101
|
Cruz-Haces M, Tang J, Acosta G, Fernandez J, Shi R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl Neurodegener 2017; 6:20. [PMID: 28702179 PMCID: PMC5504572 DOI: 10.1186/s40035-017-0088-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury is among the most common causes of death and disability in youth and young adults. In addition to the acute risk of morbidity with moderate to severe injuries, traumatic brain injury is associated with a number of chronic neurological and neuropsychiatric sequelae including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. However, despite the high incidence of traumatic brain injuries and the established clinical correlation with neurodegeneration, the causative factors linking these processes have not yet been fully elucidated. Apart from removal from activity, few, if any prophylactic treatments against post-traumatic brain injury neurodegeneration exist. Therefore, it is imperative to understand the pathophysiological mechanisms of traumatic brain injury and neurodegeneration in order to identify potential factors that initiate neurodegenerative processes. Oxidative stress, neuroinflammation, and glutamatergic excitotoxicity have previously been implicated in both secondary brain injury and neurodegeneration. In particular, reactive oxygen species appear to be key in mediating molecular insult in neuroinflammation and excitotoxicity. As such, it is likely that post injury oxidative stress is a key mechanism which links traumatic brain injury to increased risk of neurodegeneration. Consequently, reactive oxygen species and their subsequent byproducts may serve as novel fluid markers for identification and monitoring of cellular damage. Furthermore, these reactive species may further serve as a suitable therapeutic target to reduce the risk of post-injury neurodegeneration and provide long term quality of life improvements for those suffering from traumatic brain injury.
Collapse
Affiliation(s)
- Marcela Cruz-Haces
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Jonathan Tang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Glen Acosta
- Department of Basic Medical Sciences, Purdue University, West Lafayette, USA
| | - Joseph Fernandez
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Riyi Shi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
- Department of Basic Medical Sciences, Purdue University, West Lafayette, USA
| |
Collapse
|
102
|
Volkman R, Offen D. Concise Review: Mesenchymal Stem Cells in Neurodegenerative Diseases. Stem Cells 2017; 35:1867-1880. [PMID: 28589621 DOI: 10.1002/stem.2651] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/06/2017] [Indexed: 12/13/2022]
Abstract
Stem cell-based therapies for neurodegenerative diseases aim at halting clinical deterioration by regeneration and by providing local support for damaged tissue. Mesenchymal stem cells (MSCs) hold great potential for cell therapy as they can be efficiently derived from adult tissue, ex vivo expanded in culture and safely transplanted autologously. MSCs were also shown to be able to differentiate toward neural fates and to secrete a broad range of factors able to promote nervous tissue maintenance and repair. Moreover, upon transplantation, MSCs were shown capable of homing toward lesioned areas, implying their potential use as vehicles for therapeutic agents administration. Indeed, various advantageous effects were reported following human MSCs transplantation into rodent models of neurodegenerative diseases, such as neurotrophic factor-mediated protection, enhanced neurogenesis, modulation of inflammation, and abnormal protein aggregate clearance. Per journal style, most nonstandard abbreviations must be used at least two times in the abstract to be retained; NTF was used once and thus has been deleted. Recent studies have also used ex vivo manipulation for enhanced expression of potentially favorable factors, by so exploiting the homing capacity of MSCs for effective expression at the lesion site. Here, we will summarize current advancements in MSCs-based therapies for neurodegenerative diseases. We will examine the roles of central mechanisms suggested to mediate the beneficial effects of MSCs-based therapy and consider the augmentation of these mechanisms for superior clinical outcomes in rodent models of neurodegeneration as well as in clinical trials. Stem Cells 2017;35:1867-1880.
Collapse
|
103
|
Jara JH, Genç B, Stanford MJ, Pytel P, Roos RP, Weintraub S, Mesulam MM, Bigio EH, Miller RJ, Özdinler PH. Evidence for an early innate immune response in the motor cortex of ALS. J Neuroinflammation 2017. [PMID: 28651542 PMCID: PMC5485686 DOI: 10.1186/s12974-017-0896-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Recent evidence indicates the importance of innate immunity and neuroinflammation with microgliosis in amyotrophic lateral sclerosis (ALS) pathology. The MCP1 (monocyte chemoattractant protein-1) and CCR2 (CC chemokine receptor 2) signaling system has been strongly associated with the innate immune responses observed in ALS patients, but the motor cortex has not been studied in detail. Methods After revealing the presence of MCP1 and CCR2 in the motor cortex of ALS patients, to elucidate, visualize, and define the timing, location and the extent of immune response in relation to upper motor neuron vulnerability and progressive degeneration in ALS, we developed MCP1-CCR2-hSOD1G93A mice, an ALS reporter line, in which cells expressing MCP1 and CCR2 are genetically labeled by monomeric red fluorescent protein-1 and enhanced green fluorescent protein, respectively. Results In the motor cortex of MCP1-CCR2-hSOD1G93A mice, unlike in the spinal cord, there was an early increase in the numbers of MCP1+ cells, which displayed microglial morphology and selectively expressed microglia markers. Even though fewer CCR2+ cells were present throughout the motor cortex, they were mainly infiltrating monocytes. Interestingly, MCP1+ cells were found in close proximity to the apical dendrites and cell bodies of corticospinal motor neurons (CSMN), further implicating the importance of their cellular interaction to neuronal pathology. Similar findings were observed in the motor cortex of ALS patients, where MCP1+ microglia were especially in close proximity to the degenerating apical dendrites of Betz cells. Conclusions Our findings reveal that the intricate cellular interplay between immune cells and upper motor neurons observed in the motor cortex of ALS mice is indeed recapitulated in ALS patients. We generated and characterized a novel model system, to study the cellular and molecular basis of this close cellular interaction and how that relates to motor neuron vulnerability and progressive degeneration in ALS. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0896-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Javier H Jara
- Department of Neurology and Clinical Neurological Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 10-120, Chicago, IL, 60611, USA.
| | - Barış Genç
- Department of Neurology and Clinical Neurological Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 10-120, Chicago, IL, 60611, USA
| | - Macdonell J Stanford
- Department of Neurology and Clinical Neurological Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 10-120, Chicago, IL, 60611, USA
| | - Peter Pytel
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Raymond P Roos
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Sandra Weintraub
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Chicago, IL, 60611, USA
| | - M Marsel Mesulam
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Chicago, IL, 60611, USA
| | - Eileen H Bigio
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Chicago, IL, 60611, USA
| | - Richard J Miller
- Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - P Hande Özdinler
- Department of Neurology and Clinical Neurological Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 10-120, Chicago, IL, 60611, USA. .,Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Chicago, IL, 60611, USA. .,Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
104
|
Therapeutic Strategies Under Development Targeting Inflammatory Mechanisms in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2017; 55:2789-2813. [PMID: 28455693 DOI: 10.1007/s12035-017-0532-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
|
105
|
Garbuzova-Davis S, Kurien C, Thomson A, Falco D, Ahmad S, Staffetti J, Steiner G, Abraham S, James G, Mahendrasah A, Sanberg PR, Borlongan CV. Endothelial and Astrocytic Support by Human Bone Marrow Stem Cell Grafts into Symptomatic ALS Mice towards Blood-Spinal Cord Barrier Repair. Sci Rep 2017; 7:884. [PMID: 28408761 PMCID: PMC5429840 DOI: 10.1038/s41598-017-00993-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/17/2017] [Indexed: 12/11/2022] Open
Abstract
Vascular pathology, including blood-CNS barrier (B-CNS-B) damage via endothelial cell (EC) degeneration, is a recently recognized hallmark of Amyotrophic Lateral Sclerosis (ALS) pathogenesis. B-CNS-B repair may be a new therapeutic approach for ALS. This study aimed to determine effects of transplanted unmodified human bone marrow CD34+ (hBM34+) cells into symptomatic G93A mice towards blood-spinal cord barrier (BSCB) repair. Thirteen weeks old G93A mice intravenously received one of three different doses of hBM34+ cells. Cell-treated, media-treated, and control mice were euthanized at 17 weeks of age. Immunohistochemical (anti-human vWF, CD45, GFAP, and Iba-1) and motor neuron histological analyses were performed in cervical and lumbar spinal cords. EB levels in spinal cord parenchyma determined capillary permeability. Transplanted hBM34+ cells improved behavioral disease outcomes and enhanced motor neuron survival, mainly in high-cell-dose mice. Transplanted cells differentiated into ECs and engrafted within numerous capillaries. Reduced astrogliosis, microgliosis, and enhanced perivascular end-feet astrocytes were also determined in spinal cords, mostly in high-cell-dose mice. These mice also showed significantly decreased parenchymal EB levels. EC differentiation, capillary engraftment, reduced capillary permeability, and re-established perivascular end-feet astrocytes in symptomatic ALS mice may represent BSCB repair processes, supporting hBM34+ cell transplantation as a future therapeutic strategy for ALS patients.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America. .,Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America. .,Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America.
| | - Crupa Kurien
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Avery Thomson
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Dimitri Falco
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Sohaib Ahmad
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Joseph Staffetti
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - George Steiner
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Sophia Abraham
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Greeshma James
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Ajay Mahendrasah
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Paul R Sanberg
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America.,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America.,Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America.,Department of Psychiatry, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Cesario V Borlongan
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America.,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| |
Collapse
|
106
|
Yin L, Dai Q, Jiang P, Zhu L, Dai H, Yao Z, Liu H, Ma X, Qu L, Jiang J. Manganese exposure facilitates microglial JAK2-STAT3 signaling and consequent secretion of TNF-a and IL-1β to promote neuronal death. Neurotoxicology 2017; 64:195-203. [PMID: 28385490 DOI: 10.1016/j.neuro.2017.04.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/02/2017] [Accepted: 04/03/2017] [Indexed: 12/31/2022]
Abstract
Chronic manganese (Mn) exposure can lead to neuroinflammation and neurological deficit, which resemble idiopathic Parkinson's disease (IPD). However, the precise mechanisms underlying Mn exposure-induced neurotoxicity remain incompletely understood. Microglia can become hyperactivated and plays a vital role in neuroinflammation and consequent neurodegeneration in response to pro-inflammatory stimuli. In the present study, we found that HAPI microglial cells exhibited increased secretion of pro-inflammatory TNF-α and IL-1β following Mn exposure in dose- and time-dependent manners. In addition, we showed that Mn exposure could trigger the activation of JAK2/STAT3 signaling pathway in microglia. Notably, Mn-induced secretion of TNF-α and IL-1β was significantly attenuated by the treatment of JAK2 inhibitor. Finally, through incubating PC12 neuronal cells with Mn-treated microglial conditioned medium, we demonstrated that Mn-induced secretion of microglial TNF-α and IL-1β facilitated neuronal apoptosis. Thus, we speculate that Mn exposure might trigger JAK2-STAT3 signal pathway in microglia, leading to resultant neuroinflammation and neuronal loss.
