1
|
Ye Q, Lu W, Li T, Li Y, Tang S, Xiao P, Wei T, Zhao J, Wang Z, Huang J. Edaravone attenuates cerebral inflammation by inhibiting mast cells degranulation via ROS/STIM1 signaling pathway in HIE model. Int Immunopharmacol 2025; 159:114880. [PMID: 40394799 DOI: 10.1016/j.intimp.2025.114880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
Mast cells (MCs) degranulation is responsible for the occurrence and development of neuroinflammation after hypoxic-ischemic encephalopathy (HIE). Stromal interaction molecule 1 (STIM1) serves as a Ca2+ sensor on the endoplasmic reticulum. It has been demonstrated that the supression of STIM1 impedes degranulation of MCs in numerous prior investigations. This study aimed to explore the impact of edaravone, an oxygen radical scavenger, on MCs degranulation in HIE rat model, and to explore the contribution of reactive oxygen species (ROS)/STIM1 pathway in mediating MCs degranulation. Nine-day old undetermined gender rat pups were experienced hypoxic-ischemic (HI) injury and edaravone was administered intraperitoneally at 10 min after HI insults. CM4620, an inhibitor of STIM1, was administered intraperitoneally at 10 min after HI insults to elucidate the possible mechanisms. TTC staining, Western blot analysis, immunofluorescence staining, brain water content, cerebral blood flow, toluidine blue staining, Nissl staining, and neurobehavioral test were conducted. The results demonstrated that tryptase, STIM1, tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) were increased after HI, and edaravone significantly improved neurobehavioral outcomes, reduced brain water content, decreased infarct area, reduced the accumulation of ROS, decreased the degranulation of MCs, and downregulated the protein expression of tryptase, STIM1, IL-6 and TNF-α. CM4620 inhibited MCs degranulation and downregulated the expression of STIM1, tryptase, IL-6, TNF-α. In conclusion, the current investigation revealed that edaravone attenuates MCs degranulation and neuroinflammation, at least partially, via ROS/STIM1 pathway after HI injury.
Collapse
Affiliation(s)
- Qingqing Ye
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China; Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Weitian Lu
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China; Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Tingsong Li
- Department of Rehabilitation, Children's Hospital of Chongqing Medical University (CHCMU), Chongqing, China; National Clinical Research Center for Child Health and Disorders, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Yuan Li
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China; Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Shilong Tang
- National Clinical Research Center for Child Health and Disorders, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Pengyu Xiao
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China; Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Ting Wei
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China; Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Jiayi Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zhenhua Wang
- Technical Department of Criminal Investigation Branch, Deyang Police Office, Deyang 618000, China
| | - Juan Huang
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China; Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Prado C, Herrada AA, Hevia D, Goiry LG, Escobedo N. Role of innate immune cells in multiple sclerosis. Front Immunol 2025; 16:1540263. [PMID: 40034690 PMCID: PMC11872933 DOI: 10.3389/fimmu.2025.1540263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune, inflammatory and neurodegenerative disease affecting the central nervous system (CNS). MS is associated with a complex interplay between neurodegenerative and inflammatory processes, mostly attributed to pathogenic T and B cells. However, a growing body of preclinical and clinical evidence indicates that innate immunity plays a crucial role in MS promotion and progression. Accordingly, preclinical and clinical studies targeting different innate immune cells to control MS are currently under study, highlighting the importance of innate immunity in this pathology. Here, we reviewed recent findings regarding the role played by innate immune cells in the pathogenesis of MS. Additionally, we discuss potential new treatments for MS based on targets against innate immune components.
Collapse
Affiliation(s)
- Carolina Prado
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Andrés A. Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Daniel Hevia
- Center for Studies and Innovation in Dentistry, Facultad de Odontología, Universidad Finis Terrae, Santiago, Chile
| | - Lorna Galleguillos Goiry
- Neurology and Psychiatry Department, Clínica Alemana, Neurology and Neurosurgery Department, Clínica Dávila, Santiago, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
3
|
Patel PU, Regmi A, Dass AI, Rojas OL. Immune conversations at the border: meningeal immunity in health and disease. Front Immunol 2025; 16:1531068. [PMID: 39944687 PMCID: PMC11813769 DOI: 10.3389/fimmu.2025.1531068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/10/2025] [Indexed: 05/09/2025] Open
Abstract
The brain and spinal cord, collectively known as the central nervous system, are encapsulated by an overlapping series of membranes known as the meninges. Once considered primarily a physical barrier for central nervous system protection, the bordering meninges are now recognized as highly immunologically active. The meninges host diverse resident immune cells and serve as a critical interface with peripheral immunity, playing multifaceted roles in maintaining central nervous system homeostasis, responding to pathogenic threats, and neurological disorders. This review summarizes recent advancements in our understanding of meningeal immunity including its structural composition, physiological functions, and role in health and disease.
