distinct manifestations of Sjögren’s syndrome (SS) patients with neurological complications and the potential risk factors associated with neurological complications in SS, and to produce a disease evaluation and neurological involvement prediction for SS. Methods 566 patients who fulfilled the 2002 classification criteria for SS from the Rheumatology Department of the First Affiliated Hospital of Wenzhou Medical University were included in the cross-sectional study. Clinical, immunological and histological characteristics were surveyed, and potential risk factors for neurological complications were examined by multivariate analysis. Results Among 566 SS patients, 184 (32.5%) patients had neurological involvement, with more than 10% got limbs pain, limbs numbness and cerebral infarction, respectively. Of these 184 SS patients with neurological complications, secondary SS (sSS) patients had a higher prevalence of peripheral nervous system (PNS) involvement than primary SS (pSS) patients (31.1 vs. 19%). And sSS patients showed higher total ESSPRI score and higher prevalence of xerostomia and low C3, C4 levels with more liver, articular involvement and saliva gland atrophy, and more severe lymphocyte infiltration in salivary glands than pSS patients. As for the specific factors associated with neurological involvement, low C3 level were found to be significant in pSS or sSS patients who were younger 50 year old, and ANA positivity, cardiac involvement, saliva gland atrophy were demonstrated to be associated in elder pSS patients. And xerophthalmia was found to be associated in sSS patients. Conclusion Low complement (C3) levels, xerophthalmia, ANA positive, cardiac involvement and labial salivary gland histological result were good ways to predict neurological complications in different subgroups of SS, which might provide insight into better clinical decision-making, especially at early stages of the disease.
Collapse
Affiliation(s)
- Wenjing Ye
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Rheumatology Department, Ruian People's Hospital, Wenzhou, China
| | - Siyan Chen
- Neurology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinshi Huang
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Qin
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ting Zhang
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaofang Zhu
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaochun Zhu
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chongxiang Lin
- Stomatological Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xiaobing Wang
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
43
|
Induced neural stem cell-derived astrocytes modulate complement activation and mediate neuroprotection following closed head injury. Cell Death Dis 2018; 9:101. [PMID: 29367701 PMCID: PMC5833559 DOI: 10.1038/s41419-017-0172-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/25/2017] [Accepted: 11/22/2017] [Indexed: 11/09/2022]
Abstract
The complement system is a crucial component of immunity, and its activation has critical roles in neuroinflammatory response and cellular damage following closed head injury (CHI). We previously demonstrated that systemically injected induced neural stem cells (iNSCs) could modulate complement activation to ameliorate neuronal apoptosis in mouse CHI models. However, it remains unknown whether iNSC derivatives can regulate complement activation. In the present study, after CHI mouse serum treatment, we found dramatic decreases in the cellular viabilities of differentiated iNSCs. Interestingly, following CHI mouse serum treatment, the death of astrocytes derived from iNSCs which were pre-treated with CHI mouse serum was significantly decreased. Meanwhile, the deposition of C3 (C3d) and C5b-9 in these astrocytes was substantially reduced. Remarkably, we detected increased expression of complement receptor type 1-related protein y (Crry) in these astrocytes. Moreover, these astrocytes could reduce the numbers of apoptotic neurons via Crry expression post-CHI mouse serum treatment. Additionally, intracerebral-transplanted iNSCs, pre-treated with CHI mouse serum, significantly increased the levels of Crry expression in astrocytes to reduce the accumulation of C3d and C9 and the death of neurons in the brains of CHI mice. In summary, iNSCs receiving CHI mouse serum pre-treatment could enhance the expression of Crry in iNSC-derived astrocytes to modulate complement activation and mediate neuroprotection following CHI.