Collapse
Affiliation(s)
- Lifeng Yin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Qijun Dai
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Peipei Jiang
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Lin Zhu
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Haifeng Dai
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Zhigang Yao
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Hua Liu
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Xiaoping Ma
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China
| | - Lianxia Qu
- Department of Encephalopathy, Traditional Chinese Medicine Hospital of Haian County Affiliated Hospital, Nanjing University of Chinese Medicine of Hanlin College, Nantong, Jiangsu 226600, People's Republic of China.
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
107
|
Genung NE, Guckian KM. Small Molecule Inhibition of Interleukin-1 Receptor-Associated Kinase 4 (IRAK4). PROGRESS IN MEDICINAL CHEMISTRY 2017; 56:117-163. [PMID: 28314411 DOI: 10.1016/bs.pmch.2016.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In recent years, interleukin-1 receptor-associated kinase 4, IRAK4, has become an attractive target for many medicinal chemistry programmes. Target inhibition is of potential therapeutic value in areas including autoimmune disorders, cancer, inflammatory diseases, and possibly neurodegenerative diseases. Results from high-throughput screening efforts have led, in conjunction with structure-based drug design, to the identification of highly potent and selective small molecule IRAK4 inhibitors from many diverse chemical series. In vitro and in vivo studies with entities from distinct structural classes have helped elucidate the downstream pharmacological responses associated with IRAK4 inhibition as a proof of concept in disease models, leading to the recent initiation of human clinical trials. Within this review, we will highlight the considerable effort by numerous groups dedicated to the development of small molecule IRAK4 inhibitors for the treatment of human disease.
Collapse
|
108
|
Lee JM, Tan V, Lovejoy D, Braidy N, Rowe DB, Brew BJ, Guillemin GJ. Involvement of quinolinic acid in the neuropathogenesis of amyotrophic lateral sclerosis. Neuropharmacology 2017; 112:346-364. [DOI: 10.1016/j.neuropharm.2016.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
|
109
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a highly heterogeneous disease with no effective treatment. Drug development has been hampered by the lack of biomarkers that aid in early diagnosis, demonstrate target engagement, monitor disease progression, and can serve as surrogate endpoints to assess the efficacy of treatments. Fluid-based biomarkers may potentially address these issues. An ideal biomarker should exhibit high specificity and sensitivity for distinguishing ALS from control (appropriate disease mimics and other neurologic diseases) populations and monitor disease progression within individual patients. Significant progress has been made using cerebrospinal fluid, serum, and plasma in the search for ALS biomarkers, with urine and saliva biomarkers still in earlier stages of development. A few of these biomarker candidates have demonstrated use in patient stratification, predicting disease course (fast vs slow progression) and severity, or have been used in preclinical and clinical applications. However, while ALS biomarker discovery has seen tremendous advancements in the last decade, validating biomarkers and moving them towards the clinic remains more elusive. In this review, we highlight biomarkers that are moving towards clinical utility and the challenges that remain in order to implement biomarkers at all stages of the ALS drug development process.
Collapse
Affiliation(s)
- Lucas T Vu
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA
| | - Robert Bowser
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA.
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA.
| |
Collapse
|
110
|
Tefera TW, Tan KN, McDonald TS, Borges K. Alternative Fuels in Epilepsy and Amyotrophic Lateral Sclerosis. Neurochem Res 2016; 42:1610-1620. [PMID: 27868154 DOI: 10.1007/s11064-016-2106-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/12/2022]
Abstract
This review summarises the recent findings on metabolic treatments for epilepsy and Amyotrophic Lateral Sclerosis (ALS) in honour of Professor Ursula Sonnewald. The metabolic impairments in rodent models of these disorders as well as affected patients are being discussed. In both epilepsy and ALS, there are defects in glucose uptake and reduced tricarboxylic acid (TCA) cycling, at least in part due to reduced amounts of C4 TCA cycle intermediates. In addition there are impairments in glycolysis in ALS. A reduction in glucose uptake can be addressed by providing the brain with alternative fuels, such as ketones or medium-chain triglycerides. As anaplerotic fuels, such as the triglyceride of heptanoate, triheptanoin, refill the TCA cycle C4/C5 intermediate pool that is deficient, they are ideal to boost TCA cycling and thus the oxidative metabolism of all fuels.
Collapse
Affiliation(s)
- Tesfaye W Tefera
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia
| | - Kah Ni Tan
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia
| | - Tanya S McDonald
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia
| | - Karin Borges
- Department of Pharmacology, School of Biomedical Sciences, The University of Queensland, Skerman Building 65, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
111
|
Won YH, Lee MY, Choi YC, Ha Y, Kim H, Kim DY, Kim MS, Yu JH, Seo JH, Kim M, Cho SR, Kang SW. Elucidation of Relevant Neuroinflammation Mechanisms Using Gene Expression Profiling in Patients with Amyotrophic Lateral Sclerosis. PLoS One 2016; 11:e0165290. [PMID: 27812125 PMCID: PMC5094695 DOI: 10.1371/journal.pone.0165290] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 10/10/2016] [Indexed: 11/19/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by damage of motor neurons. Recent reports indicate that inflammatory responses occurring within the central nervous system contribute to the pathogenesis of ALS. We aimed to investigate disease-specific gene expression associated with neuroinflammation by conducting transcriptome analysis on fibroblasts from three patients with sporadic ALS and three normal controls. Several pathways were found to be upregulated in patients with ALS, among which the toll-like receptor (TLR) and NOD-like receptor (NLR) signaling pathways are related to the immune response. Genes—toll-interacting protein (TOLLIP), mitogen-activated protein kinase 9 (MAPK9), interleukin-1β (IL-1β), interleukin-8 (IL-8), and chemokine (C-X-C motif) ligand 1 (CXCL1)—related to these two pathways were validated using western blotting. This study validated the genes that are associated with TLR and NLR signaling pathways from different types of patient-derived cells. Not only fibroblasts but also induced pluripotent stem cells (iPSCs) and neural rosettes from the same origins showed similar expression patterns. Furthermore, expression of TOLLIP, a regulator of TLR signaling pathway, decreased with cellular aging as judged by changes in its expression through multiple passages. TOLLIP expression was downregulated in ALS cells under conditions of inflammation induced by lipopolysaccharide. Our data suggest that the TLR and NLR signaling pathways are involved in pathological innate immunity and neuroinflammation associated with ALS and that TOLLIP, MAPK9, IL-1β, IL-8, and CXCL1 play a role in ALS-specific immune responses. Moreover, changes of TOLLIP expression might be associated with progression of ALS.
Collapse
Affiliation(s)
- Yu Hui Won
- Department of Physical Medicine and Rehabilitation, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
- Department of Medicine, the Graduate School of Yonsei University, Seoul, Korea
| | - Min-Young Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Chul Choi
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Ha
- Department of Neurosurgery, Spine & Spinal Cord Institute, College of Medicine, Yonsei University, Seoul, Korea
| | - Hyongbum Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Do-Young Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Myung-Sun Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hea Yu
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Jung Hwa Seo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - MinGi Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- * E-mail: (SWK); (SRC)
| | - Seong-Woong Kang
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea
- Department of Medicine, the Graduate School of Yonsei University, Seoul, Korea
- * E-mail: (SWK); (SRC)
| |
Collapse
|
112
|
Miller ZA, Sturm VE, Camsari GB, Karydas A, Yokoyama JS, Grinberg LT, Boxer AL, Rosen HJ, Rankin KP, Gorno-Tempini ML, Coppola G, Geschwind DH, Rademakers R, Seeley WW, Graff-Radford NR, Miller BL. Increased prevalence of autoimmune disease within C9 and FTD/MND cohorts: Completing the picture. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e301. [PMID: 27844039 PMCID: PMC5087253 DOI: 10.1212/nxi.0000000000000301] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022]
Abstract
Objective: To determine the prevalence of autoimmune disease in symptomatic C9ORF72 (C9) mutation carriers and frontotemporal dementia with motor neuron disease (FTD/MND) cohorts. Methods: In this case-control study, we reviewed the clinical histories of 66 patients with FTD/MND and 57 symptomatic C9 carriers (24 overlapping cases), a total of 99 charts, for history of autoimmune disease. The prevalence of autoimmune disease in C9 and FTD/MND cohorts was determined by χ2 and Fisher exact comparisons between the combined C9 and FTD/MND group with normal control, Alzheimer disease, and progressive supranuclear palsy cohorts, as well as comparisons within C9 and FTD/MND cohorts. Results: Our combined C9 and FTD/MND cohort has a 12% prevalence of nonthyroid autoimmune disease. The prevalence of nonthyroid autoimmune disease in C9 and FTD/MND is similar to the rates in previously detailed progranulin and semantic variant primary progressive aphasia cohorts and elevated in comparison to previously collected normal control and typical Alzheimer disease cohorts, as well as a newly screened progressive supranuclear palsy cohort. Furthermore, the types of autoimmune disease in this combined C9 and FTD/MND cohort cluster within the same 3 categories previously described in progranulin and semantic variant primary progressive aphasia: inflammatory arthritides, cutaneous conditions, and gastrointestinal disorders. Conclusions: The association between selective autoimmune disease and neurodegenerative disorders unified by the underlying pathology frontotemporal lobar degeneration with TDP-43–positive inclusions (FTLD-TDP) extends to C9 and FTD/MND cohorts, providing further evidence that select autoimmune inflammation may be intrinsically linked to FTLD-TDP pathophysiology.
Collapse
Affiliation(s)
- Zachary A Miller
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Virginia E Sturm
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Gamze Balci Camsari
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Anna Karydas
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Jennifer S Yokoyama
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Lea T Grinberg
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Adam L Boxer
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Howard J Rosen
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Katherine P Rankin
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Giovanni Coppola
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Daniel H Geschwind
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Rosa Rademakers
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - William W Seeley
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Neill R Graff-Radford
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Bruce L Miller
- Memory and Aging Center (Z.A.M., V.E.S., A.K., J.S.Y., L.T.G., A.L.B., H.J.R., K.P.R., M.L.G.-T., W.W.S., B.L.M.) and Department of Pathology (L.T.G., W.W.S.), University of California, San Francisco; Departments of Neurology (G.B.C., N.R.G.-R.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Department of Neurology (G.C., D.H.G.), David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
113
|
Oxymatrine inhibits microglia activation via HSP60-TLR4 signaling. Biomed Rep 2016; 5:623-628. [PMID: 27882228 DOI: 10.3892/br.2016.776] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/16/2016] [Indexed: 12/30/2022] Open
Abstract
Oxymatrine (OMT) is an alkaloid extracted from Sophora flavescens, which has broad anti-inflammatory, antitumor and immunosuppressant actions. However, the underlying molecular mechanisms have remained elusive. Heat shock protein 60 (HSP60) has recently been shown to have an important role in autoimmune reactions. The present study aimed to investigate whether OMT exerts its anti-inflammatory effects by inhibiting microglial activation and examined the role of HSP60 in this process. Western blot analysis and ELISA showed that OMT decreased the expression and release of HSP60 by LPS-activated BV2 cells. The expression of heat shock factor 1, the transcription factor of HSP60, was also suppressed by OMT. Extracellular HSP60 has been previously indicated to induce microglial apoptosis through the Toll-like receptor (TLR)-4 pathway. Flow cytometric analysis demonstrated that LPS treatment induced apoptosis of BV2 cells, which was inhibited by OMT in parallel with inhibition of LPS-induced expression of TLR-4. Furthermore, OMT was shown to suppress the levels of myeloid differentiation factor (MYD)88, nuclear factor (NF)-κB, caspase-3, inducible nitric oxide synthase, tumor necrosis factor-α, interleukin (IL)-1β and IL-6. In light of these results, it was concluded that OMT may exert its neuroprotective effects via HSP60/TLR-4/MYD88/NF-κB signaling pathways to inhibit microglial activation. OMT may therefore offer substantial therapeutic potential for treating neurodegenerative diseases associated with microglial activation.
