1
|
Koehn LM, Steele JR, Schittenhelm RB, Nicolazzo JA. Sex-Specific Markers of Neuroinflammation and Neurodegeneration in the Spinal Cord Proteome of the SOD1 G93A Mouse Model of Amyotrophic Lateral Sclerosis. J Proteome Res 2025; 24:1956-1970. [PMID: 40117341 DOI: 10.1021/acs.jproteome.4c00990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that has no cure. The underlying mechanistic details of sex differences in the ALS spinal cord, the site of disease onset, are not understood to an extent that could guide novel drug development. To address this, the spinal cords of 120-day-old wild-type (WT) and SOD1G93A (familial mouse model of ALS with mutant superoxide dismutase 1) mice were subjected to untargeted, quantitative proteomics using tandem mass tag acquisition on high-resolution mass spectrometric instrumentation. Compared to WT, both male and female SOD1G93A spinal cords exhibited an upregulation of neuroinflammatory cascades of both peripheral and central origins, as well as a downregulation of proteins reflective of death and dysfunction of cells within the spinal cord. However, female and male SOD1G93A mouse spinal cords exhibited sex-specific differences in proteins compared to respective WT that related to immune response, as well as cellular structure, function, and homeostasis. The proteomic datasets presented provide entire cohort and sex-specific spinal cord drug targets and disease biomarkers in the SOD1G93A mouse model of ALS that may guide future drug development and sex selection in preclinical study designs utilizing the SOD1G93A model.
Collapse
Affiliation(s)
- Liam M Koehn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3168, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3168, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
2
|
Faller KME, Chaytow H, Gillingwater TH. Targeting common disease pathomechanisms to treat amyotrophic lateral sclerosis. Nat Rev Neurol 2025; 21:86-102. [PMID: 39743546 DOI: 10.1038/s41582-024-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
The motor neuron disease amyotrophic lateral sclerosis (ALS) is a devastating condition with limited treatment options. The past few years have witnessed a ramping up of translational ALS research, offering the prospect of disease-modifying therapies. Although breakthroughs using gene-targeted approaches have shown potential to treat patients with specific disease-causing mutations, the applicability of such therapies remains restricted to a minority of individuals. Therapies targeting more general mechanisms that underlie motor neuron pathology in ALS are therefore of considerable interest. ALS pathology is associated with disruption to a complex array of key cellular pathways, including RNA processing, proteostasis, metabolism and inflammation. This Review details attempts to restore cellular homeostasis by targeting these pathways in order to develop effective, broadly-applicable ALS therapeutics.
Collapse
Affiliation(s)
- Kiterie M E Faller
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Helena Chaytow
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Vecchiarelli HA, Lopes LT, Paolicelli RC, Stevens B, Wake H, Tremblay MÈ. Synapse Regulation. ADVANCES IN NEUROBIOLOGY 2024; 37:179-208. [PMID: 39207693 DOI: 10.1007/978-3-031-55529-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are the resident immune cells of the brain. As such, they rapidly detect changes in normal brain homeostasis and accurately respond by fine-tuning in a tightly regulated manner their morphology, gene expression, and functional behavior. Depending on the nature of these changes, microglia can thicken and retract their processes, proliferate and migrate, release numerous signaling factors and compounds influencing neuronal physiology (e.g., cytokines and trophic factors), in addition to secreting proteases able to transform the extracellular matrix, and phagocytosing various types of cellular debris, etc. Because microglia also transform rapidly (on a time scale of minutes) during experimental procedures, studying these very special cells requires methods that are specifically non-invasive. The development of such methods has provided unprecedented insights into the roles of microglia during normal physiological conditions. In particular, transcranial two-photon in vivo imaging revealed that presumably "resting" microglia continuously survey the brain parenchyma with their highly motile processes, in addition to modulating their structural and functional interactions with neuronal circuits along the changes in neuronal activity and behavioral experience occurring throughout the lifespan. In this chapter, we will describe how surveillant microglia interact with synaptic elements and modulate the number, maturation, function, and plasticity of synapses in the healthy developing, mature, and aging brain, with consequences on neuronal activity, learning and memory, and the behavioral outcome.
Collapse
Affiliation(s)
| | | | - Rosa C Paolicelli
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
| | - Beth Stevens
- Department of Neurology, Harvard Medical School, Center for Life Science, Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
| | - Hiroaki Wake
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji-cho, Okazaki, Japan
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
4
|
Stanley ER, Biundo F, Gökhan Ş, Chitu V. Differential regulation of microglial states by colony stimulating factors. Front Cell Neurosci 2023; 17:1275935. [PMID: 37964794 PMCID: PMC10642290 DOI: 10.3389/fncel.2023.1275935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
Recent studies have emphasized the role of microglia in the progression of many neurodegenerative diseases. The colony stimulating factors, CSF-1 (M-CSF), granulocyte-macrophage CSF (GM-CSF) and granulocyte CSF (G-CSF) regulate microglia through different cognate receptors. While the receptors for GM-CSF (GM-CSFR) and G-CSF (G-CSFR) are specific for their ligands, CSF-1 shares its receptor, the CSF-1 receptor-tyrosine kinase (CSF-1R), with interleukin-34 (IL-34). All four cytokines are expressed locally in the CNS. Activation of the CSF-1R in macrophages is anti-inflammatory. In contrast, the actions of GM-CSF and G-CSF elicit different activated states. We here review the roles of each of these cytokines in the CNS and how they contribute to the development of disease in a mouse model of CSF-1R-related leukodystrophy. Understanding their roles in this model may illuminate their contribution to the development or exacerbation of other neurodegenerative diseases.
Collapse
Affiliation(s)
- E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Şölen Gökhan
- Department of Neurology, Albert Einstein College of Medicine, Institute for Brain Disorders and Neural Regeneration, Bronx, NY, United States
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
5
|
Genge A, van den Berg LH, Frick G, Han S, Abikoff C, Simmons A, Lin Q, Patra K, Kupperman E, Berry JD. Efficacy and Safety of Ravulizumab, a Complement C5 Inhibitor, in Adults With Amyotrophic Lateral Sclerosis: A Randomized Clinical Trial. JAMA Neurol 2023; 80:1089-1097. [PMID: 37695623 PMCID: PMC10495927 DOI: 10.1001/jamaneurol.2023.2851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/02/2023] [Indexed: 09/12/2023]
Abstract
Importance Additional therapies for amyotrophic lateral sclerosis (ALS) are urgently needed. Immune-mediated complement activation may be involved in ALS pathogenesis as evidenced by the upregulation of terminal components; thus, complement inhibition could potentially slow progression. Objective To evaluate the safety and efficacy of the terminal complement C5 inhibitor ravulizumab in adults with ALS. Design, Setting, and Participants This double-blind, placebo-controlled, parallel-group, multinational, randomized, phase 3 clinical trial was conducted from March 30, 2020, to October 17, 2021, in 81 ALS specialty centers across 17 countries. A preplanned, unmasked, nonbinding interim futility analysis was conducted when 33% of participants had completed week 26, wherein a conditional power of less than 10% would halt the trial. A total of 478 individuals were screened, and 96 were excluded. Inclusion criteria were weight of 40 kg or more, fulfillment of the El Escorial diagnostic criteria, and a minimal prestudy Revised Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS-R) progression score of -0.3 points per month. Interventions Study treatment consisted of placebo or a weight-based dose of intravenous ravulizumab every 8 weeks until week 42. Participants could continue standard-of-care treatment. Main Outcomes and Measures The primary end point was change from baseline in ALSFRS-R score at week 50 based on the Combined Assessment of Function and Survival (CAFS). Results A total of 382 participants were randomly assigned 2:1 to receive ravulizumab (n = 255; mean [SD] age, 58.6 [10.6] years; 94 female [36.9%] and 161 male [63.1%]) or placebo (n = 127; mean [SD] age, 58.0 [11.0] years; 58 female [45.7%] and 69 male [54.3%]). The interim analysis showed that the observed mean change from baseline in ALSFRS-R at week 50 was -14.67 points (SE, 0.89 points; 95% CI, -16.42 to -12.91 points) for ravulizumab and -13.33 points (SE, 1.22 points; 95% CI, -15.72 to -10.93 points) for placebo, with no significant difference between the groups (mean [SE] difference, -1.34 [1.46] points; 95% CI, -4.21 to 1.53 points). Based on these data, the trial was terminated for futility. The primary analysis at week 50 showed no significant difference in CAFS between groups (mean [SE], 5.5 [10.8] points; 95% CI, -15.7 to 26.6 points; P = .61). Overall incidence rates for treatment-emergent adverse events were similar for ravulizumab (204 participants [80.0%]) and placebo (108 participants [85.0%]). Conclusions and Relevance This trial rapidly showed that terminal complement C5 inhibition with ravulizumab did not slow functional decline in participants with ALS and that the safety profiles of ravulizumab and placebo were similar. Highly effective, novel treatments are critically needed to slow functional decline and extend survival in patients with ALS. Trial Registration ClinicalTrials.gov Identifier: NCT04248465.
Collapse
Affiliation(s)
| | | | - Glen Frick
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
| | - Steve Han
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
- Now with Takeda Pharmaceuticals, Cambridge, Massachusetts
| | - Cori Abikoff
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
- Now with Takeda Pharmaceuticals, Cambridge, Massachusetts
| | - Adam Simmons
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
- Now with Alector, West Hartford, Connecticut
| | - Qun Lin
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
| | - Kaushik Patra
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
- Now with Ultragenyx, Lexington, Massachusetts
| | - Erik Kupperman
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts
- Now with Viridian Therapeutics, Waltham, Massachusetts
| | - James D. Berry
- Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Boston
| |
Collapse
|
6
|
Kioutchoukova IP, Foster D, Thakkar RN, Kurz HN, Lucke-Wold B. Amyotrophic Lateral Sclerosis: From Mechanisms to Current, Emerging, and Alternative Therapeutics. MED DISCOVERIES 2023; 2:1059. [PMID: 37799543 PMCID: PMC10552707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a severe neurodegenerative disease affecting the motor neurons. Although the etiology remains unknown, mutations in superoxide dismutase 1 have been observed in patients with familial ALS, resulting in increased calcium in the cells and leading to cell death. Additionally, studies in patients with the C9orf72 repeat expansion have shown lower age of onset, cognitive and behavioral impairments, and reduced survival. Accumulation of TDP-43 in the cytoplasm of neurons and glial cells caused by the loss of UBQLN2 has been shown to lead to mitotoxicity and proteasomal overload. Early diagnosis of ALS is necessary for the optimization of care between a patient's neurologist and interdisciplinary team members to ensure the best outcomes possible. Proper management between physical therapy, occupation therapy, and pharmaceutical medications can improve ALS symptoms, achieving the highest quality of life possible for the patient. The current therapeutic medication recommended for ALS is Riluzole, but new therapies are emerging. This paper analyzes mechanisms of injury and progression of ALS along while analyzing current, emerging, and alternative therapeutics targeting ALS.