Collapse
|
44
|
Hammad A, Westacott L, Zaben M. The role of the complement system in traumatic brain injury: a review. J Neuroinflammation 2018; 15:24. [PMID: 29357880 PMCID: PMC5778697 DOI: 10.1186/s12974-018-1066-z] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of disability and mortality in the western world. While the initial injury sustained results in damage, it is the subsequent secondary cascade that is thought to be the significant determinant of subsequent outcomes. The changes associated with the secondary injury do not become irreversible until some time after the start of the cascade. This may present a window of opportunity for therapeutic interventions aiming to improve outcomes subsequent to TBI. A prominent contributor to the secondary injury is a multifaceted inflammatory reaction. The complement system plays a notable role in this inflammatory reaction; however, it has often been overlooked in the context of TBI secondary injury. The complement system has homeostatic functions in the uninjured central nervous system (CNS), playing a part in neurodevelopment as well as having protective functions in the fully developed CNS, including protection from infection and inflammation. In the context of CNS injury, it can have a number of deleterious effects, evidence for which primarily comes not only from animal models but also, to a lesser extent, from human post-mortem studies. In stark contrast to this, complement may also promote neurogenesis and plasticity subsequent to CNS injury. This review aims to explore the role of the complement system in TBI secondary injury, by examining evidence from both clinical and animal studies. We examine whether specific complement activation pathways play more prominent roles in TBI than others. We also explore the potential role of complement in post-TBI neuroprotection and CNS repair/regeneration. Finally, we highlight the therapeutic potential of targeting the complement system in the context of TBI and point out certain areas on which future research is needed.
Collapse
Affiliation(s)
- Adnan Hammad
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Laura Westacott
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Room 4FT 80E, 4th Floor, Heath Park, Cardiff, CF14 4XN UK
| | - Malik Zaben
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Room 4FT 80E, 4th Floor, Heath Park, Cardiff, CF14 4XN UK
| |
Collapse
|
45
|
Xiao J, Zhang J, Zhao Y, Huang W, Guo Z, Su B, Guo Q. Sex differences of steroid receptor coactivator-1 expression after spinal cord injury in mice. Neurol Res 2017; 39:1022-1027. [DOI: 10.1080/01616412.2017.1367077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jiayu Xiao
- Student Brigade, Third Military Medical University, Chongqing, China
| | - Jiqiang Zhang
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Yangang Zhao
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Wenjie Huang
- Student Brigade, Third Military Medical University, Chongqing, China
| | - Zhikai Guo
- Student Brigade, Third Military Medical University, Chongqing, China
| | - Bingyin Su
- Development and Regeneration Key Lab of Sichuan Province, Department of Anatomy and Histology and Embryology, Department of Pathology, Chengdu Medical College, Chengdu, China
| | - Qiang Guo
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Third Military Medical University, Chongqing, China
| |
Collapse
|
46
|
The Complement System Component C5a Produces Thermal Hyperalgesia via Macrophage-to-Nociceptor Signaling That Requires NGF and TRPV1. J Neurosci 2017; 36:5055-70. [PMID: 27147658 DOI: 10.1523/jneurosci.3249-15.2016] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The complement cascade is a principal component of innate immunity. Recent studies have underscored the importance of C5a and other components of the complement system in inflammatory and neuropathic pain, although the underlying mechanisms are largely unknown. In particular, it is unclear how the complement system communicates with nociceptors and which ion channels and receptors are involved. Here we demonstrate that inflammatory thermal and mechanical hyperalgesia induced by complete Freund's adjuvant was accompanied by C5a upregulation and was markedly reduced by C5a receptor (C5aR1) knock-out or treatment with the C5aR1 antagonist PMX53. Direct administration of C5a into the mouse hindpaw produced strong thermal hyperalgesia, an effect that was absent in TRPV1 knock-out mice, and was blocked by the TRPV1 antagonist AMG9810. Immunohistochemistry of mouse plantar skin showed prominent expression of C5aR1 in macrophages. Additionally, C5a evoked strong Ca(2+) mobilization in macrophages. Macrophage depletion in transgenic macrophage Fas-induced apoptosis mice abolished C5a-dependent thermal hyperalgesia. Examination of inflammatory mediators following C5a injection revealed a rapid upregulation of NGF, a mediator known to sensitize TRPV1. Preinjection of an NGF-neutralizing antibody or Trk inhibitor GNF-5837 prevented C5a-induced thermal hyperalgesia. Notably, NGF-induced thermal hyperalgesia was unaffected by macrophage depletion. Collectively, these results suggest that complement fragment C5a induces thermal hyperalgesia by triggering macrophage-dependent signaling that involves mobilization of NGF and NGF-dependent sensitization of TRPV1. Our findings highlight the importance of macrophage-to-neuron signaling in pain processing and identify C5a, NGF, and TRPV1 as key players in this cross-cellular communication. SIGNIFICANCE STATEMENT This study provides mechanistic insight into how the complement system, a key component of innate immunity, regulates the development of pain hypersensitivity. We demonstrate a crucial role of the C5a receptor, C5aR1, in the development of inflammatory thermal and mechanical sensitization. By focusing on the mechanisms of C5a-induced thermal hyperalgesia, we show that this process requires recruitment of macrophages and initiation of macrophage-to-nociceptor signaling. At the molecular level, we demonstrate that this signaling depends on NGF and is mediated by the heat-sensitive nociceptive channel TRPV1. This deeper understanding of how immune cells and neurons interact to regulate pain processing is expected to facilitate mechanism-based approaches in the development of new analgesics.