Collapse
|
114
|
Lee J, Hyeon SJ, Im H, Ryu H, Kim Y, Ryu H. Astrocytes and Microglia as Non-cell Autonomous Players in the Pathogenesis of ALS. Exp Neurobiol 2016; 25:233-240. [PMID: 27790057 PMCID: PMC5081469 DOI: 10.5607/en.2016.25.5.233] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder that leads to a progressive muscle wasting and paralysis. The pathological phenotypes are featured by severe motor neuron death and glial activation in the lumbar spinal cord. Proposed ALS pathogenic mechanisms include glutamate cytotoxicity, inflammatory pathway, oxidative stress, and protein aggregation. However, the exact mechanisms of ALS pathogenesis are not fully understood yet. Recently, a growing body of evidence provides a novel insight on the importance of glial cells in relation to the motor neuronal damage via the non-cell autonomous pathway. Accordingly, the aim of the current paper is to overview the role of astrocytes and microglia in the pathogenesis of ALS and to better understand the disease mechanism of ALS.
Collapse
Affiliation(s)
- Junghee Lee
- Veterans Affairs Boston Healthcare System, Boston, MA 02130, USA.; Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Seung Jae Hyeon
- Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 04535, Korea
| | - Hyeonjoo Im
- Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 04535, Korea
| | - Hyun Ryu
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Yunha Kim
- Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 04535, Korea
| | - Hoon Ryu
- Veterans Affairs Boston Healthcare System, Boston, MA 02130, USA.; Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA.; Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 04535, Korea
| |
Collapse
|
115
|
Triheptanoin Protects Motor Neurons and Delays the Onset of Motor Symptoms in a Mouse Model of Amyotrophic Lateral Sclerosis. PLoS One 2016; 11:e0161816. [PMID: 27564703 PMCID: PMC5001695 DOI: 10.1371/journal.pone.0161816] [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: 11/12/2015] [Accepted: 08/14/2016] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence that energy metabolism is disturbed in Amyotrophic Lateral Sclerosis (ALS) patients and animal models. Treatment with triheptanoin, the triglyceride of heptanoate, is a promising approach to provide alternative fuel to improve oxidative phosphorylation and aid ATP generation. Heptanoate can be metabolized to propionyl-CoA, which after carboxylation can produce succinyl-CoA and thereby re-fill the tricarboxylic acid (TCA) cycle (anaplerosis). Here we tested the hypothesis that treatment with triheptanoin prevents motor neuron loss and delays the onset of disease symptoms in female mice overexpressing the mutant human SOD1G93A (hSOD1G93A) gene. When oral triheptanoin (35% of caloric content) was initiated at P35, motor neuron loss at 70 days of age was attenuated by 33%. In untreated hSOD1G93A mice, the loss of hind limb grip strength began at 16.7 weeks. Triheptanoin maintained hind limb grip strength for 2.8 weeks longer (p<0.01). Loss of balance on the rotarod and reduction of body weight were delayed by 13 and 11 days respectively (both p<0.01). Improved motor function occurred in parallel with alterations in the expression of genes associated with muscle metabolism. In gastrocnemius muscles, the mRNA levels of pyruvate, 2-oxoglutarate and succinate dehydrogenases and methyl-malonyl mutase were reduced by 24–33% in 10 week old hSOD1G93A mice when compared to wild-type mice, suggesting that TCA cycling in skeletal muscle may be slowed in this ALS mouse model at a stage when muscle strength is still normal. At 25 weeks of age, mRNA levels of succinate dehydrogenases, glutamic pyruvic transaminase 2 and the propionyl carboxylase β subunit were reduced by 69–84% in control, but not in triheptanoin treated hSOD1G93A animals. Taken together, our results suggest that triheptanoin slows motor neuron loss and the onset of motor symptoms in ALS mice by improving TCA cycling.
Collapse
|
116
|
Saberi S, Stauffer JE, Schulte DJ, Ravits J. Neuropathology of Amyotrophic Lateral Sclerosis and Its Variants. Neurol Clin 2016; 33:855-76. [PMID: 26515626 DOI: 10.1016/j.ncl.2015.07.012] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neuropathologic molecular signature common to almost all sporadic amyotrophic lateral sclerosis (ALS) and most familial ALS is TDP-43 immunoreactive neuronal cytoplasmic inclusions. The neuropathologic and molecular neuropathologic features of ALS variants, primarily lateral sclerosis and progressive muscular atrophy, are less certain but also seem to share the primary features of ALS. Genetic causes, including mutations in SOD1, TDP-43, FUS, and C9orf72, all have distinctive molecular neuropathologic signatures. Neuropathology will continue to play an increasingly key role in solving the puzzle of ALS pathogenesis.
Collapse
Affiliation(s)
- Shahram Saberi
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA
| | - Jennifer E Stauffer
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA
| | - Derek J Schulte
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA
| | - John Ravits
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA.
| |
Collapse
|
117
|
Lam L, Chin L, Halder RC, Sagong B, Famenini S, Sayre J, Montoya D, Rubbi L, Pellegrini M, Fiala M. Epigenetic changes in T-cell and monocyte signatures and production of neurotoxic cytokines in ALS patients. FASEB J 2016; 30:3461-3473. [PMID: 27368295 DOI: 10.1096/fj.201600259rr] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/21/2016] [Indexed: 12/13/2022]
Abstract
We have investigated transcriptional and epigenetic differences in peripheral blood mononuclear cells (PBMCs) of monozygotic female twins discordant in the diagnosis of amyotrophic lateral sclerosis (ALS). Exploring DNA methylation differences by reduced representation bisulfite sequencing (RRBS), we determined that, over time, the ALS twin developed higher abundances of the CD14 macrophages and lower abundances of T cells compared to the non-ALS twin. Higher macrophage signature in the ALS twin was also shown by RNA sequencing (RNA-seq). Moreover, the twins differed in the methylome at loci near several genes, including EGFR and TNFRSF11A, and in the pathways related to the tretinoin and H3K27me3 markers. We also tested cytokine production by PBMCs. The ALS twin's PBMCs spontaneously produced IL-6 and TNF-α, whereas PBMCs of the healthy twin produced these cytokines only when stimulated by superoxide dismutase (SOD)-1. These results and flow cytometric detection of CD45 and CD127 suggest the presence of memory T cells in both twins, but effector T cells only in the ALS twin. The ALS twin's PBMC supernatants, but not the healthy twin's, were toxic to rat cortical neurons, and this toxicity was strongly inhibited by an IL-6 receptor antibody (tocilizumab) and less well by TNF-α and IL-1β antibodies. The putative neurotoxicity of IL-6 and TNF-α is in agreement with a high expression of these cytokines on infiltrating macrophages in the ALS spinal cord. We hypothesize that higher macrophage abundance and increased neurotoxic cytokines have a fundamental role in the phenotype and treatment of certain individuals with ALS.-Lam, L., Chin, L., Halder, R. C., Sagong, B., Famenini, S., Sayre, J., Montoya, D., Rubbi L., Pellegrini, M., Fiala, M. Epigenetic changes in T-cell and monocyte signatures and production of neurotoxic cytokines in ALS patients.
Collapse
Affiliation(s)
- Larry Lam
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Lydia Chin
- Department of Surgery, UCLA School of Medicine, University of California, Los Angeles, Los Angeles, California, USA; and
| | - Ramesh C Halder
- Department of Surgery, UCLA School of Medicine, University of California, Los Angeles, Los Angeles, California, USA; and
| | - Bien Sagong
- Department of Surgery, UCLA School of Medicine, University of California, Los Angeles, Los Angeles, California, USA; and
| | - Sam Famenini
- Department of Surgery, UCLA School of Medicine, University of California, Los Angeles, Los Angeles, California, USA; and
| | - James Sayre
- Department of Biostatistics, UCLA School of Public Health, University of California, Los Angeles, Los Angeles, California, USA
| | - Dennis Montoya
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Liudmilla Rubbi
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Milan Fiala
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA; Department of Surgery, UCLA School of Medicine, University of California, Los Angeles, Los Angeles, California, USA; and
| |
Collapse
|
118
|
Neuroprotective Activity of (-)-Epigallocatechin Gallate against Lipopolysaccharide-Mediated Cytotoxicity. J Immunol Res 2016; 2016:4962351. [PMID: 27191001 PMCID: PMC4844887 DOI: 10.1155/2016/4962351] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/15/2016] [Accepted: 03/17/2016] [Indexed: 12/14/2022] Open
Abstract
Lipopolysaccharide- (LPS-) mediated systemic inflammation plays a critical role in neurodegenerative diseases. The present study was conducted to evaluate the protective effects of epigallocatechin gallate (EGCG), the major component in green tea, on LPS-mediated inflammation and neurotoxicity. LPS treatment of macrophages induced expression of proinflammatory cytokines (TNF-α, IL-1β, and IL-6). However, EGCG pretreatment of macrophages significantly inhibited LPS-mediated induction of these cytokines. In addition, EGCG significantly diminished LPS-induced inflammatory cytokines in the peripheral mononuclear blood cells (PBMCs). Supernatant from EGCG-pretreated and LPS-activated macrophage cultures was found to be less cytotoxic to neurons than that from non-EGCG-pretreated and LPS-activated macrophage cultures. Furthermore, EGCG treatment of neurons could inhibit LPS-induced production of reactive oxygen species (ROS). Thus EGCG represents a potent and useful neuroprotective agent for inflammation-mediated neurological disorders.