Collapse
Affiliation(s)
| | - Devon Foster
- University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Rajvi N Thakkar
- University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Hayley N Kurz
- University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
7
|
Yang W, Liu LB, Liu FL, Wu YH, Zhen ZD, Fan DY, Sheng ZY, Song ZR, Chang JT, Zheng YT, An J, Wang PG. Single-cell RNA sequencing reveals the fragility of male spermatogenic cells to Zika virus-induced complement activation. Nat Commun 2023; 14:2476. [PMID: 37120617 PMCID: PMC10148584 DOI: 10.1038/s41467-023-38223-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/21/2023] [Indexed: 05/01/2023] Open
Abstract
Zika virus (ZIKV) is a potential threat to male reproductive health but the mechanisms underlying its influence on testes during ZIKV infection remain obscure. To address this question, we perform single-cell RNA sequencing using testes from ZIKV-infected mice. The results reveal the fragility of spermatogenic cells, especially spermatogonia, to ZIKV infection and show that the genes of the complement system are significantly upregulated mainly in infiltrated S100A4 + monocytes/macrophages. Complement activation and its contribution to testicular damage are validated by ELISA, RT‒qPCR and IFA and further verify in ZIKV-infected northern pigtailed macaques by RNA genome sequencing and IFA, suggesting that this might be the common response to ZIKV infection in primates. On this basis, we test the complement inhibitor C1INH and S100A4 inhibitors sulindac and niclosamide for their effects on testis protection. C1INH alleviates the pathological change in the testis but deteriorates ZIKV infection in general. In contrast, niclosamide effectively reduces S100A4 + monocyte/macrophage infiltration, inhibits complement activation, alleviates testicular damage, and rescues the fertility of male mice from ZIKV infection. This discovery therefore encourages male reproductive health protection during the next ZIKV epidemic.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Li-Bo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Feng-Liang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Yan-Hua Wu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zi-Da Zhen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zi-Yang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zheng-Ran Song
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100093, China.
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
8
|
Zhang W, Chen Y, Pei H. C1q and central nervous system disorders. Front Immunol 2023; 14:1145649. [PMID: 37033981 PMCID: PMC10076750 DOI: 10.3389/fimmu.2023.1145649] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
C1q is a crucial component of the complement system, which is activated through the classical pathway to perform non-specific immune functions, serving as the first line of defense against pathogens. C1q can also bind to specific receptors to carry out immune and other functions, playing a vital role in maintaining immune homeostasis and normal physiological functions. In the developing central nervous system (CNS), C1q functions in synapse formation and pruning, serving as a key player in the development and homeostasis of neuronal networks in the CNS. C1q has a close relationship with microglia and astrocytes, and under their influence, C1q may contribute to the development of CNS disorders. Furthermore, C1q can also have independent effects on neurological disorders, producing either beneficial or detrimental outcomes. Most of the evidence for these functions comes from animal models, with some also from human specimen studies. C1q is now emerging as a promising target for the treatment of a variety of diseases, and clinical trials are already underway for CNS disorders. This article highlights the role of C1q in CNS diseases, offering new directions for the diagnosis and treatment of these conditions.
Collapse
Affiliation(s)
- Wenjie Zhang
- Department of Emergency Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of General Practice, Xingyang Sishui Central Health Center, Zhengzhou, China
| | - Yuan Chen
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Pei
- Department of Emergency Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Jiang J, Wang Y, Deng M. New developments and opportunities in drugs being trialed for amyotrophic lateral sclerosis from 2020 to 2022. Front Pharmacol 2022; 13:1054006. [PMID: 36518658 PMCID: PMC9742490 DOI: 10.3389/fphar.2022.1054006] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 08/31/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that primarily affects motor neurons in the brain and spinal cord. In the recent past, there have been just two drugs approved for treatment, riluzole and edaravone, which only prolong survival by a few months. However, there are many novel experimental drugs in development. In this review, we summarize 53 new drugs that have been evaluated in clinical trials from 2020 to 2022, which we have classified into eight mechanistic groups (anti-apoptotic, anti-inflammatory, anti-excitotoxicity, regulated integrated stress response, neurotrophic factors and neuroprotection, anti-aggregation, gene therapy and other). Six were tested in phase 1 studies, 31 were in phase 2 studies, three failed in phase 3 studies and stopped further development, and the remaining 13 drugs were being tested in phase 3 studies, including methylcobalamin, masitinib, MN-166, verdiperstat, memantine, AMX0035, trazodone, CNM-Au8, pridopidine, SLS-005, IONN363, tofersen, and reldesemtiv. Among them, five drugs, including methylcobalamin, masitinib, AMX0035, CNM-Au8, and tofersen, have shown potent therapeutic effects in clinical trials. Recently, AMX0035 has been the third medicine approved by the FDA for the treatment of ALS after riluzole and edaravone.
Collapse
Affiliation(s)
| | | | - Min Deng
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| |
Collapse
|
10
|
Schulz K, Trendelenburg M. C1q as a target molecule to treat human disease: What do mouse studies teach us? Front Immunol 2022; 13:958273. [PMID: 35990646 PMCID: PMC9385197 DOI: 10.3389/fimmu.2022.958273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
The complement system is a field of growing interest for pharmacological intervention. Complement protein C1q, the pattern recognition molecule at the start of the classical pathway of the complement cascade, is a versatile molecule with additional non-canonical actions affecting numerous cellular processes. Based on observations made in patients with hereditary C1q deficiency, C1q is protective against systemic autoimmunity and bacterial infections. Accordingly, C1q deficient mice reproduce this phenotype with susceptibility to autoimmunity and infections. At the same time, beneficial effects of C1q deficiency on disease entities such as neurodegenerative diseases have also been described in murine disease models. This systematic review provides an overview of all currently available literature on the C1q knockout mouse in disease models to identify potential target diseases for treatment strategies focusing on C1q, and discusses potential side-effects when depleting and/or inhibiting C1q.
Collapse
Affiliation(s)
- Kristina Schulz
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- *Correspondence: Kristina Schulz,
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
11
|
The Complement System in the Central Nervous System: From Neurodevelopment to Neurodegeneration. Biomolecules 2022; 12:biom12020337. [PMID: 35204837 PMCID: PMC8869249 DOI: 10.3390/biom12020337] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/31/2022] [Accepted: 02/13/2022] [Indexed: 12/13/2022] Open
Abstract
The functions of the complement system to both innate and adaptive immunity through opsonization, cell lysis, and inflammatory activities are well known. In contrast, the role of complement in the central nervous system (CNS) which extends beyond immunity, is only beginning to be recognized as important to neurodevelopment and neurodegeneration. In addition to protecting the brain against invasive pathogens, appropriate activation of the complement system is pivotal to the maintenance of normal brain function. Moreover, overactivation or dysregulation may cause synaptic dysfunction and promote excessive pro-inflammatory responses. Recent studies have provided insights into the various responses of complement components in different neurological diseases and the regulatory mechanisms involved in their pathophysiology, as well as a glimpse into targeting complement factors as a potential therapeutic modality. However, there remain significant knowledge gaps in the relationship between the complement system and different brain disorders. This review summarizes recent key findings regarding the role of different components of the complement system in health and pathology of the CNS and discusses the therapeutic potential of anti-complement strategies for the treatment of neurodegenerative conditions.
Collapse
|
12
|
Xu X, Zhang J, Li S, Al-Nusaif M, Zhou Q, Chen S, Le W. Bone Marrow Stromal Cell Antigen 2: Is a Potential Neuroinflammation Biomarker of SOD1G93A Mouse Model of Amyotrophic Lateral Sclerosis in Pre-symptomatic Stage. Front Neurosci 2022; 15:788730. [PMID: 35197819 PMCID: PMC8858987 DOI: 10.3389/fnins.2021.788730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation has long been thought to be associated with amyotrophic lateral sclerosis (ALS) development and progression. However, the exact molecular mechanisms of neuroinflammation underlying ALS remain largely unknown. In the present study, we attempted to elucidate the genetic basis of neuroinflammation in ALS by comparing the transcriptomic profile of the anterior horns of the lumbar spinal cord (AHLSC) between SOD1G93A mice and their wild-type (WT) littermates. Our results revealed that immune-related genes were selectively up-regulated in the AHLSC of pre-symptomatic ALS mice (40 days of age) compared to age-matched WT control mice. Notably, the differential expression level of these immune-related genes became more significant at the symptomatic stage of disease (90 days of age) in the ALS mice. Subsequently, eight genes involved in innate immune response in the AHLSC of ALS mice were further validated by qRT-PCR analysis. Of these genes, bone marrow stromal cell antigen 2 (BST2) was found for the first time to be significantly higher in the AHLSC of pre-symptomatic ALS mice when compared with WT mice. The increasing trend of BST2 expression became more obvious in the symptomatic stage. Immunofluorescent staining further confirmed that BST2 is mainly expressed on microglia in the AHLSC of ALS mice. These findings support the view that immune-related neuroinflammation is involved in the early pathogenesis of ALS, and BST2 may serve as a potential target for ameliorating microglia-mediated neuroinflammation pathologies in ALS.
Collapse
Affiliation(s)
- Xiaojiao Xu
- Institute of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingjing Zhang
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Murad Al-Nusaif
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Qinming Zhou
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
- *Correspondence: Weidong Le,
| |
Collapse
|
13
|
Wang M, Kang L, Wang Y, Yang B, Zhang C, Lu Y, Kang L. Microglia in motor neuron disease: Signaling evidence from last 10 years. Dev Neurobiol 2022; 82:625-638. [PMID: 36309345 PMCID: PMC9828749 DOI: 10.1002/dneu.22905] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 07/09/2022] [Accepted: 10/19/2022] [Indexed: 01/30/2023]
Abstract
Motor neuron disease (MND), including amyotrophic lateral sclerosis, spinal muscular atrophy and others, involved the upper or lower motor neurons selective loss, is characterized by neurodegeneration and neuroinflammation, in conjunction with microglia. We summarized that pathways and key mediators are associated with microglia, such as fractalkine signaling, purinergic signaling, NF-κB signaling, p38 MAPK signaling, TREM2-APOE signaling, ROCK signaling, C1q signaling, and Ion channel, which are involved in the activation, proliferation, and inflammation of microglia. This review aims to identify the microglia-related molecular target and explore potential treatment strategies for MND based on that target.