Collapse
Affiliation(s)
- Preya U. Patel
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Aryan Regmi
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Angelina I. Dass
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Olga L. Rojas
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
4
|
Boziki M, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Tzitiridou-Chatzopoulou M, Doulberis M, Kazakos E, Deretzi G, Grigoriadis N, Kountouras J. Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut-Brain Axis and Helicobacter pylori Infection. Neurol Int 2024; 16:1750-1778. [PMID: 39728753 DOI: 10.3390/neurolint16060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND The innate immune response aims to prevent pathogens from entering the organism and/or to facilitate pathogen clearance. Innate immune cells, such as macrophages, mast cells (MCs), natural killer cells and neutrophils, bear pattern recognition receptors and are thus able to recognize common molecular patterns, such as pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), the later occurring in the context of neuroinflammation. An inflammatory component in the pathology of otherwise "primary cerebrovascular and neurodegenerative" disease has recently been recognized and targeted as a means of therapeutic intervention. Activated MCs are multifunctional effector cells generated from hematopoietic stem cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens and/or tissue destruction. METHODS This review aims to summarize evidence of MC implication in the pathogenesis of neurodegenerative diseases, namely, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. RESULTS In view of recent evidence that the gut-brain axis may be implicated in the pathogenesis of neurodegenerative diseases and the characterization of the neuroinflammatory component in the pathology of these diseases, this review also focuses on MCs as potential mediators in the gut-brain axis bi-directional communication and the possible role of Helicobacter pylori, a gastric pathogen known to alter the gut-brain axis homeostasis towards local and systemic pro-inflammatory responses. CONCLUSION As MCs and Helicobacter pylori infection may offer targets of intervention with potential therapeutic implications for neurodegenerative disease, more clinical and translational evidence is needed to elucidate this field.
Collapse
Affiliation(s)
- Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Nella
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Midwifery Department, School of Healthcare Sciences, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, 5001 Aarau, Switzerland
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgia Deretzi
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Department of Neurology, Papageorgiou General Hospital, 54629 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
5
|
Rajalingam A, Ganjiwale A. Identification of common genetic factors and immune-related pathways associating more than two autoimmune disorders: implications on risk, diagnosis, and treatment. Genomics Inform 2024; 22:10. [PMID: 38956704 PMCID: PMC11221123 DOI: 10.1186/s44342-024-00004-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/22/2023] [Indexed: 07/04/2024] Open
Abstract
Autoimmune disorders (ADs) are chronic conditions resulting from failure or breakdown of immunological tolerance, resulting in the host immune system attacking its cells or tissues. Recent studies report shared effects, mechanisms, and evolutionary origins among ADs; however, the possible factors connecting them are unknown. This study attempts to identify gene signatures commonly shared between different autoimmune disorders and elucidate their molecular pathways linking the pathogenesis of these ADs using an integrated gene expression approach. We employed differential gene expression analysis across 19 datasets of whole blood/peripheral blood cell samples with five different autoimmune disorders (rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, Crohn's disease, and type 1 diabetes) to get nine key genes-EGR1, RUNX3, SMAD7, NAMPT, S100A9, S100A8, CYBB, GATA2, and MCEMP1 that were primarily involved in cell and leukocyte activation, leukocyte mediated immunity, IL-17, AGE-RAGE signaling in diabetic complications, prion disease, and NOD-like receptor signaling confirming its role in immune-related pathways. Combined with biological interpretations such as gene ontology (GO), pathway enrichment, and protein-protein interaction (PPI) network, our current study sheds light on the in-depth research on early detection, diagnosis, and prognosis of different ADs.