Collapse
|
119
|
Simone C, Ramirez A, Bucchia M, Rinchetti P, Rideout H, Papadimitriou D, Re DB, Corti S. Is spinal muscular atrophy a disease of the motor neurons only: pathogenesis and therapeutic implications? Cell Mol Life Sci 2016; 73:1003-20. [PMID: 26681261 PMCID: PMC4756905 DOI: 10.1007/s00018-015-2106-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 01/16/2023]
Abstract
Spinal muscular atrophy (SMA) is a genetic neurological disease that causes infant mortality; no effective therapies are currently available. SMA is due to homozygous mutations and/or deletions in the survival motor neuron 1 gene and subsequent reduction of the SMN protein, leading to the death of motor neurons. However, there is increasing evidence that in addition to motor neurons, other cell types are contributing to SMA pathology. In this review, we will discuss the involvement of non-motor neuronal cells, located both inside and outside the central nervous system, in disease onset and progression. Even if SMN restoration in motor neurons is needed, it has been shown that optimal phenotypic amelioration in animal models of SMA requires a more widespread SMN correction. It has been demonstrated that non-motor neuronal cells are also involved in disease pathogenesis and could have important therapeutic implications. For these reasons it will be crucial to take this evidence into account for the clinical translation of the novel therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Simone
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Agnese Ramirez
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Monica Bucchia
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Paola Rinchetti
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Hardy Rideout
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens (BRFAA), Soranou Efesiou 4, 115 27, Athens, Greece
| | - Dimitra Papadimitriou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens (BRFAA), Soranou Efesiou 4, 115 27, Athens, Greece
| | - Diane B Re
- Department of Environmental Health Sciences, Columbia University, New York, NY, 10032, USA
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
| | - Stefania Corti
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
120
|
Kynurenine-3-monooxygenase: a review of structure, mechanism, and inhibitors. Drug Discov Today 2016; 21:315-24. [DOI: 10.1016/j.drudis.2015.11.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/14/2015] [Accepted: 11/05/2015] [Indexed: 01/04/2023]
|
121
|
Gargiulo S, Anzilotti S, Coda ARD, Gramanzini M, Greco A, Panico M, Vinciguerra A, Zannetti A, Vicidomini C, Dollé F, Pignataro G, Quarantelli M, Annunziato L, Brunetti A, Salvatore M, Pappatà S. Imaging of brain TSPO expression in a mouse model of amyotrophic lateral sclerosis with (18)F-DPA-714 and micro-PET/CT. Eur J Nucl Med Mol Imaging 2016; 43:1348-59. [PMID: 26816193 DOI: 10.1007/s00259-016-3311-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
PURPOSE To evaluate the feasibility and sensitivity of (18)F-DPA-714 for the study of microglial activation in the brain and spinal cord of transgenic SOD1(G93A) mice using high-resolution PET/CT and to evaluate the Iba1 and TSPO expression with immunohistochemistry. METHODS Nine symptomatic SOD1(G93A) mice (aged 117 ± 12.7 days, clinical score range 1 - 4) and five WT SOD1 control mice (aged 108 ± 28.5 days) underwent (18)F-DPA-714 PET/CT. SUV ratios were calculated by normalizing the cerebellar (rCRB), brainstem (rBS), motor cortex (rMCX) and cervical spinal cord (rCSC) activities to that of the frontal association cortex. Two WT SOD1 and six symptomatic SOD1(G93A) mice were studied by immunohistochemistry. RESULTS In the symptomatic SOD1(G93A) mice, rCRB, rBS and rCSC were increased as compared to the values in WT SOD1 mice, with a statistically significantly difference in rBS (2.340 ± 0.784 vs 1.576 ± 0.287, p = 0.014). Immunofluorescence studies showed that TSPO expression was increased in the trigeminal, facial, ambiguus and hypoglossal nuclei, as well as in the spinal cord, of symptomatic SOD1(G93A) mice and was colocalized with increased Iba1 staining. CONCLUSION Increased (18)F-DPA-714 uptake can be detected with high-resolution PET/CT in the brainstem of transgenic SOD1(G93A) mice, a region known to be a site of degeneration and increased microglial activation in amyotrophic lateral sclerosis, in agreement with increased TSPO expression in the brainstem nuclei shown by immunostaining. Therefore, (18)F-DPA-714 PET/CT might be a suitable tool to evaluate microglial activation in the SOD1(G93A) mouse model.
Collapse
Affiliation(s)
- S Gargiulo
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy.,Ceinge Biotecnologie Avanzate s. c. a r. l., Via G. Salvatore 486, 80145, Naples, Italy
| | - S Anzilotti
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - A R D Coda
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - M Gramanzini
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy.,Ceinge Biotecnologie Avanzate s. c. a r. l., Via G. Salvatore 486, 80145, Naples, Italy
| | - A Greco
- Ceinge Biotecnologie Avanzate s. c. a r. l., Via G. Salvatore 486, 80145, Naples, Italy.,Department of Advanced Biomedical Sciences, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - M Panico
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - A Vinciguerra
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - A Zannetti
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - C Vicidomini
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - F Dollé
- CEA, Institute for Biomedical Imaging, 4 Place du Général Leclerc, 91401, Orsay, France
| | - G Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - M Quarantelli
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - L Annunziato
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy.,Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - A Brunetti
- Ceinge Biotecnologie Avanzate s. c. a r. l., Via G. Salvatore 486, 80145, Naples, Italy.,Department of Advanced Biomedical Sciences, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - M Salvatore
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - S Pappatà
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy.
| |
Collapse
|
122
|
Cornejo F, von Bernhardi R. Age-Dependent Changes in the Activation and Regulation of Microglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:205-226. [DOI: 10.1007/978-3-319-40764-7_10] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
123
|
Cistaro A. Neuroimaging in Amyotrophic Lateral Sclerosis. PET-CT AND PET-MRI IN NEUROLOGY 2016:231-246. [DOI: 10.1007/978-3-319-31614-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
124
|
Salleh WMNHW, Ahmad F, Yen KH, Zulkifli RM. Anticholinesterase and Anti-inflammatory Constituents fromBeilschmiedia pulverulentaKosterm. ACTA ACUST UNITED AC 2016. [DOI: 10.20307/nps.2016.22.4.225] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Farediah Ahmad
- Department of Chemistry, Faculty of Science, University, Teknologi Malaysia (UTM), 81310 Skudai, Johor, Malaysia
| | - Khong Heng Yen
- School of Chemistry and Environment Studies, Faculty of Applied Sciences, University Teknologi MARA (UiTM) Sarawak Jalan Meranek, 94300 Kota Samarahan, Sarawak, Malaysia
| | - Razauden Mohamed Zulkifli
- Department of Bioscience and Health Sciences, Faculty of Biosciences and Medical Engineering, University Teknologi Malaysia (UTM), 81310 Skudai, Johor, Malaysia
| |
Collapse
|
125
|
Ray R, Juranek JK, Rai V. RAGE axis in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis. Neurosci Biobehav Rev 2015; 62:48-55. [PMID: 26724598 DOI: 10.1016/j.neubiorev.2015.12.006] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 10/19/2015] [Accepted: 12/10/2015] [Indexed: 12/13/2022]
Abstract
RAGE, the receptor of advanced glycation end-products, is thought to be one of the potential contributors to the neurodegeneration. It has been shown that RAGE activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines. RAGE involvement has been documented in the pathogenesis of a number of neurodegenerative diseases such amyotrophic lateral sclerosis (ALS), Alzheimer's, Parkinson's, Huntington's, Creutzfeld-Jakob' diseases and various neurodegenerative conditions such as diabetic neuropathy, familial amyloid polyneuropathy, Charcot neuroarthropathy and vasculitic neuropathy. Although the detailed mechanisms of RAGE contribution to the neurodegeneration remains unclear, studies indicate that RAGE detrimental actions are exerted via its binding to the pro-inflammatory ligands such as advanced glycation end-products, S100/calgranulin and amphoterin and subsequent activation of downstream regulatory pathways such as NF-κB, STAT and JKN pathways. Here, in this review we attempt to shed light onto molecular events and pathological pathways involved in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis (ALS)--a progressive and fatal neurodegenerative disorder, summarizing current knowledge and the prospect of RAGE in the pathogenesis of this disastrous disease.
Collapse
Affiliation(s)
- Rashmi Ray
- Institute of Life Sciences, Bhubaneswar 751023, India; Manipal University, Karnataka 576104, India
| | - Judyta K Juranek
- Department of Medicine, New York University Medical Center, New York, USA; Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-557 Olsztyn, Poland.
| | - Vivek Rai
- Institute of Life Sciences, Bhubaneswar 751023, India.
| |
Collapse
|
126
|
Central nervous system myeloid cells as drug targets: current status and translational challenges. Nat Rev Drug Discov 2015; 15:110-24. [DOI: 10.1038/nrd.2015.14] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
127
|
Jeyachandran A, Mertens B, McKissick EA, Mitchell CS. Type I Vs. Type II Cytokine Levels as a Function of SOD1 G93A Mouse Amyotrophic Lateral Sclerosis Disease Progression. Front Cell Neurosci 2015; 9:462. [PMID: 26648846 PMCID: PMC4664727 DOI: 10.3389/fncel.2015.00462] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/13/2015] [Indexed: 01/01/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal motoneuron disease that is characterized by the degradation of neurons throughout the central nervous system. Inflammation have been cited a key contributor to ALS neurodegeneration, but the timeline of cytokine upregulation remains unresolved. The goal of this study was to temporally examine the correlation between the varying levels of pro-inflammatory type I cytokines (IL-1β, IL-1α, IL-12, TNF-α, and GFAP) and anti-inflammatory type II cytokines (IL-4, IL-6, IL-10) throughout the progression of ALS in the SOD1 G93A mouse model. Cytokine level data from high copy SOD1 G93A transgenic mice was collected from 66 peer-reviewed studies. For each corresponding experimental time point, the ratio of transgenic to wild type (TG/WT) cytokine was calculated. One-way ANOVA and t-tests with Bonferonni correction were used to analyze the data. Meta-analysis was performed for four discrete stages: early, pre-onset, post-onset, and end stage. A significant increase in TG cytokine levels was found when compared to WT cytokine levels across the entire SOD1 G93A lifespan for majority of the cytokines. The rates of change of the individual cytokines, and type I and type II were not significantly different; however, the mean fold change of type I was expressed at significantly higher levels than type II levels across all stages with the difference between the means becoming more pronounced at the end stage. An overexpression of cytokines occurred both before and after the onset of ALS symptoms. The trend between pro-inflammatory type I and type II cytokine mean levels indicate a progressive instability of the dynamic balance between pro- and anti-inflammatory cytokines as anti-inflammatory cytokines fail to mediate the pronounced increase in pro-inflammatory cytokines. Very early immunoregulatory treatment is necessary to successfully interrupt ALS-induced neuroinflammation.