Collapse
Affiliation(s)
- Min‐Jia Wang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Lu Kang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Yao‐Zheng Wang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Bi‐Ru Yang
- Department of Postpartum RehabilitationSichuan Jinxin Women & Children HospitalChengduChina
| | - Chun Zhang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Yu‐Feng Lu
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Liang Kang
- Institute of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| |
Collapse
|
14
|
Wells TL, Myles JR, Akay T. C-Boutons and Their Influence on Amyotrophic Lateral Sclerosis Disease Progression. J Neurosci 2021; 41:8088-8101. [PMID: 34380764 PMCID: PMC8460137 DOI: 10.1523/jneurosci.0660-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 11/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease with progressive motor neuron death, where patients usually die within 5 years of diagnosis. Previously, we showed that the C-boutons, which are large cholinergic synapses to motor neurons that modulate motor neuron activity, are necessary for behavioral compensation in mSOD1G93A mice, a mouse model for ALS. We reasoned that, since the C-boutons likely increase the excitability of surviving motor neurons to compensate for motor neuron loss during ALS disease progression, then amplitude modulation through the C-boutons likely increases motor neuron stress and worsens disease progression. By comparing male and female mSOD1G93A mice to mSOD1G93A mice with genetically silenced C-boutons [mSOD1G93A ; Dbx1::cre; ChATfl/fl (mSOD1G93A/Coff )], we show that the C-boutons do not influence the humane end point of mSOD1G93A mice; however, our histologic analysis shows that C-bouton silencing significantly improves fast-twitch muscle innervation over time. Using immunohistology, we also show that the C-boutons are active in a task-dependent manner, and that symptomatic mSOD1G93A mice show significantly higher C-bouton activity than wild-type mice during low-intensity walking. Last, by using behavioral analysis, we provide evidence that C-bouton silencing in combination with swimming is beneficial for the behavioral capabilities of mSOD1G93A mice. Our observations suggest that manipulating the C-boutons in combination with a modulatory-targeted training program may therefore be beneficial for ALS patients and could result in improved mobility and quality of life.SIGNIFICANCE STATEMENT Despite decades of research on amyotrophic lateral sclerosis (ALS), there have been little improvements in treatments and therapies. We sought to better understand how the activation of C-boutons, which are large cholinergic modulatory synapses on motor neurons, change and affect the disease as it progresses. When these C-boutons are genetically silenced and exercises designed to otherwise activate the C-boutons are frequently performed in ALS model mice, the mice perform better than their untreated counterparts over time. C-bouton-targeted therapies could therefore be beneficial for ALS patients and could result in improved mobility and quality of life.
Collapse
Affiliation(s)
- Tyler L Wells
- Atlantic Mobility Action Project, Brain Repair Center, Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 0A8, Canada
| | - Jacob R Myles
- Atlantic Mobility Action Project, Brain Repair Center, Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 0A8, Canada
| | - Turgay Akay
- Atlantic Mobility Action Project, Brain Repair Center, Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 0A8, Canada
| |
Collapse
|
15
|
Wang M, Liu Z, Du J, Yuan Y, Jiao B, Zhang X, Hou X, Shen L, Guo J, Jiang H, Xia K, Tang J, Zhang R, Tang B, Wang J. Evaluation of Peripheral Immune Activation in Amyotrophic Lateral Sclerosis. Front Neurol 2021; 12:628710. [PMID: 34248812 PMCID: PMC8264193 DOI: 10.3389/fneur.2021.628710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/27/2021] [Indexed: 01/11/2023] Open
Abstract
Accumulating evidence has revealed that immunity plays an important role in amyotrophic lateral sclerosis (ALS) progression. However, the results regarding the serum levels of immunoglobulin and complement are inconsistent in patients with ALS. Although immune dysfunctions have also been reported in patients with other neurodegenerative diseases, few studies have explored whether immune dysfunction in ALS is similar to that in other neurodegenerative diseases. Therefore, we performed this study to address these gaps. In the present study, serum levels of immunoglobulin and complement were measured in 245 patients with ALS, 65 patients with multiple system atrophy (MSA), 60 patients with Parkinson's disease (PD), and 82 healthy controls (HCs). Multiple comparisons revealed that no significant differences existed between patients with ALS and other neurodegenerative diseases in immunoglobulin and complement levels. Meta-analysis based on data from our cohort and eight published articles was performed to evaluate the serum immunoglobulin and complement between patients with ALS and HCs. The pooled results showed that patients with ALS had higher C4 levels than HCs. In addition, we found that the IgG levels were lower in early-onset ALS patients than in late-onset ALS patients and HCs, and the correlations between age at onset of ALS and IgG or IgA levels were significant positive. In conclusion, our data supplement existing literature on understanding the role of peripheral immunity in ALS.
Collapse
Affiliation(s)
- Mengli Wang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanchun Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuewei Zhang
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Hou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Medical Genetics, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Medical Genetics, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Kun Xia
- Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Jianguang Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Medical Genetics, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Medical Genetics, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Abstract
The complement cascade is an evolutionary ancient innate immune defense system, playing a major role in the defense against infections. Its function in maintaining host homeostasis on activated cells has been emphasized by the crucial role of its overactivation in ever growing number of diseases, such as atypical hemolytic uremic syndrome (aHUS), autoimmune diseases as systemic lupus erythematosus (SLE), C3 glomerulopathies (C3GN), age-related macular degeneration (AMD), graft rejection, Alzheimer disease, and cancer, to name just a few. The last decade of research on complement has extended its implication in many pathological processes, offering new insights to potential therapeutic targets and asserting the necessity of reliable, sensitive, specific, accurate, and reproducible biomarkers to decipher complement role in pathology. We need to evaluate accurately which pathway or role should be targeted pharmacologically, and optimize treatment efficacy versus toxicity. This chapter is an introduction to the role of complement in human diseases and the use of complement-related biomarkers in the clinical practice. It is a part of a book intending to give reliable and standardized methods to evaluate complement according to nowadays needs and knowledge.
Collapse
|
17
|
Ziabska K, Ziemka-Nalecz M, Pawelec P, Sypecka J, Zalewska T. Aberrant Complement System Activation in Neurological Disorders. Int J Mol Sci 2021; 22:4675. [PMID: 33925147 PMCID: PMC8125564 DOI: 10.3390/ijms22094675] [Citation(s) in RCA: 29] [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: 03/24/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
The complement system is an assembly of proteins that collectively participate in the functions of the healthy and diseased brain. The complement system plays an important role in the maintenance of uninjured (healthy) brain homeostasis, contributing to the clearance of invading pathogens and apoptotic cells, and limiting the inflammatory immune response. However, overactivation or underregulation of the entire complement cascade within the brain may lead to neuronal damage and disturbances in brain function. During the last decade, there has been a growing interest in the role that this cascading pathway plays in the neuropathology of a diverse array of brain disorders (e.g., acute neurotraumatic insult, chronic neurodegenerative diseases, and psychiatric disturbances) in which interruption of neuronal homeostasis triggers complement activation. Dysfunction of the complement promotes a disease-specific response that may have either beneficial or detrimental effects. Despite recent advances, the explicit link between complement component regulation and brain disorders remains unclear. Therefore, a comprehensible understanding of such relationships at different stages of diseases could provide new insight into potential therapeutic targets to ameliorate or slow progression of currently intractable disorders in the nervous system. Hence, the aim of this review is to provide a summary of the literature on the emerging role of the complement system in certain brain disorders.
Collapse
Affiliation(s)
| | | | | | | | - Teresa Zalewska
- Mossakowski Medical Research Centre, NeuroRepair Department, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland; (K.Z.); (M.Z.-N.); (P.P.); (J.S.)
| |
Collapse
|
18
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|
19
|
Propson NE, Gedam M, Zheng H. Complement in Neurologic Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2020; 16:277-298. [PMID: 33234021 DOI: 10.1146/annurev-pathol-031620-113409] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Classic innate immune signaling pathways provide most of the immune response in the brain. This response activates many of the canonical signaling mechanisms identified in peripheral immune cells, despite their relative absence in this immune-privileged tissue. Studies over the past decade have strongly linked complement protein production and activation to age-related functional changes and neurodegeneration. The reactivation of the complement signaling pathway in aging and disease has opened new avenues for understanding brain aging and neurological disease pathogenesis and has implicated cell types such as astrocytes, microglia, endothelial cells, oligodendrocytes, neurons, and even peripheral immune cells in these processes. In this review, we aim to unravel the past decade of research related to complement activation and its numerous consequences in aging and neurological disease.
Collapse
Affiliation(s)
- Nicholas E Propson
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Manasee Gedam
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
20
|
He J, Fu J, Fan D. The complement C7 variant rs3792646 is associated with amyotrophic lateral sclerosis in a Han Chinese population. Neurobiol Aging 2020; 99:103.e1-103.e7. [PMID: 33303220 DOI: 10.1016/j.neurobiolaging.2020.10.012] [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: 04/06/2020] [Revised: 09/27/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
The complement system has been shown to have a critical pathogenetic role in amyotrophic lateral sclerosis (ALS). Recently a C7 variant in rs3792646 was linked to neurodegenerative diseases in a Chinese population. We used whole exome sequencing to evaluate the role of C7 (rs3792646) in ALS in a Chinese cohort with 1970 individuals. The minor allele frequency in cases was 0.032 while 0.016 in controls, suggesting this variant was associated with ALS. Further analyses showed the prevalence of the variant was significantly higher in Chinese than Caucasian, suggesting its importance in Han individuals. rs3792646-C was significantly associated with a lower onset age in both genders, and a survival analysis revealed a significant relationship between the variant and decreased survival. There was no significant association between the variant and other common ALS-related variants. Our study further elucidated the relationship between the complement system and ALS from a genetic perspective. In addition, the results suggested C7 (rs3792646) could be a potential predictive factor for poor prognosis in ALS.