Collapse
|
47
|
Osthoff M, Walder B, Delhumeau C, Trendelenburg M, Turck N. Association of Lectin Pathway Protein Levels and Genetic Variants Early after Injury with Outcomes after Severe Traumatic Brain Injury: A Prospective Cohort Study. J Neurotrauma 2017; 34:2560-2566. [PMID: 28482760 DOI: 10.1089/neu.2016.4941] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The lectin pathway of the complement system has been implicated in secondary ischemic/inflammatory injury after traumatic brain injury (TBI). However, previous experimental studies have yielded conflicting results, and human studies are scarce. In this exploratory study, we investigated associations of several lectin pathway proteins early after injury and single-nucleotide polymorphisms (SNP) with outcomes after severe TBI (mortality at 14 days [primary outcome] and consciousness assessed with the Glasgow Coma Scale [GCS] at 14 days, disability assessed with the Glasgow Outcome Scale Extended [GOSE] at 90 days). Forty-four patients with severe TBI were included. Plasma levels of lectin pathway proteins were sampled at 6, 12, 24, and 48 h after injury and eight mannose-binding lectin (MBL) and ficolin (FCN)2 SNPs were analyzed by enzyme-linked immunosorbent assay (ELISA) and genotyping, respectively. Plasma protein levels were stable with only a slight increase in mannose-binding protein-associated serine protease (MASP)-2 and FCN2 levels after 48 h (p < 0.05), respectively. Neither lectin protein plasma levels (6 h or mean levels) nor MBL2 genotypes or FCN2 variant alleles were associated with 14 day mortality or 14 day consciousness. However, FCN2, FCN3, and MASP-2 levels were higher in patients with an unfavorable outcome (GOSE 1-4) at 90 days (p < 0.05), whereas there was no difference in MBL2 genotypes or FCN2 variant alleles. In particular, higher mean MASP-2 levels over 48 h were independently associated with a GOSE score < 4 at 90 days after adjustment (odds ratio 3.46 [95% confidence interval 1.12-10.68] per 100 ng/mL increase, p = 0.03). No association was observed between the lectin pathway of the complement system and 14 day mortality or 14 day consciousness. However, higher plasma FCN2, FCN3, and, in particular, MASP-2 levels early after injury were associated with an unfavorable outcome at 90 days (death, vegetative state, and severe disability) which may be related to an increased activation of the lectin pathway.