Collapse
Affiliation(s)
- Amilia Jeyachandran
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Benjamin Mertens
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Eric A McKissick
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University Atlanta, GA, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University Atlanta, GA, USA
| |
Collapse
|
128
|
Waisman A, Liblau RS, Becher B. Innate and adaptive immune responses in the CNS. Lancet Neurol 2015; 14:945-55. [PMID: 26293566 DOI: 10.1016/s1474-4422(15)00141-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 04/22/2015] [Accepted: 06/11/2015] [Indexed: 12/11/2022]
Abstract
Almost every disorder of the CNS is said to have an inflammatory component, but the precise nature of inflammation in the CNS is often imprecisely defined, and the role of CNS-resident cells is uncertain compared with that of cells that invade the tissue from the systemic immune compartment. To understand inflammation in the CNS, the term must be better defined, and the response of tissue to disturbances in homoeostasis (eg, neurodegenerative processes) should be distinguished from disorders in which aberrant immune responses lead to CNS dysfunction and tissue destruction (eg, autoimmunity). Whether the inflammatory tissue response to injury is reparative or degenerative seems to be dependent on context and timing, as are the windows of opportunity for therapeutic intervention in inflammatory CNS diseases.
Collapse
Affiliation(s)
- Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Roland S Liblau
- Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse, France
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
129
|
Kumari R, Astafurov K, Genis A, Danias J. Differential Effects of C1qa Ablation on Glaucomatous Damage in Two Sexes in DBA/2NNia Mice. PLoS One 2015; 10:e0142199. [PMID: 26544197 PMCID: PMC4636422 DOI: 10.1371/journal.pone.0142199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 10/18/2015] [Indexed: 11/19/2022] Open
Abstract
PURPOSE To determine the sex and age-related effects of C1qa ablation on retinal ganglion cell (RGC) and optic nerve (ON) axonal loss in a mouse model of glaucomatous neurodegeneration. METHODS Congenic C1qa mice were generated in the DBA/2NNia background. Female and male knockout (-/-), heterozygous (+/-), and wild type (+/+) mice were aged up to 14 months and IOPs were recorded in a subset of animals. Retinas of mice from all three groups at 5-6, 9-10 and 11-13 months of age were flat-mounted after retrograde labeling with Fluorogold. Imaged retinas were scored (RGC score) semi-quantitatively on a 10 point scale by two independent observers. A subset of retinas and optic nerves were also used for measurement of total number of RGCs. Semi-thin sections of ON were imaged and graded (ON score) for the amount of axonal damage semi-quantitatively, by two masked observers. Analysis of covariance (ANCOVA) was used for statistical comparisons. Microglial cells in flat-mounted retinas of 5-6 month old C1qa -/- and C1qa +/+ mice were used for assessment of microglial activation utilizing morphological criteria. RESULTS Female C1qa -/- mice had significantly higher IOP (p<0.000001, ANOVA) between 8 and 13 months of age compared to C1qa +/+ animals. No differences in IOPs between animals of the three genotypes were observed in males. At 5-6 months of age, there was no difference in RGC or ON scores between the three genotypes in animals of either sex. At 9-10 months of age, female mice didn't show significant differences in RGC or ON scores between the three genotypes. However, male C1qa -/- and C1qa +/- mice of the same age had better RGC and ON scores (p<0.003 and p<0.05, ANCOVA, for RGC and ON scores, respectively) compared with C1qa +/+ mice. At 11-13 months of age, female C1qa -/- mice had better RGC scores (p<0.006, ANCOVA) compared to C1qa +/+ and C1qa +/- animals. Accordingly, C1qa -/- mice had higher RGC counts (p<0.03, t-test) compared to C1qa +/+ animals. In male mice, there was a tendency for 12 month old C1qa -/- animals to have better RGC scores and higher RGC counts, but this didn't reach statistical significance. ON scores in 11-13 month old animals of either sex were not different between all three genotype. Microglial activation in male 5-6 month old C1qa -/- mice was decreased compared to C1qa +/+ animals; no such effect was seen in females. CONCLUSIONS Absence of C1qa ameliorates RGC and ON loss in the DBA/2NNia strain, but this effect differs between the two sexes. C1q-mediated RGC damage seems to be more potent than IOP-mediated RGC loss. In contrast, C1qa absence provides axonal protection early on, but this protection cannot overcome the effects of significant IOP elevation.
Collapse
Affiliation(s)
- Ruma Kumari
- Department of Cell Biology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- State University of New York (SUNY) Eye Institute, Brooklyn, New York, United States of America
- * E-mail: (RK); (JD)
| | - Konstantin Astafurov
- Department of Cell Biology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- State University of New York (SUNY) Eye Institute, Brooklyn, New York, United States of America
| | - Alina Genis
- Department of Ophthalmology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- State University of New York (SUNY) Eye Institute, Brooklyn, New York, United States of America
| | - John Danias
- Department of Cell Biology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Ophthalmology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- State University of New York (SUNY) Eye Institute, Brooklyn, New York, United States of America
- * E-mail: (RK); (JD)
| |
Collapse
|
130
|
Maier A, Deigendesch N, Müller K, Weishaupt JH, Krannich A, Röhle R, Meissner F, Molawi K, Münch C, Holm T, Meyer R, Meyer T, Zychlinsky A. Interleukin-1 Antagonist Anakinra in Amyotrophic Lateral Sclerosis--A Pilot Study. PLoS One 2015; 10:e0139684. [PMID: 26444282 PMCID: PMC4596620 DOI: 10.1371/journal.pone.0139684] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 09/15/2015] [Indexed: 12/11/2022] Open
Abstract
Preclinical studies show that blocking Interleukin–1 (IL–1) retards the progression of Amyotrophic Lateral Sclerosis (ALS). We assessed the safety of Anakinra (ANA), an IL–1 receptor antagonist, in ALS patients. In a single arm pilot study we treated 17 ALS patients with ANA (100 mg) daily for one year. We selected patients with dominant or exclusive lower motor neuron degeneration (LMND) presentation, as peripheral nerves may be more accessible to the drug. Our primary endpoint was safety and tolerability. Secondary endpoints included measuring disease progression with the revised ALS functional rating scale (ALSFRSr). We also quantified serum inflammatory markers. For comparison, we generated a historical cohort of 47 patients that fit the criteria for enrolment, disease characteristics and rate of progression of the study group. Only mild adverse events occurred in ALS patients treated with ANA. Notably, we observed lower levels of cytokines and the inflammatory marker fibrinogen during the first 24 weeks of treatment. Despite of this, we could not detect a significant reduction in disease progression during the same period in patients treated with ANA compared to controls as measured by the ALSFRSr. In the second part of the treatment period we observed an increase in serum inflammatory markers. Sixteen out of the 17 patients (94%) developed antibodies against ANA. This study showed that blocking IL–1 is safe in patients with ALS. Further trials should test whether targeting IL–1 more efficiently can help treating this devastating disease.
Collapse
Affiliation(s)
- André Maier
- Department of Neurology, Charité-University Hospital, Campus Virchow-Klinikum, Berlin, Germany
| | | | | | | | - Alexander Krannich
- Department of Biostatistics, Coordination Center for Clinical Trials, Charité-University Hospital, Berlin, Germany
| | - Robert Röhle
- Department of Biostatistics, Coordination Center for Clinical Trials, Charité-University Hospital, Berlin, Germany
| | - Felix Meissner
- Max-Planck Institute for Infection Biology, Berlin, Germany
| | - Kaaweh Molawi
- Max-Planck Institute for Infection Biology, Berlin, Germany
| | - Christoph Münch
- Department of Neurology, Charité-University Hospital, Campus Virchow-Klinikum, Berlin, Germany
| | - Teresa Holm
- Department of Neurology, Charité-University Hospital, Campus Virchow-Klinikum, Berlin, Germany
| | - Robert Meyer
- Department of Neurology, Charité-University Hospital, Campus Virchow-Klinikum, Berlin, Germany
| | - Thomas Meyer
- Department of Neurology, Charité-University Hospital, Campus Virchow-Klinikum, Berlin, Germany
| | - Arturo Zychlinsky
- Max-Planck Institute for Infection Biology, Berlin, Germany
- * E-mail:
| |
Collapse
|
131
|
Boison D, Aronica E. Comorbidities in Neurology: Is adenosine the common link? Neuropharmacology 2015; 97:18-34. [PMID: 25979489 PMCID: PMC4537378 DOI: 10.1016/j.neuropharm.2015.04.031] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
Abstract
Comorbidities in Neurology represent a major conceptual and therapeutic challenge. For example, temporal lobe epilepsy (TLE) is a syndrome comprised of epileptic seizures and comorbid symptoms including memory and psychiatric impairment, depression, and sleep dysfunction. Similarly, Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic Lateral Sclerosis (ALS) are accompanied by various degrees of memory dysfunction. Patients with AD have an increased likelihood for seizures, whereas all four conditions share certain aspects of psychosis, depression, and sleep dysfunction. This remarkable overlap suggests common pathophysiological mechanisms, which include synaptic dysfunction and synaptotoxicity, as well as glial activation and astrogliosis. Astrogliosis is linked to synapse function via the tripartite synapse, but astrocytes also control the availability of gliotransmitters and adenosine. Here we will specifically focus on the 'adenosine hypothesis of comorbidities' implying that astrocyte activation, via overexpression of adenosine kinase (ADK), induces a deficiency in the homeostatic tone of adenosine. We present evidence from patient-derived samples showing astrogliosis and overexpression of ADK as common pathological hallmark of epilepsy, AD, PD, and ALS. We discuss a transgenic 'comorbidity model', in which brain-wide overexpression of ADK and resulting adenosine deficiency produces a comorbid spectrum of seizures, altered dopaminergic function, attentional impairment, and deficits in cognitive domains and sleep regulation. We conclude that dysfunction of adenosine signaling is common in neurological conditions, that adenosine dysfunction can explain co-morbid phenotypes, and that therapeutic adenosine augmentation might be effective for the treatment of comorbid symptoms in multiple neurological conditions.
Collapse
Affiliation(s)
- Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR 97232, USA.
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands; Stichting Epilepsie Instellingen (SEIN) Nederland, Heemstede, The Netherlands
| |
Collapse
|
132
|
Abdelhak A, Junker A, Brettschneider J, Kassubek J, Ludolph AC, Otto M, Tumani H. Brain-Specific Cytoskeletal Damage Markers in Cerebrospinal Fluid: Is There a Common Pattern between Amyotrophic Lateral Sclerosis and Primary Progressive Multiple Sclerosis? Int J Mol Sci 2015; 16:17565-88. [PMID: 26263977 PMCID: PMC4581209 DOI: 10.3390/ijms160817565] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/20/2015] [Accepted: 07/23/2015] [Indexed: 11/25/2022] Open
Abstract
Many neurodegenerative disorders share a common pathophysiological pathway involving axonal degeneration despite different etiological triggers. Analysis of cytoskeletal markers such as neurofilaments, protein tau and tubulin in cerebrospinal fluid (CSF) may be a useful approach to detect the process of axonal damage and its severity during disease course. In this article, we review the published literature regarding brain-specific CSF markers for cytoskeletal damage in primary progressive multiple sclerosis and amyotrophic lateral sclerosis in order to evaluate their utility as a biomarker for disease progression in conjunction with imaging and histological markers which might also be useful in other neurodegenerative diseases associated with affection of the upper motor neurons. A long-term benefit of such an approach could be facilitating early diagnostic and prognostic tools and assessment of treatment efficacy of disease modifying drugs.