Collapse
Affiliation(s)
- Aruna Rajalingam
- Department of Life Science, Bangalore University, Bangalore, Karnataka, 560056, India
| | - Anjali Ganjiwale
- Department of Life Science, Bangalore University, Bangalore, Karnataka, 560056, India.
| |
Collapse
|
6
|
Sagües‐Sesé E, García‐Casares N, Álvarez‐Twose I. Cognitive, neuropsychiatric and neurological alterations in mastocytosis: A systematic review. Clin Transl Allergy 2023; 13:e12319. [PMID: 38146805 PMCID: PMC10718195 DOI: 10.1002/clt2.12319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Mastocytosis manifests with multisystemic symptoms, often involving the nervous system. Numerous cognitive, neuropsychiatric and neurological alterations have been reported in multiple observational studies. METHODS We performed a qualitative systematic literature review of reported data consulting the electronic databases Medline, Scopus, Web of Science, Cochrane, and BASE until June 2023. RESULTS We selected 24 studies in which the majority showed that a high proportion of mastocytosis patients suffer cognitive, neuropsychiatric and neurological alterations. The most common disorders and estimated ranges of frequency observed in adults were depression (68%-75%), anxiety, high stress or irritability (27%-54%), cognitive impairment (27%-39%, primarily affecting memory skills), and headaches (55%-69%). Attention challenges and learning difficulties were reported in children at a rate of 13%, while neurodevelopmental disorders occurred at rates of 8%-12%. Frequent white abnormalities in mastocytosis patients with concomitant psychocognitive symptoms have been reported although neuroimaging studies have been performed rarely in this population. CONCLUSION Further studies with more comprehensive and homogeneous evaluations and neuroimaging and histological analysis should be performed for a better understanding of these manifestations. An earlier detection and proper management of these symptoms could greatly improve the quality of life of these patients.
Collapse
Affiliation(s)
- Elena Sagües‐Sesé
- Departamento de Medicina, Facultad de MedicinaUniversidad de MálagaMálagaSpain
| | - Natalia García‐Casares
- Departamento de Medicina, Facultad de MedicinaUniversidad de MálagaMálagaSpain
- Centro de Investigaciones Médico‐Sanitarias (CIMES), Fundación General de la Universidad de Málaga, Universidad de MálagaMálagaSpain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND)MálagaSpain
| | - Ivan Álvarez‐Twose
- Instituto de Estudios de Mastocitosis de Castilla‐La Mancha (CLMast)Reference Center for Mastocytosis and CIBERONCToledoSpain
- Spanish Network on Mastocytosis (REMA)Toledo and SalamancaSpain
- Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)ToledoSpain
| |
Collapse
|
7
|
Mamuladze T, Kipnis J. Type 2 immunity in the brain and brain borders. Cell Mol Immunol 2023; 20:1290-1299. [PMID: 37429945 PMCID: PMC10616183 DOI: 10.1038/s41423-023-01043-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/16/2023] [Indexed: 07/12/2023] Open
Abstract
Recent research in neuroimmunology has revolutionized our understanding of the intricate interactions between the immune system and the central nervous system (CNS). The CNS, an "immune-privileged organ", is now known to be intimately connected to the immune system through different cell types and cytokines. While type 2 immune responses have traditionally been associated with allergy and parasitic infections, emerging evidence suggests that these responses also play a crucial role in CNS homeostasis and disease pathogenesis. Type 2 immunity encompasses a delicate interplay among stroma, Th2 cells, innate lymphoid type 2 cells (ILC2s), mast cells, basophils, and the cytokines interleukin (IL)-4, IL-5, IL-13, IL-25, TSLP and IL-33. In this review, we discuss the beneficial and detrimental roles of type 2 immune cells and cytokines in CNS injury and homeostasis, cognition, and diseases such as tumors, Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Tornike Mamuladze
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Immunology Graduate Program, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Immunology Graduate Program, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Cocchi M, Mondo E, Romeo M, Traina G. The Inflammatory Conspiracy in Multiple Sclerosis: A Crossroads of Clues and Insights through Mast Cells, Platelets, Inflammation, Gut Microbiota, Mood Disorders and Stem Cells. Int J Mol Sci 2022; 23:ijms23063253. [PMID: 35328673 PMCID: PMC8950240 DOI: 10.3390/ijms23063253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 01/27/2023] Open
Abstract
Multiple Sclerosis is a chronic neurological disease characterized by demyelination and axonal loss. This pathology, still largely of unknown etiology, carries within it a complex series of etiopathogenetic components of which it is difficult to trace the origin. An inflammatory state is likely to be the basis of the pathology. Crucial elements of the inflammatory process are the interactions between platelets and mast cells as well as the bacterial component of the intestinal microbiota. In addition, the involvement of mast cells in autoimmune demyelinating diseases has been shown. The present work tries to hang up on that Ariadne’s thread which, in the molecular complexity of the interactions between mast cells, platelets, microbiota and inflammation, characterizes Multiple Sclerosis and attempts to bring the pathology back to the causal determinism of psychopathological phenomenology. Therefore, we consider the possibility that the original error of Multiple Sclerosis can be investigated in the genetic origin of the depressive pathology.