Collapse
Affiliation(s)
- Ji He
- Department of Neurology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Jiayu Fu
- Department of Neurology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, People's Republic of China; Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, People's Republic of China.
| |
Collapse
|
21
|
Gavriilaki M, Kimiskidis VK, Gavriilaki E. Precision Medicine in Neurology: The Inspirational Paradigm of Complement Therapeutics. Pharmaceuticals (Basel) 2020; 13:E341. [PMID: 33114553 PMCID: PMC7693884 DOI: 10.3390/ph13110341] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Precision medicine has emerged as a central element of healthcare science. Complement, a component of innate immunity known for centuries, has been implicated in the pathophysiology of numerous incurable neurological diseases, emerging as a potential therapeutic target and predictive biomarker. In parallel, the innovative application of the first complement inhibitor in clinical practice as an approved treatment of myasthenia gravis (MG) and neuromyelitis optica spectrum disorders (NMOSD) related with specific antibodies raised hope for the implementation of personalized therapies in detrimental neurological diseases. A thorough literature search was conducted through May 2020 at MEDLINE, EMBASE, Cochrane Library and ClinicalTrials.gov databases based on medical terms (MeSH)" complement system proteins" and "neurologic disease". Complement's role in pathophysiology, monitoring of disease activity and therapy has been investigated in MG, multiple sclerosis, NMOSD, spinal muscular atrophy, amyotrophic lateral sclerosis, Parkinson, Alzheimer, Huntington disease, Guillain-Barré syndrome, chronic inflammatory demyelinating polyneuropathy, stroke, and epilepsy. Given the complexity of complement diagnostics and therapeutics, this state-of-the-art review aims to provide a brief description of the complement system for the neurologist, an overview of novel complement inhibitors and updates of complement studies in a wide range of neurological disorders.
Collapse
Affiliation(s)
- Maria Gavriilaki
- Postgraduate Course, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasilios K. Kimiskidis
- Postgraduate Course, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, 57010 Thessaloniki, Greece;
| |
Collapse
|
22
|
Knockout of reactive astrocyte activating factors slows disease progression in an ALS mouse model. Nat Commun 2020; 11:3753. [PMID: 32719333 PMCID: PMC7385161 DOI: 10.1038/s41467-020-17514-9] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Reactive astrocytes have been implicated in the pathogenesis of neurodegenerative diseases, including a non-cell autonomous effect on motor neuron survival in ALS. We previously defined a mechanism by which microglia release three factors, IL-1α, TNFα, and C1q, to induce neurotoxic astrocytes. Here we report that knocking out these three factors markedly extends survival in the SOD1G93A ALS mouse model, providing evidence for gliosis as a potential ALS therapeutic target.
Collapse
|
23
|
Hammond JW, Bellizzi MJ, Ware C, Qiu WQ, Saminathan P, Li H, Luo S, Ma SA, Li Y, Gelbard HA. Complement-dependent synapse loss and microgliosis in a mouse model of multiple sclerosis. Brain Behav Immun 2020; 87:739-750. [PMID: 32151684 PMCID: PMC8698220 DOI: 10.1016/j.bbi.2020.03.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, neurodegenerative disease of the CNS characterized by both grey and white matter injury. Microglial activation and a reduction in synaptic density are key features of grey matter pathology that can be modeled with MOG35-55 experimental autoimmune encephalomyelitis (EAE). Complement deposition combined with microglial engulfment has been shown during normal development and in disease as a mechanism for pruning synapses. We tested whether there is excess complement production in the EAE hippocampus and whether complement-dependent synapse loss is a source of degeneration in EAE using C1qa and C3 knockout mice. We found that C1q and C3 protein and mRNA levels were elevated in EAE mice. Genetic loss of C3 protected mice from EAE-induced synapse loss, reduced microglial activation, decreased the severity of the EAE clinical score, and protected memory/freezing behavior after contextual fear conditioning. C1qa KO mice with EAE showed little to no change on these measurements compared to WT EAE mice. Thus, pathologic expression and activation of the early complement pathway, specifically at the level of C3, contributes to hippocampal grey matter pathology in the EAE.
Collapse
Affiliation(s)
- Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Correspondence: Jennetta W. Hammond,
University of Rochester, Center for Neurotherapeutics Discovery, 601 Elmwood
Avenue, Box 645, Rochester, NY 14642, USA,
, Phone:
1-585-273-2872
| | - Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Caroline Ware
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Wen Q. Qiu
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Priyanka Saminathan
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Microbiology and Immunology, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Shaopeiwen Luo
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Stefanie A. Ma
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Yuanhao Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| |
Collapse
|
24
|
Sex-Specific Differences in Motor-Unit Remodeling in a Mouse Model of ALS. eNeuro 2020; 7:ENEURO.0388-19.2020. [PMID: 32033983 PMCID: PMC7044502 DOI: 10.1523/eneuro.0388-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Progressive loss of neuromuscular junctions (NMJs) is an early event in amyotrophic lateral sclerosis (ALS), preceding the global degeneration of motor axons and being accompanied by new axonal sprouting within the same axonal arbor. Some aspects of ALS onset and progression seem to be affected by sex in animal models of the disease. However, whether there are sex-specific differences in the pattern or time course of NMJ loss and repair within single motor axons remains unknown. We performed further analysis of a previously published in vivo dataset, obtained from male and female SOD1G37R mice. We found that NMJ losses are as frequent in male and female motor axons but, intriguingly, axonal sprouting is more frequent in female than male mice, resulting in a net increase of axonal arborization. Interestingly, these numerous new axonal branches in female mice are associated with a slightly faster decline in grip strength, increased NMJ denervation, and reduced α-motor neuron survival. Collectively, these results suggest that excessive axonal sprouting and motor-unit (MU) expansion in female SOD1G37R mice are maladaptive during ALS progression.
Collapse
|
25
|
Waegaert R, Dirrig-Grosch S, Parisot F, Keime C, Henriques A, Loeffler JP, René F. Longitudinal transcriptomic analysis of altered pathways in a CHMP2B intron5-based model of ALS-FTD. Neurobiol Dis 2019; 136:104710. [PMID: 31837425 DOI: 10.1016/j.nbd.2019.104710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/28/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are two neurodegenerative diseases with currently no cure. These two diseases share a clinical continuum with overlapping genetic causes. Mutations in the CHMP2B gene are found in patients with ALS, FTD and ALS-FTD. To highlight deregulated mechanisms occurring in ALS-FTD linked to the CHMP2B gene, we performed a whole transcriptomic study on lumbar spinal cord from CHMP2Bintron5 mice, a model that develops progressive motor alterations associated with dementia symptoms reminiscent of both ALS and FTD. To gain insight into the transcriptomic changes taking place during disease progression this study was performed at three stages: asymptomatic, symptomatic and end stage. We showed that before appearance of motor symptoms, the major disrupted mechanisms were linked with the immune system/inflammatory response and lipid metabolism. These processes were progressively replaced by alterations of neuronal electric activity as motor symptoms appeared, alterations that could lead to motor neuron dysfunction. To investigate overlapping alterations in gene expression between two ALS-causing genes, we then compared the transcriptome of symptomatic CHMP2Bintron5 mice with the one of symptomatic SOD1G86R mice and found the same families deregulated providing further insights into common underlying dysfunction of biological pathways, disrupted or disturbed in ALS. Altogether, this study provides a database to explore potential new candidate genes involved in the CHMP2Bintron5-based pathogenesis of ALS, and provides molecular clues to further understand the functional consequences that diseased neurons expressing CHMP2B mutant may have on their neighbor cells.
Collapse
Affiliation(s)
- Robin Waegaert
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Sylvie Dirrig-Grosch
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Florian Parisot
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS, UMR7104, Université de Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Alexandre Henriques
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Jean-Philippe Loeffler
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Frédérique René
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France.
| |
Collapse
|
26
|
Lee JD, Coulthard LG, Woodruff TM. Complement dysregulation in the central nervous system during development and disease. Semin Immunol 2019; 45:101340. [PMID: 31708347 DOI: 10.1016/j.smim.2019.101340] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/15/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
The complement cascade is an important arm of the immune system that plays a key role in protecting the central nervous system (CNS) from infection. Recently, it has also become clear that complement proteins have fundamental roles in the developing and aging CNS that are distinct from their roles in immunity. During neurodevelopment, complement signalling is involved in diverse processes including neural tube closure, neural progenitor proliferation and differentiation, neuronal migration, and synaptic pruning. In acute neurotrauma and ischamic brain injury, complement drives inflammation and neuronal death, but also neuroprotection and regeneration. In diseases of the aging CNS including dementias and motor neuron disease, chronic complement activation is associated with glial activation, and synapse and neuron loss. Proper regulation of complement is thus essential to allow for an appropriately developed CNS and prevention of excessive damage following neurotrauma or during neurodegeneration. This review provides a comprehensive overview of the evidence for functional roles of complement in brain formation, and its dysregulation during acute and chronic disease. We also provide working models for how complement can lead to neurodevelopmental disorders such as schizophrenia and autism, and either protect, or propagate neurodegenerative diseases including Alzheimer's disease and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Liam G Coulthard
- Royal Brisbane and Women's Hospital, Herston, Australia; School of Clinical Medicine, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
27
|
Parker SE, Hanton AM, Stefanou SN, Noakes PG, Woodruff TM, Lee JD. Revisiting the role of the innate immune complement system in ALS. Neurobiol Dis 2019; 127:223-232. [DOI: 10.1016/j.nbd.2019.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/21/2019] [Accepted: 03/04/2019] [Indexed: 12/11/2022] Open
|
28
|
苏 丹, 张 勇, 毕 方, 肖 波. [Proteomic analysis of the cerebrospinal fluid from patients with amyotrophic lateral sclerosis based on tandem mass spectrometry technique]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:428-436. [PMID: 31068286 PMCID: PMC6743996 DOI: 10.12122/j.issn.1673-4254.2019.04.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the differentially expressed proteins in the cerebrospinal fluid (CSF) of patients with amyotrophic lateral sclerosis (ALS) at the proteomics level using tandem mass spectrometry label (TMT) technique and explore the pathogenic mechanism and related pathways of ALS. METHODS Between November, 2017 and April, 2018, 5 patients with medulla oblongata onset ALS and 5 patients with limb onset ALS were selected from the Departments of Neurology of 928 Hospital of Army Joint Logistics Support Force of PLA and Xiangya Hospital of Central South University, with 5 patients with migraine and low intracranial pressure headache serving as the healthy controls.CSF samples were obtained from all the participants, and the differentially expressed proteins in the CSF were identified using tandem mass spectrometry (TMT) technique with bioinformatics analysis. RESULTS A total of 1530 proteins were identified and quantified in the CSF samples.The expression of 48 proteins was up-regulated and 6 proteins were down-regulated in medulla oblongata onset ALS patients; 16 proteins were up-regulated and 19 were down-regulated in limb onset ALS patients.GO analysis showed that these proteins, which were distributed both within and outside the cells, were involved in cell physiological process, single organ process and biological regulation and had binding function, catalytic activity, and receptor activity.KEGG pathway analysis showed that the up-regulated proteins in the CSF from patients with medulla oblongata onset ALS participated in 3 pathways involving the lysosomes, metabolism, and measles.The down-regulated proteins in the CSF from patients with limb onset ALS participated in 7 pathways involving the complement and coagulation cascade, Staphylococcus aureus infection and herpes simplex infection, and all the pathways contained complement components. CONCLUSIONS The CSF samples of ALS patients with medullary onset and limb onset have differentially expressed proteins.The lysosomal pathway is involved in the occurrence and progression of ALS with medullary onset, and the immune responses are involved in the occurrence and progression of ALS with limb onset.