Collapse
Affiliation(s)
- Ahmed Abdelhak
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany.
| | - Andreas Junker
- Institute of Neuropathology, University Hospital Göttingen, Robert-Koch-Str 40, 37075 Göttingen, Germany.
| | | | - Jan Kassubek
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany.
| | - Albert C Ludolph
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany.
| | - Markus Otto
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany.
| | - Hayrettin Tumani
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany.
| |
Collapse
|
133
|
Lee SH, Choi SM, Yang EJ. Bee Venom Acupuncture Augments Anti-Inflammation in the Peripheral Organs of hSOD1G93A Transgenic Mice. Toxins (Basel) 2015; 7:2835-44. [PMID: 26230709 PMCID: PMC4549727 DOI: 10.3390/toxins7082835] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/06/2015] [Indexed: 11/16/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) includes progressively degenerated motor neurons in the brainstem, motor cortex, and spinal cord. Recent reports demonstrate the dysfunction of multiple organs, including the lungs, spleen, and liver, in ALS animals and patients. Bee venom acupuncture (BVA) has been used for treating inflammatory diseases in Oriental Medicine. In a previous study, we demonstrated that BV prevented motor neuron death and increased anti-inflammation in the spinal cord of symptomatic hSOD1G93A transgenic mice. In this study, we examined whether BVA's effects depend on acupuncture point (ST36) in the organs, including the liver, spleen and kidney, of hSOD1G93A transgenic mice. We found that BV treatment at ST36 reduces inflammation in the liver, spleen, and kidney compared with saline-treatment at ST36 and BV injected intraperitoneally in symptomatic hSOD1G93A transgenic mice. Those findings suggest that BV treatment combined with acupuncture stimulation is more effective at reducing inflammation and increasing immune responses compared with only BV treatment, at least in an ALS animal model.
Collapse
Affiliation(s)
- Sun-Hwa Lee
- Division of Clinical Research, Korea Institute of Oriental Medicine, Daejeon 305-811, Korea.
| | - Sun-Mi Choi
- Executive Director of R&D, Korea Institute of Oriental Medicine, Daejeon 305-811, Korea.
| | - Eun Jin Yang
- Division of Clinical Research, Korea Institute of Oriental Medicine, Daejeon 305-811, Korea.
| |
Collapse
|
134
|
Abstract
Neurodegenerative diseases (NDs) collectively afflict more than 40 million people worldwide. The majority of these diseases lack therapies to slow or stop progression due in large part to the challenge of disentangling the simultaneous presentation of broad, multifaceted pathophysiologic changes. Present technologies and computational capabilities suggest an optimistic future for deconvolving these changes to identify novel mechanisms driving ND onset and progression. In particular, integration of highly multi-dimensional omic analytical techniques (e.g., microarray, mass spectrometry) with computational systems biology approaches provides a systematic methodology to elucidate new mechanisms driving NDs. In this review, we begin by summarizing the complex pathophysiology of NDs associated with protein aggregation, emphasizing the shared complex dysregulation found in all of these diseases, and discuss available experimental ND models. Next, we provide an overview of technological and computational techniques used in systems biology that are applicable to studying NDs. We conclude by reviewing prior studies that have applied these approaches to NDs and comment on the necessity of combining analysis from both human tissues and model systems to identify driving mechanisms. We envision that the integration of computational approaches with multiple omic analyses of human tissues, and mouse and in vitro models, will enable the discovery of new therapeutic strategies for these devastating diseases.
Collapse
Affiliation(s)
- Levi B Wood
- Cancer Research Institute, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
135
|
Ehrhart J, Smith AJ, Kuzmin-Nichols N, Zesiewicz TA, Jahan I, Shytle RD, Kim SH, Sanberg CD, Vu TH, Gooch CL, Sanberg PR, Garbuzova-Davis S. Humoral factors in ALS patients during disease progression. J Neuroinflammation 2015; 12:127. [PMID: 26126965 PMCID: PMC4487852 DOI: 10.1186/s12974-015-0350-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/19/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting upper and lower motor neurons in the CNS and leading to paralysis and death. There are currently no effective treatments for ALS due to the complexity and heterogeneity of factors involved in motor neuron degeneration. A complex of interrelated effectors have been identified in ALS, yet systemic factors indicating and/or reflecting pathological disease developments are uncertain. The purpose of the study was to identify humoral effectors as potential biomarkers during disease progression. METHODS Thirteen clinically definite ALS patients and seven non-neurological controls enrolled in the study. Peripheral blood samples were obtained from each ALS patient and control at two visits separated by 6 months. The Revised ALS Functional Rating Scale (ALSFRS-R) was used to evaluate overall ALS-patient functional status at each visit. Eleven humoral factors were analyzed in sera. Cytokine levels (GM-CSF, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, and TNF-α) were determined using the Bio-Rad Bio-Plex® Luminex 200 multiplex assay system. Nitrite, a breakdown product of NO, was quantified using a Griess Reagent System. Glutathione (GSH) concentrations were measured using a Glutathione Fluorometric Assay Kit. RESULTS ALS patients had ALSFRS-R scores of 30.5 ± 1.9 on their first visit and 27.3 ± 2.7 on the second visit, indicating slight disease progression. Serum multiplex cytokine panels revealed statistically significant changes in IL-2, IL-5, IL-6, and IL-8 levels in ALS patients depending on disease status at each visit. Nitrite serum levels trended upwards in ALS patients while serum GSH concentrations were drastically decreased in sera from ALS patients versus controls at both visits. CONCLUSIONS Our results demonstrated a systemic pro-inflammatory state and impaired antioxidant system in ALS patients during disease progression. Increased levels of pro-inflammatory IL-6, IL-8, and nitrite and significantly decreased endogenous antioxidant GSH levels could identify these humoral constituents as systemic biomarkers for ALS. However, systemic changes in IL-2, IL-5, and IL-6 levels determined between visits in ALS patients might indicate adaptive immune system responses dependent on current disease stage. These novel findings, showing dynamic changes in humoral effectors during disease progression, could be important for development of an effective treatment for ALS.
Collapse
Affiliation(s)
| | - Adam J Smith
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | | | - Theresa A Zesiewicz
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Israt Jahan
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - R Douglas Shytle
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612, USA.
| | - Seol-Hee Kim
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | | | - Tuan H Vu
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Clifton L Gooch
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Paul R Sanberg
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612, USA. .,Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Psychiatry, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612, USA. .,Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
136
|
Gunia-Krzyżak A, Pańczyk K, Waszkielewicz AM, Marona H. Cinnamamide Derivatives for Central and Peripheral Nervous System Disorders--A Review of Structure-Activity Relationships. ChemMedChem 2015; 10:1302-25. [PMID: 26083325 DOI: 10.1002/cmdc.201500153] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Indexed: 12/17/2022]
Abstract
The cinnamamide scaffold has been incorporated in to the structure of numerous organic compounds with therapeutic potential. The scaffold enables multiple interactions, such as hydrophobic, dipolar, and hydrogen bonding, with important molecular targets. Additionally, the scaffold has multiple substitution options providing the opportunity to optimize and modify the pharmacological activity of the derivatives. In particular, cinnamamide derivatives have exhibited therapeutic potential in animal models of both central and peripheral nervous system disorders. Some have undergone clinical trials and were introduced on to the pharmaceutical market. The diverse activities observed in the nervous system included anticonvulsant, antidepressant, neuroprotective, analgesic, anti-inflammatory, muscle relaxant, and sedative properties. Over the last decade, research has focused on the molecular mechanisms of action of these derivatives, and the data reported in the literature include targeting the γ-aminobutyric acid type A (GABAA ) receptors, N-methyl-D-aspartate (NMDA) receptors, transient receptor potential (TRP) cation channels, voltage-gated potassium channels, histone deacetylases (HDACs), prostanoid receptors, opioid receptors, and histamine H3 receptors. Here, the literature data from reports evaluating cinnamic acid amide derivatives for activity in target-based or phenotypic assays, both in vivo and in vitro, relevant to disorders of the central and peripheral nervous systems are analyzed and structure-activity relationships discussed.
Collapse
Affiliation(s)
- Agnieszka Gunia-Krzyżak
- Department of Bioorganic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow (Poland).
| | - Katarzyna Pańczyk
- Department of Bioorganic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow (Poland)
| | - Anna M Waszkielewicz
- Department of Bioorganic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow (Poland)
| | - Henryk Marona
- Department of Bioorganic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow (Poland)
| |
Collapse
|
137
|
Deleidi M, Jäggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Front Neurosci 2015; 9:172. [PMID: 26089771 PMCID: PMC4453474 DOI: 10.3389/fnins.2015.00172] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
As we age, the immune system undergoes a process of senescence accompanied by the increased production of proinflammatory cytokines, a chronic subclinical condition named as “inflammaging”. Emerging evidence from human and experimental models suggest that immune senescence also affects the central nervous system and promotes neuronal dysfunction, especially within susceptible neuronal populations. In this review we discuss the potential role of immune aging, inflammation and metabolic derangement in neurological diseases. The discovery of novel therapeutic strategies targeting age-linked inflammation may promote healthy brain aging and the treatment of neurodegenerative as well as neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michela Deleidi
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Madeline Jäggle
- Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Hertie Institute for Clinical Brain Research, University of Tübingen Tübingen, Germany
| | - Graziella Rubino
- Department of Internal Medicine II, Center for Medical Research, University of Tübingen Tübingen, Germany
| |
Collapse
|
138
|
Absence of toll-like receptor 4 (TLR4) extends survival in the hSOD1 G93A mouse model of amyotrophic lateral sclerosis. J Neuroinflammation 2015; 12:90. [PMID: 25962427 PMCID: PMC4431460 DOI: 10.1186/s12974-015-0310-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/28/2015] [Indexed: 12/13/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a devastating late onset neurodegenerative disorder that is characterised by the progressive loss of upper and lower motor neurons. The mechanisms underlying ALS pathogenesis are unclear; however, there is emerging evidence the innate immune system, including components of the toll-like receptor (TLR) system, may drive disease progression. For example, toll-like receptor 4 (TLR4) antagonism in a spontaneous ‘wobbler mouse’ model of ALS increased motor function, associated with a decrease in microglial activation. This study therefore aimed to extend from these findings and determine the expression and function of TLR4 signalling in hSOD1G93A mice, the most widely established preclinical model of ALS. Findings TLR4 and one of its major endogenous ligands, high-mobility group box 1 (HMGB1), were increased during disease progression in hSOD1G93A mice, with TLR4 and HMGB1 expressed by activated microglia and astrocytes. hSOD1G93A mice lacking TLR4 showed transient improvements in hind-limb grip strength and significantly extended survival when compared to TLR4-sufficient hSOD1G93A mice. Conclusion These results suggest that enhanced glial TLR4 signalling during disease progression contributes to end-stage ALS pathology in hSOD1G93A mice.