Collapse
Affiliation(s)
- Massimo Cocchi
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (M.C.); (E.M.)
| | - Elisabetta Mondo
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, 40064 Bologna, Italy; (M.C.); (E.M.)
| | - Marcello Romeo
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy;
| | - Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
- Correspondence:
| |
Collapse
|
9
|
Germundson DL, Nagamoto-Combs K. Potential Role of Intracranial Mast Cells in Neuroinflammation and Neuropathology Associated with Food Allergy. Cells 2022; 11:738. [PMID: 35203387 PMCID: PMC8870724 DOI: 10.3390/cells11040738] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/29/2022] Open
Abstract
Mast cells (MCs) are the major effector cells of allergic responses and reside throughout the body, including in the brain and meninges. Previously, we showed in a mouse model of subclinical cow's milk allergy that brain MC numbers were elevated in sensitized mice. However, the neurophysiological consequences of intracranial MC accumulation and activation are unclear. We hypothesized that centrally recruited MCs in sensitized mice could be activated by the allergen via the IgE/FcεRI mechanism and increase the blood-brain barrier (BBB) permeability to promote neuroinflammation. Furthermore, we suspected that repeated allergen exposure could sustain MC activation. To investigate our hypothesis, we sensitized C57BL6/J mice to a bovine whey allergen, β-lactoglobulin (BLG), and subsequently placed them on a whey-containing diet for two weeks. MC activity and associated changes in the brain were examined. BLG-sensitized mice showed mobility changes and depression-like behavior with significantly increased MC numbers and histamine levels in select brain regions. IgG extravasation and perivascular astrogliosis were also evident. Importantly, myelin staining revealed cortical demyelination in the BLG-sensitized mice, suggesting a potential neural substrate for their behavioral changes. Our findings support the ability of brain MCs to release histamine and other mediators to increase BBB permeability and facilitate neuroinflammatory responses in the brain.
Collapse
Affiliation(s)
- Danielle L. Germundson
- Clinical and Translational Sciences Graduate Program, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202-9037, USA;
| | - Kumi Nagamoto-Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202-9037, USA
| |
Collapse
|
10
|
Noto CN, Hoft SG, DiPaolo RJ. Mast Cells as Important Regulators in Autoimmunity and Cancer Development. Front Cell Dev Biol 2021; 9:752350. [PMID: 34712668 PMCID: PMC8546116 DOI: 10.3389/fcell.2021.752350] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/17/2021] [Indexed: 01/04/2023] Open
Abstract
Mast cells are an essential part of the immune system and are best known as important modulators of allergic and anaphylactic immune responses. Upon activation, mast cells release a multitude of inflammatory mediators with various effector functions that can be both protective and damage-inducing. Mast cells can have an anti-inflammatory or pro-inflammatory immunological effect and play important roles in regulating autoimmune diseases including rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. Importantly, chronic inflammation and autoimmunity are linked to the development of specific cancers including pancreatic cancer, prostate cancer, colorectal cancer, and gastric cancer. Inflammatory mediators released from activated mast cells regulate immune responses and promote vascular permeability and the recruitment of immune cells to the site of inflammation. Mast cells are present in increased numbers in tissues affected by autoimmune diseases as well as in tumor microenvironments where they co-localize with T regulatory cells and T effector cells. Mast cells can regulate immune responses by expressing immune checkpoint molecules on their surface, releasing anti-inflammatory cytokines, and promoting vascularization of solid tumor sites. As a result of these immune modulating activities, mast cells have disease-modifying roles in specific autoimmune diseases and cancers. Therefore, determining how to regulate the activities of mast cells in different inflammatory and tumor microenvironments may be critical to discovering potential therapeutic targets to treat autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Christine N Noto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Stella G Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Richard J DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
11
|