Collapse
Affiliation(s)
- 丹丹 苏
- 中国人民解放军联勤保障部队第 928医院神经内科,海南 海口 571100Department of Neurology, 928 Hospital of Joint Logistics Support Force of PLA, Haikou 571100, China
| | - 勇 张
- 中国人民解放军联勤保障部队第 928医院神经内科,海南 海口 571100Department of Neurology, 928 Hospital of Joint Logistics Support Force of PLA, Haikou 571100, China
| | - 方方 毕
- 中南大学湘雅医院神经内科,湖南 长沙 410008Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - 波 肖
- 中南大学湘雅医院神经内科,湖南 长沙 410008Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
29
|
Carpanini SM, Torvell M, Morgan BP. Therapeutic Inhibition of the Complement System in Diseases of the Central Nervous System. Front Immunol 2019; 10:362. [PMID: 30886620 PMCID: PMC6409326 DOI: 10.3389/fimmu.2019.00362] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system plays critical roles in development, homeostasis, and regeneration in the central nervous system (CNS) throughout life; however, complement dysregulation in the CNS can lead to damage and disease. Complement proteins, regulators, and receptors are widely expressed throughout the CNS and, in many cases, are upregulated in disease. Genetic and epidemiological studies, cerebrospinal fluid (CSF) and plasma biomarker measurements and pathological analysis of post-mortem tissues have all implicated complement in multiple CNS diseases including multiple sclerosis (MS), neuromyelitis optica (NMO), neurotrauma, stroke, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Given this body of evidence implicating complement in diverse brain diseases, manipulating complement in the brain is an attractive prospect; however, the blood-brain barrier (BBB), critical to protect the brain from potentially harmful agents in the circulation, is also impermeable to current complement-targeting therapeutics, making drug design much more challenging. For example, antibody therapeutics administered systemically are essentially excluded from the brain. Recent protocols have utilized "Trojan horse" techniques to transport therapeutics across the BBB or used osmotic shock or ultrasound to temporarily disrupt the BBB. Most research to date exploring the impact of complement inhibition on CNS diseases has been in animal models, and some of these studies have generated convincing data; for example, in models of MS, NMO, and stroke. There have been a few recent clinical trials of available anti-complement drugs in CNS diseases associated with BBB impairment, for example the use of the anti-C5 monoclonal antibody (mAb) eculizumab in NMO, but for most CNS diseases there have been no human trials of anti-complement therapies. Here we will review the evidence implicating complement in diverse CNS disorders, from acute, such as traumatic brain or spine injury, to chronic, including demyelinating, neuroinflammatory, and neurodegenerative diseases. We will discuss the particular problems of drug access into the CNS and explore ways in which anti-complement therapies might be tailored for CNS disease.
Collapse
Affiliation(s)
- Sarah M Carpanini
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Megan Torvell
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Bryan Paul Morgan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
30
|
Henstridge CM, Tzioras M, Paolicelli RC. Glial Contribution to Excitatory and Inhibitory Synapse Loss in Neurodegeneration. Front Cell Neurosci 2019; 13:63. [PMID: 30863284 PMCID: PMC6399113 DOI: 10.3389/fncel.2019.00063] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Synapse loss is an early feature shared by many neurodegenerative diseases, and it represents the major correlate of cognitive impairment. Recent studies reveal that microglia and astrocytes play a major role in synapse elimination, contributing to network dysfunction associated with neurodegeneration. Excitatory and inhibitory activity can be affected by glia-mediated synapse loss, resulting in imbalanced synaptic transmission and subsequent synaptic dysfunction. Here, we review the recent literature on the contribution of glia to excitatory/inhibitory imbalance, in the context of the most common neurodegenerative disorders. A better understanding of the mechanisms underlying pathological synapse loss will be instrumental to design targeted therapeutic interventions, taking in account the emerging roles of microglia and astrocytes in synapse remodeling.
Collapse
Affiliation(s)
- Christopher M Henstridge
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Dementia Research Institute UK, The University of Edinburgh, Edinburgh, United Kingdom
| | - Makis Tzioras
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Dementia Research Institute UK, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rosa C Paolicelli
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
31
|
Hammond JW, Qiu WQ, Marker DF, Chamberlain JM, Greaves-Tunnell W, Bellizzi MJ, Lu SM, Gelbard HA. HIV Tat causes synapse loss in a mouse model of HIV-associated neurocognitive disorder that is independent of the classical complement cascade component C1q. Glia 2018; 66:2563-2574. [PMID: 30325063 DOI: 10.1002/glia.23511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
Microglial activation, increased proinflammatory cytokine production, and a reduction in synaptic density are key pathological features associated with HIV-associated neurocognitive disorders (HAND). Even with combination antiretroviral therapy (cART), more than 50% of HIV-positive individuals experience some type of cognitive impairment. Although viral replication is inhibited by cART, HIV proteins such as Tat are still produced within the nervous system that are neurotoxic, involved in synapse elimination, and provoke enduring neuroinflammation. As complement deposition on synapses followed by microglial engulfment has been shown during normal development and disease to be a mechanism for pruning synapses, we have tested whether complement is required for the loss of synapses that occurs after a cortical Tat injection mouse model of HAND. In Tat-injected animals evaluated 7 or 28 days after injection, levels of early complement pathway components, C1q and C3, are significantly elevated and associated with microgliosis and a loss of synapses. However, C1qa knockout mice have the same level of Tat-induced synapse loss as wild-type (WT) mice, showing that the C1q-initiated classical complement cascade is not driving synapse removal during HIV1 Tat-induced neuroinflammation.
Collapse
Affiliation(s)
- Jennetta W Hammond
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Wen Q Qiu
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Daniel F Marker
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Jeffrey M Chamberlain
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Will Greaves-Tunnell
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Matthew J Bellizzi
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Shao-Ming Lu
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
32
|
Konishi H, Kiyama H. Microglial TREM2/DAP12 Signaling: A Double-Edged Sword in Neural Diseases. Front Cell Neurosci 2018; 12:206. [PMID: 30127720 PMCID: PMC6087757 DOI: 10.3389/fncel.2018.00206] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022] Open
Abstract
Microglia are activated after neuronal injury and in neurodegenerative diseases, and trigger neuroinflammation in the central nervous system (CNS). Microglia-derived neuroinflammation has both beneficial and detrimental effects on neurons. Because the timing and magnitude of microglial activation is thought to be a critical determinant of neuronal fate, understanding the molecular mechanisms underlying microglial activation is required to enable establishment of microglia-targeted therapies for neural diseases. Plasma membrane receptors play primary roles as activators of microglia and in this review, we focus on a receptor complex involving triggering receptor expressed on myeloid cells 2 (TREM2) and DNAX-activating protein of 12 kDa (DAP12), both of which are causative genes for Nasu-Hakola disease, a dementia with bone cysts. Recent transcriptome approaches demonstrated TREM2/DAP12 signaling as the principal regulator that transforms microglia from a homeostatic to a neural disease-associated state. Furthermore, animal model studies revealed critical roles for TREM2/DAP12 in the regulation of microglial activity, including survival, phagocytosis, and cytokine production, not only in Alzheimer's disease but also in other neural diseases, such as Parkinson's disease, demyelinating disease, ischemia, and peripheral nerve injury. Intriguingly, while TREM2/DAP12-mediated microglial activation is detrimental for some diseases, including peripheral nerve injury, it is beneficial for other diseases. As the role of activated microglia differs among disease models, TREM2/DAP12 signaling may result in different outcomes in different diseases. In this review we discuss recent perspectives on the role of TREM2/DAP12 in microglia and their contribution to neural diseases.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
33
|
Tenner AJ, Stevens B, Woodruff TM. New tricks for an ancient system: Physiological and pathological roles of complement in the CNS. Mol Immunol 2018; 102:3-13. [PMID: 29958698 DOI: 10.1016/j.molimm.2018.06.264] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022]
Abstract
While the mechanisms underlying the functions of the complement system in the central nervous system (CNS) and systemically, namely opsonization, chemotaxis, membrane lysis, and regulation of inflammation are the same, the plethora of functions that complement orchestrates in the central nervous system (CNS) is complex. Strictly controlled expression of complement effector molecules, regulators and receptors across the gamut of life stages (embryogenesis, development and maturation, aging and disease) dictate fascinating contributions for this ancient system. Furthermore, it is becoming apparent that complement functions differ widely across distinct brain regions. This review provides a comprehensive overview of the newly identified roles for complement in the brain, including its roles in CNS development and function, during aging and in the processes of neurodegeneration. The diversity and selectively of beneficial and detrimental activities of complement, while challenging, should lead to precision targeting of specific components to provide disease modifying treatments for devastating psychiatric and neurodegenerative disorders that are still without effective treatment.