Collapse
|
139
|
Appolinário PP, Medinas DB, Chaves-Filho AB, Genaro-Mattos TC, Cussiol JRR, Netto LES, Augusto O, Miyamoto S. Oligomerization of Cu,Zn-Superoxide Dismutase (SOD1) by Docosahexaenoic Acid and Its Hydroperoxides In Vitro: Aggregation Dependence on Fatty Acid Unsaturation and Thiols. PLoS One 2015; 10:e0125146. [PMID: 25928076 PMCID: PMC4415921 DOI: 10.1371/journal.pone.0125146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/16/2015] [Indexed: 12/21/2022] Open
Abstract
Docosahexaenoic acid (C22:6, n-3, DHA) is a polyunsaturated fatty acid highly enriched in the brain. This fatty acid can be easily oxidized yielding hydroperoxides as primary products. Cu, Zn-Superoxide dismutase (SOD1) aggregation is a common hallmark of Amyotrophic Lateral Sclerosis (ALS) and the molecular mechanisms behind their formation are not completely understood. Here we investigated the effect of DHA and its hydroperoxides (DHAOOH) on human SOD1 oligomerization in vitro. DHA induced the formation of high-molecular-weight (HMW) SOD1 species (>700 kDa). Aggregation was dependent on free thiols and occurred primarily with the protein in its apo-form. SOD1 incubation with DHA was accompanied by changes in protein structure leading to exposure of protein hydrophobic patches and formation of non-amyloid aggregates. Site-directed mutagenesis studies demonstrated that Cys 6 and Cys 111 in wild-type and Cys 6 in ALS-linked G93A mutant are required for aggregation. In contrast, DHAOOH did not induce HMW species formation but promoted abnormal covalent dimerization of apo-SOD1 that was resistant to SDS and thiol reductants. Overall, our data demonstrate that DHA and DHAOOH induce distinct types of apo-SOD1 oligomerization leading to the formation of HMW and low-molecular-weight species, respectively.
Collapse
Affiliation(s)
| | - Danilo Bilches Medinas
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Adriano B. Chaves-Filho
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Thiago C. Genaro-Mattos
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - José Renato Rosa Cussiol
- Departamento de Biologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ohara Augusto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
140
|
Hooten KG, Beers DR, Zhao W, Appel SH. Protective and Toxic Neuroinflammation in Amyotrophic Lateral Sclerosis. Neurotherapeutics 2015; 12:364-75. [PMID: 25567201 PMCID: PMC4404435 DOI: 10.1007/s13311-014-0329-3] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a clinically heterogeneous disorder characterized by loss of motor neurons, resulting in paralysis and death. Multiple mechanisms of motor neuron injury have been implicated based upon the more than 20 different genetic causes of familial ALS. These inherited mutations compromise diverse motor neuron pathways leading to cell-autonomous injury. In the ALS transgenic mouse models, however, motor neurons do not die alone. Cell death is noncell-autonomous dependent upon a well orchestrated dialogue between motor neurons and surrounding glia and adaptive immune cells. The pathogenesis of ALS consists of 2 stages: an early neuroprotective stage and a later neurotoxic stage. During early phases of disease progression, the immune system is protective with glia and T cells, especially M2 macrophages/microglia, and T helper 2 cells and regulatory T cells, providing anti-inflammatory factors that sustain motor neuron viability. As the disease progresses and motor neuron injury accelerates, a second rapidly progressing phase develops, characterized by M1 macrophages/microglia, and proinflammatory T cells. In rapidly progressing ALS patients, as in transgenic mice, neuroprotective regulatory T cells are significantly decreased and neurotoxicity predominates. Our own therapeutic efforts are focused on modulating these neuroinflammatory pathways. This review will focus on the cellular players involved in neuroinflammation in ALS and current therapeutic strategies to enhance neuroprotection and suppress neurotoxicity with the goal of arresting the progressive and devastating nature of ALS.
Collapse
Affiliation(s)
- Kristopher G. Hooten
- />Department of Neurology, Houston Methodist Neurological Institute, Peggy and Gary Edwards ALS Research Laboratory, Houston Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX 77030 USA
- />Department of Neurological Surgery, University of Florida, Box 100265, Gainesville, FL 32610-0261 USA
| | - David R. Beers
- />Department of Neurology, Houston Methodist Neurological Institute, Peggy and Gary Edwards ALS Research Laboratory, Houston Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX 77030 USA
| | - Weihua Zhao
- />Department of Neurology, Houston Methodist Neurological Institute, Peggy and Gary Edwards ALS Research Laboratory, Houston Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX 77030 USA
| | - Stanley H. Appel
- />Department of Neurology, Houston Methodist Neurological Institute, Peggy and Gary Edwards ALS Research Laboratory, Houston Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX 77030 USA
| |
Collapse
|
141
|
García-Escudero V, Rosales M, Muñoz JL, Scola E, Medina J, Khalique H, Garaulet G, Rodriguez A, Lim F. Patient-derived olfactory mucosa for study of the non-neuronal contribution to amyotrophic lateral sclerosis pathology. J Cell Mol Med 2015; 19:1284-95. [PMID: 25807871 PMCID: PMC4459844 DOI: 10.1111/jcmm.12488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/10/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerative motor neuron disease which currently has no cure. Research using rodent ALS models transgenic for mutant superoxide dismutase 1 (SOD1) has implicated that glial–neuronal interactions play a major role in the destruction of motor neurons, but the generality of this mechanism is not clear as SOD1 mutations only account for less than 2% of all ALS cases. Recently, this hypothesis was backed up by observation of similar effects using astrocytes derived from post-mortem spinal cord tissue of ALS patients which did not carry SOD1 mutations. However, such necropsy samples may not be easy to obtain and may not always yield viable cell cultures. Here, we have analysed olfactory mucosa (OM) cells, which can be easily isolated from living ALS patients. Disease-specific changes observed when ALS OM cells were co-cultured with human spinal cord neurons included decreased neuronal viability, aberrant neuronal morphology and altered glial inflammatory responses. Our results show the potential of OM cells as new cell models for ALS.
Collapse
Affiliation(s)
- Vega García-Escudero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (C.S.I.C.- U.A.M.), Universidad Autónoma de Madrid, Madrid, Spain
| | - María Rosales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - José Luis Muñoz
- Departamento de Neurología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Esteban Scola
- Departamento de Otorrinolaringología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Javier Medina
- Departamento de Otorrinolaringología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Hena Khalique
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Guillermo Garaulet
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Rodriguez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Filip Lim
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
142
|
Horiuchi H, Parajuli B, Wang Y, Azuma YT, Mizuno T, Takeuchi H, Suzumura A. Interleukin-19 acts as a negative autocrine regulator of activated microglia. PLoS One 2015; 10:e0118640. [PMID: 25794104 PMCID: PMC4368203 DOI: 10.1371/journal.pone.0118640] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/12/2015] [Indexed: 11/19/2022] Open
Abstract
Activated microglia can exert either neurotoxic or neuroprotective effects, and they play pivotal roles in the pathogenesis and progression of various neurological diseases. In this study, we used cDNA microarrays to show that interleukin-19 (IL-19), an IL-10 family cytokine, is markedly upregulated in activated microglia. Furthermore, we found that microglia are the only cells in the nervous system that express the IL-19 receptor, a heterodimer of the IL-20Rα and IL-20Rβ subunits. IL-19 deficiency increased the production of such pro-inflammatory cytokines as IL-6 and tumor necrosis factor-α in activated microglia, and IL-19 treatment suppressed this effect. Moreover, in a mouse model of Alzheimer's disease, we observed upregulation of IL-19 in affected areas in association with disease progression. Our findings demonstrate that IL-19 is an anti-inflammatory cytokine, produced by activated microglia, that acts negatively on microglia in an autocrine manner. Thus, microglia may self-limit their inflammatory response by producing the negative regulator IL-19.
Collapse
Affiliation(s)
- Hiroshi Horiuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Bijay Parajuli
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Yue Wang
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, Izumisano, Osaka, 598–8531, Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
- * E-mail:
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| |
Collapse
|
143
|
Zigdon H, Savidor A, Levin Y, Meshcheriakova A, Schiffmann R, Futerman AH. Identification of a biomarker in cerebrospinal fluid for neuronopathic forms of Gaucher disease. PLoS One 2015; 10:e0120194. [PMID: 25775479 PMCID: PMC4361053 DOI: 10.1371/journal.pone.0120194] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 02/06/2015] [Indexed: 11/18/2022] Open
Abstract
Gaucher disease, a recessive inherited metabolic disorder caused by defects in the gene encoding glucosylceramidase (GlcCerase), can be divided into three subtypes according to the appearance of symptoms associated with central nervous system involvement. We now identify a protein, glycoprotein non-metastatic B (GPNMB), that acts as an authentic marker of brain pathology in neurological forms of Gaucher disease. Using three independent techniques, including quantitative global proteomic analysis of cerebrospinal fluid (CSF) in samples from Gaucher disease patients that display neurological symptoms, we demonstrate a correlation between the severity of symptoms and GPNMB levels. Moreover, GPNMB levels in the CSF correlate with disease severity in a mouse model of Gaucher disease. GPNMB was also elevated in brain samples from patients with type 2 and 3 Gaucher disease. Our data suggest that GPNMB can be used as a marker to quantify neuropathology in Gaucher disease patients and as a marker of treatment efficacy once suitable treatments towards the neurological symptoms of Gaucher disease become available.