Collapse
Affiliation(s)
- Michael Osthoff
- 1 Division of Internal Medicine, University Hospital Basel , Basel, Switzerland .,2 Department of Biomedicine, University Hospital Basel , Basel, Switzerland
| | - Bernhard Walder
- 3 Division of Anaesthesiology, Department of Anaesthesiology, Intensive Care and Clinical Pharmacology, University Hospitals of Geneva , Geneva, Switzerland
| | - Cécile Delhumeau
- 3 Division of Anaesthesiology, Department of Anaesthesiology, Intensive Care and Clinical Pharmacology, University Hospitals of Geneva , Geneva, Switzerland
| | - Marten Trendelenburg
- 1 Division of Internal Medicine, University Hospital Basel , Basel, Switzerland .,2 Department of Biomedicine, University Hospital Basel , Basel, Switzerland
| | - Natacha Turck
- 4 OPTICS Group, Department of Human Protein Sciences, University of Geneva , Geneva, Switzerland
| |
Collapse
|
48
|
Recent progress in the understanding of complement activation and its role in tumor growth and anti-tumor therapy. Biomed Pharmacother 2017; 91:446-456. [DOI: 10.1016/j.biopha.2017.04.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/18/2017] [Accepted: 04/23/2017] [Indexed: 02/07/2023] Open
|
49
|
Galbavy W, Lu Y, Kaczocha M, Puopolo M, Liu L, Rebecchi MJ. Transcriptomic evidence of a para-inflammatory state in the middle aged lumbar spinal cord. IMMUNITY & AGEING 2017; 14:9. [PMID: 28413428 PMCID: PMC5390443 DOI: 10.1186/s12979-017-0091-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/05/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND We have previously reported elevated expression of multiple pro-inflammatory markers in the lumbar spinal cord (LSC) of middle-aged male rats compared to young adults suggesting a para-inflammatory state develops in the LSC by middle age, a time that in humans is associated with the greatest pain prevalence and persistence. The goal of the current study was to examine the transcriptome-wide gene expression differences between young and middle aged LSC. METHODS Young (3 month) and middle-aged (17 month) naïve Fisher 344 rats (n = 5 per group) were euthanized, perfused with heparinized saline, and the LSC were removed. RESULTS ~70% of 31,000 coding sequences were detected. After normalization, ~ 1100 showed statistically significant differential expression. Of these genes, 353 middle-aged annotated genes differed by > 1.5 fold compared to the young group. Nearly 10% of these genes belonged to the microglial sensome. Analysis of this subset revealed that the principal age-related differential pathways populated are complement, pattern recognition receptors, OX40, and various T cell regulatory pathways consistent with microglial priming and T cell invasion and modulation. Many of these pathways substantially overlap those previously identified in studies of LSC of young animals with chronic inflammatory or neuropathic pain. CONCLUSIONS Up-modulation of complement pathway, microglial priming and activation, and T cell/antigen-presenting cell communication in healthy middle-aged LSC was found. Taken together with our previous work, the results support our conclusion that an incipient or para-inflammatory state develops in the LSC in healthy middle-aged adults.
Collapse
Affiliation(s)
- William Galbavy
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Yong Lu
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Martin Kaczocha
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Michelino Puopolo
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Lixin Liu
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| | - Mario J Rebecchi
- Department of Anesthesiology, School of Medicine, Health Sciences Center L4, Stony Brook University, Stony Brook, New York, 11794-8480 USA
| |
Collapse
|
50
|
Systemic Administration of Induced Neural Stem Cells Regulates Complement Activation in Mouse Closed Head Injury Models. Sci Rep 2017; 7:45989. [PMID: 28383046 PMCID: PMC5382667 DOI: 10.1038/srep45989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/07/2017] [Indexed: 02/06/2023] Open
Abstract
Complement activation plays important roles in the pathogenesis of central nervous system (CNS) diseases. Patients face neurological disorders due to the development of complement activation, which contributes to cell apoptosis, brain edema, blood-brain barrier dysfunction and inflammatory infiltration. We previously reported that induced neural stem cells (iNSCs) can promote neurological functional recovery in closed head injury (CHI) animals. Remarkably, we discovered that local iNSC grafts have the potential to modulate CNS inflammation post-CHI. In this study, we aimed to explore the role of systemically delivered iNSCs in complement activation following CNS injury. Our data showed that iNSC grafts decreased the levels of sera C3a and C5a and down-regulated the expression of C3d, C9, active Caspase-3 and Bax in the brain, kidney and lung tissues of CHI mice. Furthermore, iNSC grafts decreased the levels of C3d+/NeuN+, C5b-9+/NeuN+, C3d+/Map2+ and C5b-9+/Map2+ neurons in the injured cortices of CHI mice. Subsequently, we explored the mechanisms underlying these effects. With flow cytometry analysis, we observed a dramatic increase in complement receptor type 1-related protein y (Crry) expression in iNSCs after CHI mouse serum treatment. Moreover, both in vitro and in vivo loss-of-function studies revealed that iNSCs could modulate complement activation via Crry expression.
Collapse
|