Collapse
Affiliation(s)
- Andrea J Tenner
- Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, CA, United States.
| | - Beth Stevens
- F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Department of Neurobiology, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Kjældgaard AL, Pilely K, Olsen KS, Pedersen SW, Lauritsen AØ, Møller K, Garred P. Amyotrophic lateral sclerosis: The complement and inflammatory hypothesis. Mol Immunol 2018; 102:14-25. [PMID: 29933890 DOI: 10.1016/j.molimm.2018.06.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/15/2018] [Accepted: 06/06/2018] [Indexed: 12/28/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, neurodegenerative motor neuron disease. The aetiology of ALS remains an enigma which hinders the design of an effective treatment to prevent, postpone, or reverse the pathophysiological changes occurring during the aggressive progression of this disease. During the last decade, basic research within the innate immune system, and in particular the complement system, has revealed new, important roles of the innate immune system during development, homeostasis, and ageing within as well as outside the central nervous system. Several lines of evidence indicate that aberrant activation of the complement system locally in the central nervous system as well as systemically may be involved in the pathophysiology of ALS. This exciting new knowledge could point towards the innate immune system as a potential target of medical intervention in ALS. Recently, the historic perception of ALS as a central neurodegenerative disease has been challenged due to the significant amount of evidence of a dying-back mechanism causing the selective destruction of the motor neurons, indicating that disease onset occurs outside the borders of the blood-brain-barrier. This review addresses the function of the innate immune system during ALS. We emphasize the role of the complement system and specifically suggest the involvement of ficolin-3 from the lectin pathway in the pathophysiology of ALS.
Collapse
Affiliation(s)
- Anne-Lene Kjældgaard
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Diagnostic Centre, Section 7631; Department of Neuroanaesthesiology.
| | - Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Diagnostic Centre, Section 7631
| | | | - Stephen Wørlich Pedersen
- Department of Neurology, Neuroscience Centre, Rigshospitalet, Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Diagnostic Centre, Section 7631
| |
Collapse
|
35
|
Lee JD, Levin SC, Willis EF, Li R, Woodruff TM, Noakes PG. Complement components are upregulated and correlate with disease progression in the TDP-43 Q331K mouse model of amyotrophic lateral sclerosis. J Neuroinflammation 2018; 15:171. [PMID: 29859100 PMCID: PMC5984816 DOI: 10.1186/s12974-018-1217-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Components of the innate immune complement system have been implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS) specifically using hSOD1 transgenic animals; however, a comprehensive examination of complement expression in other transgenic ALS models has not been performed. This study therefore aimed to determine the expression of several key complement components and regulators in the lumbar spinal cord and tibialis anterior muscle of TDP-43Q331K mice during different disease ages. METHODS Non-transgenic, TDP-43WT and TDP-43Q331K mice were examined at three different ages of disease progression. Expression of complement components and their regulators were examined using real-time quantitative PCR and enzyme-linked immunosorbent assay. Localisation of terminal complement component receptor C5aR1 within the lumbar spinal cord was also investigated using immunohistochemistry. RESULTS Altered levels of several major complement factors, including C5a, in the spinal cord and tibialis anterior muscle of TDP-43Q331K mice were observed as disease progressed, suggesting overall increased complement activation in TDP-43Q331K mice. C5aR1 increased during disease progression, with immuno-localisation demonstrating expression on motor neurons and expression on microglia surrounding the regions of motor neuron death. There was a strong negative linear relationship between spinal cord C1qB, C3 and C5aR1 mRNA levels with hind-limb grip strength. CONCLUSIONS These results indicate that similar to SOD1 transgenic animals, local complement activation and increased expression of C5aR1 may contribute to motor neuron death and neuromuscular junction denervation in the TDP-43Q331K mouse ALS model. This further validates C5aR1 as a potential therapeutic target for ALS.
Collapse
Affiliation(s)
- John D. Lee
- School of Biomedical Sciences, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
- University of Queensland Centre for Clinical Research, the University of Queensland, Herston, Brisbane, QLD 4029 Australia
| | - Samantha C. Levin
- School of Biomedical Sciences, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
| | - Emily F. Willis
- School of Biomedical Sciences, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
| | - Rui Li
- School of Biomedical Sciences, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
| | - Trent M. Woodruff
- School of Biomedical Sciences, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
| | - Peter G. Noakes
- School of Biomedical Sciences, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
- Queensland Brain Institute, the University of Queensland, St Lucia, Brisbane, QLD 4072 Australia
| |
Collapse
|
36
|
Toedebusch CM, Snyder JC, Jones MR, Garcia VB, Johnson GC, Villalón EL, Coates JR, Garcia ML. Arginase-1 expressing microglia in close proximity to motor neurons were increased early in disease progression in canine degenerative myelopathy, a model of amyotrophic lateral sclerosis. Mol Cell Neurosci 2018; 88:148-157. [PMID: 29408267 DOI: 10.1016/j.mcn.2018.01.009] [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] [Received: 10/05/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
Toxicity within superoxide dismutase-1 (SOD1)-associated familial amyotrophic lateral sclerosis (ALS) is non-cell autonomous with direct contribution from microglia. Microglia exhibit variable expression of neuroprotective and neurotoxic molecules throughout disease progression. The mechanisms regulating microglial phenotype within ALS are not well understood. This work presents a first study to examine the specific microglial phenotypic response in close association to motor neurons in a naturally occurring disease model of ALS, canine degenerative myelopathy (DM). Microglia closely associated with motor neurons were increased in all stages of DM progression, although only DM Late reached statistical significance. Furthermore, the number of arginase-1 expressing microglia per motor neuron were significantly increased in early stages of DM, whereas the number of inducible nitric oxide synthase (iNOS)-expressing microglia per motor neuron was indistinguishable from aged controls at all stages of disease. Fractalkine, a chemotactic molecule for microglia, was expressed in motor neurons, and the fractalkine receptor was specifically localized to microglia. However, we found no correlation between microglial response and lumbar spinal cord fractalkine levels. Taken together, these data suggest that arginase-1-expressing microglia are recruited to the motor neuron early in DM disease through a fractalkine-independent mechanism.
Collapse
Affiliation(s)
- Christine M Toedebusch
- Department of Veterinary Medicine and Surgery, University of Missouri, 900 E Campus Dr., Columbia, MO 65211, USA; Division of Biological Sciences, University of Missouri, Bond Life Sciences Center, 1201 E. Rollins St., Columbia, MO 65211, USA
| | - John C Snyder
- Department of Statistics, University of Missouri, Columbia, MO 65211, USA
| | - Maria R Jones
- Division of Biological Sciences, University of Missouri, Bond Life Sciences Center, 1201 E. Rollins St., Columbia, MO 65211, USA
| | - Virginia B Garcia
- Division of Biological Sciences, University of Missouri, Bond Life Sciences Center, 1201 E. Rollins St., Columbia, MO 65211, USA; Division of Biological Sciences, University of Missouri, LeFevre Hall, 1200 University Avenue, Columbia, MO 65211, USA
| | - Gayle C Johnson
- Department of Veterinary Pathobiology, University of Missouri, 800 E. Campus Loop, Columbia, MO 65211, USA
| | - Eric L Villalón
- Division of Biological Sciences, University of Missouri, Bond Life Sciences Center, 1201 E. Rollins St., Columbia, MO 65211, USA
| | - Joan R Coates
- Department of Veterinary Medicine and Surgery, University of Missouri, 900 E Campus Dr., Columbia, MO 65211, USA
| | - Michael L Garcia
- Division of Biological Sciences, University of Missouri, Bond Life Sciences Center, 1201 E. Rollins St., Columbia, MO 65211, USA; Division of Biological Sciences, University of Missouri, LeFevre Hall, 1200 University Avenue, Columbia, MO 65211, USA.
| |
Collapse
|
37
|
Chand KK, Lee KM, Lee JD, Qiu H, Willis EF, Lavidis NA, Hilliard MA, Noakes PG. Defects in synaptic transmission at the neuromuscular junction precede motor deficits in a TDP‐43
Q331K
transgenic mouse model of amyotrophic lateral sclerosis. FASEB J 2018; 32:2676-2689. [DOI: 10.1096/fj.201700835r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kirat K. Chand
- School of Biomedical Sciences Brisbane Queensland Australia
| | - Kah Meng Lee
- School of Biomedical Sciences Brisbane Queensland Australia
| | - John D. Lee
- School of Biomedical Sciences Brisbane Queensland Australia
| | - Hao Qiu
- School of Biomedical Sciences Brisbane Queensland Australia
| | | | | | | | | |
Collapse
|
38
|
Donahue RJ, Moller-Trane R, Nickells RW. Meta-analysis of transcriptomic changes in optic nerve injury and neurodegenerative models reveals a fundamental response to injury throughout the central nervous system. Mol Vis 2017; 23:987-1005. [PMID: 29386873 PMCID: PMC5757855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/22/2017] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Injury to the central nervous system (CNS) leads to transcriptional changes that effect tissue function and govern the process of neurodegeneration. Numerous microarray and RNA-Seq studies have been performed to identify these transcriptional changes in the retina following optic nerve injury and elsewhere in the CNS following a variety of insults. We reasoned that conserved transcriptional changes between injury paradigms would be important contributors to the neurodegenerative process. Therefore, we compared the expression results from heterogeneous studies of optic nerve injury and neurodegenerative models. METHODS Expression data was collected from the Gene Expression Omnibus. A uniform method for normalizing expression data and detecting differentially expressed (DE) genes was used to compare the transcriptomes from models of acute optic nerve injury (AONI), chronic optic nerve injury (CONI) and brain neurodegeneration. DE genes were split into genes that were more or less prevalent in the injured condition than the control condition (enriched and depleted, respectively) and transformed into their human orthologs so that transcriptomes from different species could be compared. Biologic significance of shared genes was assessed by analyzing lists of shared genes for gene ontology (GO) term over-representation and for representation in KEGG pathways. RESULTS There was significant overlap of enriched DE genes between transcriptomes of AONI, CONI and neurodegeneration studies even though the overall concordance between datasets was low. The depleted DE genes identified between AONI and CONI models were significantly overlapping, but this significance did not extend to comparisons between optic nerve injury models and neurodegeneration studies. The GO terms overrepresented among the enriched genes shared between AONI, CONI and neurodegeneration studies were related to innate immune processes like the complement system and interferon signaling. KEGG pathway analysis revealed that transcriptional alteration between JAK-STAT, PI3K-AKT and TNF signaling, among others, were conserved between all models that were analyzed. CONCLUSIONS There is a conserved transcriptional response to injury in the CNS. This transcriptional response is driven by the activation of the innate immune system and several regulatory pathways. Understanding the cellular origin of these pathways and the pathological consequences of their activation is essential for understanding and treating neurodegenerative disease.