Collapse
Affiliation(s)
- Hila Zigdon
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Anna Meshcheriakova
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Research Institute, Dallas, TX, United States of America
| | - Anthony H. Futerman
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
144
|
Cai M, Choi SM, Yang EJ. The effects of bee venom acupuncture on the central nervous system and muscle in an animal hSOD1G93A mutant. Toxins (Basel) 2015; 7:846-58. [PMID: 25781653 PMCID: PMC4379529 DOI: 10.3390/toxins7030846] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/17/2015] [Accepted: 03/03/2015] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is caused by the degeneration of lower and upper motor neurons, leading to muscle paralysis and respiratory failure. However, there is no effective drug or therapy to treat ALS. Complementary and alternative medicine (CAM), including acupuncture, pharmacopuncture, herbal medicine, and massage is popular due to the significant limitations of conventional therapy. Bee venom acupuncture (BVA), also known as one of pharmacopunctures, has been used in Oriental medicine to treat inflammatory diseases. The purpose of this study is to investigate the effect of BVA on the central nervous system (CNS) and muscle in symptomatic hSOD1G93A transgenic mice, an animal model of ALS. Our findings show that BVA at ST36 enhanced motor function and decreased motor neuron death in the spinal cord compared to that observed in hSOD1G93A transgenic mice injected intraperitoneally (i.p.) with BV. Furthermore, BV treatment at ST36 eliminated signaling downstream of inflammatory proteins such as TLR4 in the spinal cords of symptomatic hSOD1G93A transgenic mice. However, i.p. treatment with BV reduced the levels of TNF-α and Bcl-2 expression in the muscle hSOD1G93A transgenic mice. Taken together, our findings suggest that BV pharmacopuncture into certain acupoints may act as a chemical stimulant to activate those acupoints and subsequently engage the endogenous immune modulatory system in the CNS in an animal model of ALS.
Collapse
Affiliation(s)
- MuDan Cai
- Department of KM Fundamental Research, Korea Institute of Oriental Medicine, 483 Expo-ro, Daejeon, Yuseong-gu 305-811, Korea.
| | - Sun-Mi Choi
- Executive Director of R&D, Korea Institute of Oriental Medicine, 483 Expo-ro, Daejeon, Yuseong-gu 305-811, Korea.
| | - Eun Jin Yang
- Department of KM Fundamental Research, Korea Institute of Oriental Medicine, 483 Expo-ro, Daejeon, Yuseong-gu 305-811, Korea.
| |
Collapse
|
145
|
Lee M, Shi X, Barron AE, McGeer E, McGeer PL. Human antimicrobial peptide LL-37 induces glial-mediated neuroinflammation. Biochem Pharmacol 2015; 94:130-41. [PMID: 25686659 DOI: 10.1016/j.bcp.2015.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 01/30/2015] [Accepted: 02/06/2015] [Indexed: 12/29/2022]
Abstract
LL-37 is the sole cathelicidin-derived antimicrobial peptide found in humans. It becomes active upon C-terminal cleavage of its inactive precursor hCAP18. In addition to antimicrobial action, it also functions as an innate immune system stimulant in many tissues of the body. Here we report that hCAP18 and LL-37 are expressed in all organs of the human body that were studied with the highest basic levels being expressed in the GI tract and the brain. Its expression and functional role in the central nerve system (CNS) has not previously been reported. We found increased expression of LL-37 in IFNγ-stimulated human astrocytes and their surrogate U373 cells, as well as in LPS/IFNγ-stimulated human microglia and their surrogate monocyte-derived THP-1 cells. We found that treatment of microglia, astrocytes, THP-1 cells and U373 cells with LL-37 induced secretion of the inflammatory cytokines IL-1β and IL-6; the chemokines IL-8 and CCL-2, and other materials toxic to human neuroblastoma SH-SY5Y cells. The mechanism of LL-37 stimulation involves activation of intracellular proinflammatory pathways involving phospho-P38 MAP kinase and phospho-NFκB proteins. We blocked the inflammatory stimulant action of LL-37 by removing it with an anti-LL-37 antibody. The inflammatory effect was also prevented by treatment with inhibitors of PKC, PI3K and MEK-1/2 as well as with the intracellular Ca(2+)-chelator, BAPTA-AM. This indicates involvement of these intracellular pathways. Our data suggest that LL-37, in addition to its established roles, may play a role in the chronic neuroinflammation which is observed in neurodegenerative diseases such as Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Moonhee Lee
- Kinsmen Laboratory of Neurological Research, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Xiaolei Shi
- Kinsmen Laboratory of Neurological Research, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Annelise E Barron
- Department of Bioengineering, Stanford University, Stanford, CA 94305, United States
| | - Edith McGeer
- Kinsmen Laboratory of Neurological Research, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Patrick L McGeer
- Kinsmen Laboratory of Neurological Research, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3.
| |
Collapse
|
146
|
Zhu Y, Fotinos A, Mao LL, Atassi N, Zhou EW, Ahmad S, Guan Y, Berry JD, Cudkowicz ME, Wang X. Neuroprotective agents target molecular mechanisms of disease in ALS. Drug Discov Today 2015; 20:65-75. [DOI: 10.1016/j.drudis.2014.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/02/2014] [Accepted: 08/31/2014] [Indexed: 12/14/2022]
|
147
|
Cloutier F, Marrero A, O'Connell C, Morin P. MicroRNAs as potential circulating biomarkers for amyotrophic lateral sclerosis. J Mol Neurosci 2014; 56:102-12. [PMID: 25433762 DOI: 10.1007/s12031-014-0471-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 01/04/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a condition primarily characterized by the selective loss of upper and lower motor neurons. Motor neuron loss gives rise to muscle tissue malfunctions, including weakness, spasticity, atrophy, and ultimately paralysis, with death typically due to respiratory failure within 2 to 5 years of symptoms' onset. The mean delay in time from presentation to diagnosis remains at over 1 year. Biomarkers are urgently needed to facilitate ALS diagnosis and prognosis as well as to act as indicators of therapeutic response in clinical trials. MicroRNAs (miRNAs) are small molecules that can influence posttranscriptional gene expression of a variety of transcript targets. Interestingly, miRNAs can be released into the circulation by pathologically affected tissues. This review presents therapeutic and diagnostic challenges associated with ALS, highlights the potential role of miRNAs in ALS, and discusses the diagnostic potential of these molecules in identifying ALS-specific miRNAs or in distinguishing between the various genotypic and phenotypic forms of ALS.
Collapse
Affiliation(s)
- Frank Cloutier
- Institut de l'Atlantique en Neurosciences Atlantic Institute, Vitalité Health Network, Centre Hospitalier Universitaire Dr Georges-L.-Dumont/Dr. Georges-L.-Dumont University Hospital Centre, Moncton, NB, Canada,
| | | | | | | |
Collapse
|
148
|
Butovsky O, Jedrychowski MP, Cialic R, Krasemann S, Murugaiyan G, Fanek Z, Greco DJ, Wu PM, Doykan CE, Kiner O, Lawson RJ, Frosch MP, Pochet N, Fatimy RE, Krichevsky AM, Gygi SP, Lassmann H, Berry J, Cudkowicz ME, Weiner HL. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann Neurol 2014; 77:75-99. [PMID: 25381879 DOI: 10.1002/ana.24304] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 10/06/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate miR-155 in the SOD1 mouse model and human sporadic and familial amyotrophic lateral sclerosis (ALS). METHODS NanoString microRNA, microglia and immune gene profiles, protein mass spectrometry, and RNA-seq analyses were measured in spinal cord microglia, splenic monocytes, and spinal cord tissue from SOD1 mice and in spinal cord tissue of familial and sporadic ALS. miR-155 was targeted by genetic ablation or by peripheral or centrally administered anti-miR-155 inhibitor in SOD1 mice. RESULTS In SOD1 mice, we found loss of the molecular signature that characterizes homeostatic microglia and increased expression of miR-155. There was loss of the microglial molecules P2ry12, Tmem119, Olfml3, transcription factors Egr1, Atf3, Jun, Fos, and Mafb, and the upstream regulators Csf1r, Tgfb1, and Tgfbr1, which are essential for microglial survival. Microglia biological functions were suppressed including phagocytosis. Genetic ablation of miR-155 increased survival in SOD1 mice by 51 days in females and 27 days in males and restored the abnormal microglia and monocyte molecular signatures. Disease severity in SOD1 males was associated with early upregulation of inflammatory genes, including Apoe in microglia. Treatment of adult microglia with apolipoprotein E suppressed the M0-homeostatic unique microglia signature and induced an M1-like phenotype. miR-155 expression was increased in the spinal cord of both familial and sporadic ALS. Dysregulated proteins that we identified in human ALS spinal cord were restored in SOD1(G93A) /miR-155(-/-) mice. Intraventricular anti-miR-155 treatment derepressed microglial miR-155 targeted genes, and peripheral anti-miR-155 treatment prolonged survival. INTERPRETATION We found overexpression of miR-155 in the SOD1 mouse and in both sporadic and familial human ALS. Targeting miR-155 in SOD1 mice restores dysfunctional microglia and ameliorates disease. These findings identify miR-155 as a therapeutic target for the treatment of ALS.
Collapse
Affiliation(s)
- Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02112
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Malaspina A, Puentes F, Amor S. Disease origin and progression in amyotrophic lateral sclerosis: an immunology perspective. Int Immunol 2014; 27:117-29. [DOI: 10.1093/intimm/dxu099] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
150
|
Peng L, Parpura V, Verkhratsky A. EDITORIAL Neuroglia as a Central Element of Neurological Diseases: An Underappreciated Target for Therapeutic Intervention. Curr Neuropharmacol 2014; 12:303-7. [PMID: 25342938 PMCID: PMC4207070 DOI: 10.2174/1570159x12999140829152550] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroglia of the central nervous system (CNS), represented by cells of neural (astrocytes, oligodendrocytes and NG2 glial cells) and myeloid (microglia) origins are fundamental for homeostasis of the nervous tissue. Astrocytes are critical for the development of the CNS, they are indispensable for synaptogenesis, and they define structural organisation of the nervous tissue, as well as the generation and maintenance of CNS-blood and cerebrospinal fluid-blood barriers. Astroglial cells control homeostasis of ions and neurotransmitters and provide neurones with metabolic support. Oligodendrocytes, through the process of myelination, as well as by homoeostatic support of axons provide for interneuronal connectivity. The NG2 cells receive direct synaptic inputs, and might be important elements of adult remyelination. Microglial cells, which originate from foetal macrophages invading the brain early in embryogenesis, shape the synaptic connections through removing of redundant synapses and phagocyting apoptotic neurones. Neuroglia also form the defensive system of the CNS through complex and context-specific programmes of activation, known as reactive gliosis. Many neurological diseases are associated with neurogliopathologies represented by asthenic and atrophic changes in glial cells that, through the loss or diminution of their homeostatic and defensive functions, assist evolution of pathology. Conceptually, neurological and psychiatric disorders can be regarded as failures of neuroglial homeostatic/
defensive responses, and, hence, glia represent a (much underappreciated) target for therapeutic intervention.
Collapse
Affiliation(s)
- Liang Peng
- Laboratory of Metabolic Brain Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, P. R. China
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Atomic Force Microscopy & Nanotechnology Laboratories, Civitan International Research Center, Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL 35294, USA ; Department of Biotechnology, University or Rijeka, 51000 Rijeka, Croatia
| | - Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, Manchester, UK ; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain ; University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|