Collapse
Affiliation(s)
- Ryan J. Donahue
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI
| | - Ralph Moller-Trane
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI
| | - Robert W. Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
39
|
Let's make microglia great again in neurodegenerative disorders. J Neural Transm (Vienna) 2017; 125:751-770. [PMID: 29027011 DOI: 10.1007/s00702-017-1792-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
All of the common neurodegenerative disorders-Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and prion diseases-are characterized by accumulation of misfolded proteins that trigger activation of microglia; brain-resident mononuclear phagocytes. This chronic form of neuroinflammation is earmarked by increased release of myriad cytokines and chemokines in patient brains and biofluids. Microglial phagocytosis is compromised early in the disease process, obfuscating clearance of abnormal proteins. This review identifies immune pathologies shared by the major neurodegenerative disorders. The overarching concept is that aberrant innate immune pathways can be targeted for return to homeostasis in hopes of coaxing microglia into clearing neurotoxic misfolded proteins.
Collapse
|
40
|
Liu J, Wang F. Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular Mechanisms and Therapeutic Implications. Front Immunol 2017; 8:1005. [PMID: 28871262 PMCID: PMC5567007 DOI: 10.3389/fimmu.2017.01005] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/07/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects upper motor neurons (MNs) comprising the corticospinal tract and lower MNs arising from the brain stem nuclei and ventral roots of the spinal cord, leading to fatal paralysis. Currently, there are no effective therapies for ALS. Increasing evidence indicates that neuroinflammation plays an important role in ALS pathogenesis. The neuroinflammation in ALS is characterized by infiltration of lymphocytes and macrophages, activation of microglia and reactive astrocytes, as well as the involvement of complement. In this review, we focus on the key cellular players of neuroinflammation during the pathogenesis of ALS by discussing not only their detrimental roles but also their immunomodulatory actions. We will summarize the pharmacological therapies for ALS that target neuroinflammation, as well as recent advances in the field of stem cell therapy aimed at modulating the inflammatory environment to preserve the remaining MNs in ALS patients and animal models of the disease.
Collapse
Affiliation(s)
- Jia Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
41
|
Complement C5a-C5aR1 signalling drives skeletal muscle macrophage recruitment in the hSOD1 G93A mouse model of amyotrophic lateral sclerosis. Skelet Muscle 2017; 7:10. [PMID: 28571586 PMCID: PMC5455206 DOI: 10.1186/s13395-017-0128-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/22/2017] [Indexed: 11/15/2022] Open
Abstract
Background The terminal pathway of the innate immune complement system is implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS). Terminal complement activation leads to generation of C5a, which through its receptor, C5aR1, drives immune cell recruitment and activation. Importantly, genetic or pharmacological blockage of C5aR1 improves motor performance and reduces disease pathology in hSOD1G93A rodent models of ALS. In this study, we aimed to explore the potential mechanisms of C5aR1-mediated pathology in hSOD1G93A mice by examining their skeletal muscles. Results We found elevated levels of C1qB, C4, fB, C3, C5a, and C5aR1 in tibialis anterior muscles of hSOD1G93A mice, which increased with disease progression. Macrophage cell numbers also progressively increased in hSOD1G93A muscles in line with disease progression. Immuno-localisation demonstrated that C5aR1 was expressed predominantly on macrophages within hSOD1G93A skeletal muscles. Notably, hSOD1G93A × C5aR1-/- mice showed markedly decreased numbers of infiltrating macrophages, along with reduced neuromuscular denervation and improved grip strength in hind limb skeletal muscles, when compared to hSOD1G93A mice. Conclusion These results indicate that terminal complement activation and C5a production occur in skeletal muscle tissue of hSOD1G93A mice, and that C5a-C5aR1 signalling contributes to the recruitment of macrophages that may accelerate muscle denervation in these ALS mice.
Collapse
|
42
|
Imamura K, Izumi Y, Watanabe A, Tsukita K, Woltjen K, Yamamoto T, Hotta A, Kondo T, Kitaoka S, Ohta A, Tanaka A, Watanabe D, Morita M, Takuma H, Tamaoka A, Kunath T, Wray S, Furuya H, Era T, Makioka K, Okamoto K, Fujisawa T, Nishitoh H, Homma K, Ichijo H, Julien JP, Obata N, Hosokawa M, Akiyama H, Kaneko S, Ayaki T, Ito H, Kaji R, Takahashi R, Yamanaka S, Inoue H. The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci Transl Med 2017; 9:eaaf3962. [PMID: 28539470 DOI: 10.1126/scitranslmed.aaf3962] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 02/04/2016] [Accepted: 12/13/2016] [Indexed: 03/07/2024]
Abstract
Amyotrophic lateral sclerosis (ALS), a fatal disease causing progressive loss of motor neurons, still has no effective treatment. We developed a phenotypic screen to repurpose existing drugs using ALS motor neuron survival as readout. Motor neurons were generated from induced pluripotent stem cells (iPSCs) derived from an ALS patient with a mutation in superoxide dismutase 1 (SOD1). Results of the screen showed that more than half of the hits targeted the Src/c-Abl signaling pathway. Src/c-Abl inhibitors increased survival of ALS iPSC-derived motor neurons in vitro. Knockdown of Src or c-Abl with small interfering RNAs (siRNAs) also rescued ALS motor neuron degeneration. One of the hits, bosutinib, boosted autophagy, reduced the amount of misfolded mutant SOD1 protein, and attenuated altered expression of mitochondrial genes. Bosutinib also increased survival in vitro of ALS iPSC-derived motor neurons from patients with sporadic ALS or other forms of familial ALS caused by mutations in TAR DNA binding protein (TDP-43) or repeat expansions in C9orf72 Furthermore, bosutinib treatment modestly extended survival of a mouse model of ALS with an SOD1 mutation, suggesting that Src/c-Abl may be a potentially useful target for developing new drugs to treat ALS.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yuishin Izumi
- Department of Clinical Neuroscience, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Kayoko Tsukita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Shiho Kitaoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Akito Tanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Dai Watanabe
- Department of Biological Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Hiroshi Takuma
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Tilo Kunath
- MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Selina Wray
- Department of Molecular Neuroscience, University College London Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Hirokazu Furuya
- Department of Neurology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Kouki Makioka
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Koichi Okamoto
- Geriatrics Research Institute and Hospital, Maebashi 371-0847, Japan
| | - Takao Fujisawa
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideki Nishitoh
- Department of Medical Sciences, University of Miyazaki, Miyazaki 889-1601, Japan
| | - Kengo Homma
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hidenori Ichijo
- Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jean-Pierre Julien
- Department of Psychiatry and Neurosciences, Research Centre of Mental Health Institute of Quebec (IUSMQ), Laval University, Québec, Canada
| | - Nanako Obata
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masato Hosokawa
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruhiko Akiyama
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Satoshi Kaneko
- Department of Neurology, Kansai Medical University, Hirakata 573-1191, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Kimiidera, Wakayama 641-8509, Japan
| | - Ryuji Kaji
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
43
|
Lee JD, Kumar V, Fung JNT, Ruitenberg MJ, Noakes PG, Woodruff TM. Pharmacological inhibition of complement C5a-C5a 1 receptor signalling ameliorates disease pathology in the hSOD1 G93A mouse model of amyotrophic lateral sclerosis. Br J Pharmacol 2017; 174:689-699. [PMID: 28128456 DOI: 10.1111/bph.13730] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing motor neuron disease without effective treatment. The complement system is up-regulated in ALS, with recent studies indicating that the activation product C5a accelerates disease progression via the C5a1 receptor (C5aR1). We therefore examined the therapeutic effect of C5a1 receptor antagonism in hSOD1G93A mice, the most widely used preclinical model of ALS. EXPERIMENTAL APPROACH The selective and orally active C5a1 receptor antagonist, PMX205, was administered to hSOD1G93A mice in drinking water, both pre- and post-disease onset. Blood, brain and spinal cord pharmacokinetics were performed using LC-MS/MS methods. Effects of PMX205 on hSOD1G93A disease progression was determined using body weight, hindlimb grip strength, survival time and blood analysis. KEY RESULTS PMX205 entered the intact CNS at pharmacologically active concentrations, with increased entry observed in hSOD1G93A mice as the disease progressed, in line with augmented blood-brain barrier breakdown. hSOD1G93A mice treated with PMX205 before disease onset had significantly improved hindlimb grip strength, slower disease progression and extended survival, compared with vehicle treatment. These improvements were associated with reductions in pro-inflammatory monocytes and granulocytes and increases in T-helper lymphocytes in peripheral blood. PMX205 treatment beginning 3 weeks following disease onset also attenuated disease progression, significantly extending survival. CONCLUSION AND IMPLICATIONS These results confirm that C5a1 receptors play a pathogenic role in hSOD1G93A mice, further validating the C5a-C5a1 receptor signalling axis as a potential therapeutic target to slow disease progression in ALS.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia.,University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Qld, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia
| | - Jenny N T Fung
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia.,Trauma, Critical Care and Recovery, Brisbane Diamantina Health Partners, The University of Queensland, Brisbane, Qld, Australia
| | - Peter G Noakes
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
44
|
Alshikho MJ, Zürcher NR, Loggia ML, Cernasov P, Chonde DB, Izquierdo Garcia D, Yasek JE, Akeju O, Catana C, Rosen BR, Cudkowicz ME, Hooker JM, Atassi N. Glial activation colocalizes with structural abnormalities in amyotrophic lateral sclerosis. Neurology 2016; 87:2554-2561. [PMID: 27837005 PMCID: PMC5207001 DOI: 10.1212/wnl.0000000000003427] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/30/2016] [Indexed: 11/24/2022] Open
Abstract
Objective: In this cross-sectional study, we aimed to evaluate brain structural abnormalities in relation to glial activation in the same cohort of participants. Methods: Ten individuals with amyotrophic lateral sclerosis (ALS) and 10 matched healthy controls underwent brain imaging using integrated MR/PET and the radioligand [11C]-PBR28. Diagnosis history and clinical assessments including Upper Motor Neuron Burden Scale (UMNB) were obtained from patients with ALS. Diffusion tensor imaging (DTI) analyses including tract-based spatial statistics and tractography were applied. DTI metrics including fractional anisotropy (FA) and diffusivities (mean, axial, and radial) were measured in regions of interest. Cortical thickness was assessed using surface-based analysis. The locations of structural changes, measured by DTI and the areas of cortical thinning, were compared to regional glial activation measured by relative [11C]-PBR28 uptake. Results: In this cohort of individuals with ALS, reduced FA and cortical thinning colocalized with regions demonstrating higher radioligand binding. [11C]-PBR28 binding in the left motor cortex was correlated with FA (r = −0.68, p < 0.05) and cortical thickness (r = −0.75, p < 0.05). UMNB was correlated with glial activation (r = +0.75, p < 0.05), FA (r = −0.77, p < 0.05), and cortical thickness (r = −0.75, p < 0.05) in the motor cortex. Conclusions: Increased uptake of the glial marker [11C]-PBR28 colocalizes with changes in FA and cortical thinning. This suggests a link between disease mechanisms (gliosis and inflammation) and structural changes (cortical thinning and white and gray matter changes). In this multimodal neuroimaging work, we provide an in vivo model to investigate the pathogenesis of ALS.
Collapse
Affiliation(s)
- Mohamad J Alshikho
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Nicole R Zürcher
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Marco L Loggia
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Paul Cernasov
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Daniel B Chonde
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - David Izquierdo Garcia
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Julia E Yasek
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Oluwaseun Akeju
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Ciprian Catana
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Bruce R Rosen
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Merit E Cudkowicz
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Jacob M Hooker
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown.
| | - Nazem Atassi
- From A.A. Martinos Center for Biomedical Imaging, Department of Radiology (M.J.A., N.R.Z., M.L.L., D.B.C., D.I.G., C.C., B.R.R., J.M.H.), Neurological Clinical Research Institute, Department of Neurology (M.J.A., P.C., J.E.Y., M.E.C., N.A.), and Department of Anesthesiology (O.A.), Massachusetts General Hospital, Harvard Medical School, Charlestown.
| |
Collapse
|
45
|
Abstract
The complement system is a major component of innate immunity and a potent driver of inflammation. It has key roles in host defense against pathogens but can also contribute to pathology by driving inflammation and cell damage in diverse diseases. Complement has emerged as an important factor in the pathogenesis of numerous diseases of the CNS and PNS, including infectious, autoimmune and degenerative disorders, and is increasingly implicated in neuropsychiatric disease. Establishing the roles and relevance of complement in disease pathogenesis has become ever more important in recent years as new drugs targeting the complement system have reached the clinic, and the potential for using complement analytes as disease biomarkers has been recognized. In this brief review, the author summarizes the evidence implicating complement in these diseases and outlines ways in which this new understanding can be used to aid diagnosis and improve outcome.
Collapse
Affiliation(s)
- Bryan Paul Morgan
- a Institute of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff CF144XN, UK
| |
Collapse
|
46
|
Affiliation(s)
- Fumito Endo
- Department of Neuroscience and Pathobiology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| |
Collapse
|
47
|
Bahia El Idrissi N, Bosch S, Ramaglia V, Aronica E, Baas F, Troost D. Complement activation at the motor end-plates in amyotrophic lateral sclerosis. J Neuroinflammation 2016; 13:72. [PMID: 27056040 PMCID: PMC4823861 DOI: 10.1186/s12974-016-0538-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a fatal progressive neurodegenerative disease with no available therapy. Components of the innate immune system are activated in the spinal cord and central nervous system of ALS patients. Studies in the SOD1G93A mouse show deposition of C1q and C3/C3b at the motor end-plate before neurological symptoms are apparent, suggesting that complement activation precedes neurodegeneration in this model. To obtain a better understanding of the role of complement at the motor end-plates in human ALS pathology, we analyzed post-mortem tissue of ALS donors for complement activation and its regulators. Methods Post-mortem intercostal muscle biopsies were collected at autopsy from ALS (n = 11) and control (n = 6) donors. The samples were analyzed for C1q, membrane attack complex (MAC), CD55, and CD59 on the motor end-plates, using immunofluorescence or immunohistochemistry. Results Here, we show that complement activation products and regulators are deposited on the motor end-plates of ALS patients. C1q co-localized with neurofilament in the intercostal muscle of ALS donors and was absent in controls (P = 0.001). In addition, C1q was found deposited on the motor end-plates in the intercostal muscle. MAC was also found deposited on motor end-plates that were innervated by nerves in the intercostal muscle of ALS donors but not in controls (P = 0.001). High levels of the regulators CD55 and CD59 were detected at the motor end-plates of ALS donors but not in controls, suggesting an attempt to counteract complement activation and prevent MAC deposition on the end-plates before they are lost. Conclusions This study provides evidence that complement activation products are deposited on innervated motor end-plates in the intercostal muscle of ALS donors, indicating that complement activation may precede end-plate denervation in human ALS. This study adds to the understanding of ALS pathology in man and identifies complement as a potential modifier of the disease process. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0538-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nawal Bahia El Idrissi
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Sanne Bosch
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Valeria Ramaglia
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Eleonora Aronica
- Department of Neuropathology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands.
| | - Dirk Troost
- Department of Neuropathology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| |
Collapse
|
48
|
Brennan FH, Lee JD, Ruitenberg MJ, Woodruff TM. Therapeutic targeting of complement to modify disease course and improve outcomes in neurological conditions. Semin Immunol 2016; 28:292-308. [PMID: 27049459 DOI: 10.1016/j.smim.2016.03.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 12/14/2022]
Abstract
The recognition that complement proteins are abundantly present and can have pathological roles in neurological conditions offers broad scope for therapeutic intervention. Accordingly, an increasing number of experimental investigations have explored the potential of harnessing the unique activation pathways, proteases, receptors, complexes, and natural inhibitors of complement, to mitigate pathology in acute neurotrauma and chronic neurodegenerative diseases. Here, we review mechanisms of complement activation in the central nervous system (CNS), and explore the effects of complement inhibition in cerebral ischemic-reperfusion injury, traumatic brain injury, spinal cord injury, Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. We consider the challenges and opportunities arising from these studies. As complement therapies approach clinical translation, we provide perspectives on how promising complement-targeted therapeutics could become part of novel and effective future treatment options to improve outcomes in the initiation and progression stages of these debilitating CNS disorders.
Collapse
Affiliation(s)
- Faith H Brennan
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia; Trauma, Critical Care and Recovery, Brisbane Diamantina Health Partners, The University of Queensland, Brisbane 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
49
|
Nardo G, Trolese MC, Tortarolo M, Vallarola A, Freschi M, Pasetto L, Bonetto V, Bendotti C. New Insights on the Mechanisms of Disease Course Variability in ALS from Mutant SOD1 Mouse Models. Brain Pathol 2016; 26:237-47. [PMID: 26780365 PMCID: PMC8029191 DOI: 10.1111/bpa.12351] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 01/14/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a heterogeneous disease in terms of progression rate and survival. This is probably one of the reasons for the failure of many clinical trials and the lack of effective therapies. Similar variability is also seen in SOD1(G93A) mouse models based on their genetic background. For example, when the SOD1(G93A) transgene is expressed in C57BL6 background the phenotype is mild with slower disease progression than in the 129Sv mice expressing the same amount of transgene but showing faster progression and shorter lifespan. This review summarizes and discusses data obtained from the analysis of these two mouse models under different aspects such as the motor phenotype, neuropathological alterations in the central nervous system (CNS) and peripheral nervous system (PNS) and the motor neuron autonomous and non-cell autonomous mechanisms with the aim of finding elements to explain the different rates of disease progression. We also discuss the identification of promising prognostic biomarkers by comparative analysis of the two ALS mouse models. This analysis might possibly suggest new strategies for effective therapeutic intervention in ALS to slow significantly or even block the course of the disease.
Collapse
Affiliation(s)
- Giovanni Nardo
- Department of NeuroscienceLaboratory Molecular Neurobiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| | - Maria Chiara Trolese
- Department of NeuroscienceLaboratory Molecular Neurobiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| | - Massimo Tortarolo
- Department of NeuroscienceLaboratory Molecular Neurobiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| | - Antonio Vallarola
- Department of NeuroscienceLaboratory Molecular Neurobiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| | - Mattia Freschi
- Department of NeuroscienceLaboratory Molecular Neurobiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
- Animal Facility, AriSLA, Fondazione Italiana di ricerca per la Sclerosi Laterale Amiotrofica
| | - Laura Pasetto
- Department of Molecular Biochemistry and Pharmacology, Laboratory of Translational ProteomicsIRCCS‐Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| | - Valentina Bonetto
- Department of Molecular Biochemistry and Pharmacology, Laboratory of Translational ProteomicsIRCCS‐Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| | - Caterina Bendotti
- Department of NeuroscienceLaboratory Molecular Neurobiology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”MilanoItaly
| |
Collapse
|
50
|
Lasiene J, Komine O, Fujimori-Tonou N, Powers B, Endo F, Watanabe S, Shijie J, Ravits J, Horner P, Misawa H, Yamanaka K. Neuregulin 1 confers neuroprotection in SOD1-linked amyotrophic lateral sclerosis mice via restoration of C-boutons of spinal motor neurons. Acta Neuropathol Commun 2016; 4:15. [PMID: 26891847 PMCID: PMC4758105 DOI: 10.1186/s40478-016-0286-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/25/2022] Open
Abstract
Introduction Increasing evidence implicates the role of the cell types surrounding motor neurons, such as interneurons and glial cells, in non-cell autonomous neurodegeneration of amyotrophic lateral sclerosis (ALS). C-boutons, the large cholinergic synapses that innervate spinal α-motor neurons to control their excitability, are progressively lost from motor neurons in both human ALS and mutant Cu/Zn superoxide dismutase 1 (SOD1)-ALS mice. Neuregulin-1 (NRG1), a trophic factor implicated in neural development, transmission, and synaptic plasticity, has been reported to localize in the synapse of C-boutons. However, the roles of NRG1 in maintenance of motor neuron health and activity, as well as the functional consequences of its alteration in motor neuron disease, are not fully understood. Results NRG1 was localized to the post-synaptic face of C-boutons and its expression was significantly lost in SOD1-ALS mice and human ALS patients. Losses of NRG1 expression and C-boutons occured almost contemporaneously in SOD1-ALS mice. In addition, expressions of ErbB3 and ErbB4, receptors for NRG1, were reduced in the motor neurons of SOD1-ALS mice. Furthermore, viral-mediated delivery of type III-NRG1 to the spinal cord restored the number of C-boutons and extended the survival time of SOD1-ALS mice. Conclusions These results suggest that maintenance of NRG1-ErbB4/3 axis by supplementation of NRG1 confers neuroprotection in motor neuron disease, partly through the maintenance of C-boutons of spinal motor neurons. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0286-7) contains supplementary material, which is available to authorized users.
Collapse
|