Chacko G, Patel S, Galor A, Kumar N. Heat Exposure and Multiple Sclerosis-A Regional and Temporal Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18115962. [PMID: 34199394 PMCID: PMC8199586 DOI: 10.3390/ijerph18115962] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is a neurological disorder that progressively distorts the myelination of axons within the central nervous system (CNS). Increased core body temperature or metabolism as a result of exercise are common causes of short-term exacerbations of neurological symptoms in MS. About 60–80% of patients with MS experience a worsening of their symptoms when exposed to heat. In comparison, less data are available on the relationship between ambient meteorological conditions (e.g., temperature and relative humidity (RH)) and fluctuations in such variables in relation to MS symptoms. Thus, this study examined associations between time-lagged exposure to meteorological conditions and risk of a clinic visit due to MS among US veterans between 2010 and 2013. This study leveraged data from the Veterans Affairs (VA) and National Climactic Data Center (NCDC) for the continental US, partitioned into eight climate zones. We used a case crossover design to assess the risk of a MS clinic visit with respect to several meteorological conditions. Location-specific time-lagged daily (ambient) exposure to temperature, RH, and temperature variations (standard deviation (SD) of temperature) were computed (up to 30 days) for each case (i.e., day of MS visit) and control (a randomly assigned date ± 90–270 days prior to visit). Statistical analyses were conducted to examine independent associations between the selected meteorological conditions and risk of MS visits at the national and regional levels. A total of 533,066 patient visits received a MS diagnosis (International Classifications of Diseases (ICD)-9 code = 340). The Northeast (NE) and Upper Midwest (UMW) regions reported the highest frequency of clinic visits due to MS. Clinic visits were 9% more likely to occur in the spring, summer, and fall months (March–October) than in the winter (OR = 1.089; 95% CI = 1.076–1.103; p < 0.01). In the univariate analyses, the SD of temperature, temperature, and temperature–RH interaction were positively associated with an elevated risk of a MS clinic visit, while the RH was negatively associated with the risk for a clinic visit. In multivariate analyses, the strongest association of a MS clinic visit was observed with the SD of the temperature (OR = 1.012; 95% CI 1.008–1.017; p < 0.01). These associations between MS clinic visits and meteorological conditions varied across climate regions, with the strongest associations being observed in the LMW, UMW, DSW, and NE zones. The SD of the temperature was again the strongest associated predictor when examined regionally. Temperature variations and temperature–RH interactions (a proxy of the heat index) showed significant associations with MS clinic visits. These associations varied across climate regions when examined geographically. Our findings have implications for the management of MS in severe or recurrent cases, especially considering the impending changes in the daily temperature variations and intensity of the heatwaves expected with the intensification of global warming.
Collapse
Affiliation(s)
- Gill Chacko
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Sneh Patel
- Ophthalmology, Miami Veterans Affairs Medical Center, Miami, FL 33136, USA; (S.P.); (A.G.)
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anat Galor
- Ophthalmology, Miami Veterans Affairs Medical Center, Miami, FL 33136, USA; (S.P.); (A.G.)
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Research Services, Miami Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Naresh Kumar
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Correspondence: ; Tel.: +1-305-243-4854
| |
Collapse
|
12
|
El Ansari YS, Kanagaratham C, Lewis OL, Oettgen HC. IgE and mast cells: The endogenous adjuvant. Adv Immunol 2020; 148:93-153. [PMID: 33190734 DOI: 10.1016/bs.ai.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mast cells and IgE are most familiar as the effectors of type I hypersensitivity reactions including anaphylaxis. It is becoming clear however that this pair has important immunomodulatory effects on innate and adaptive cells of the immune system. In this purview, they act as endogenous adjuvants to ignite evolving immune responses, promote the transition of allergic disease into chronic illness and disrupt the development of active mechanisms of tolerance to ingested foods. Suppression of IgE-mediated mast cell activation can be exerted by molecules targeting IgE, FcɛRI or signaling kinases including Syk, or by IgG antibodies acting via inhibitory Fcγ receptors. In 2015 we reviewed the evidence for the adjuvant functions of mast cells. This update includes the original text, incorporates some important developments in the field over the past five years and discusses how interventions targeting these pathways might have promise in the development of strategies to treat allergic disease.
Collapse
Affiliation(s)
- Yasmeen S El Ansari
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Owen L Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
13
|
Pinke KH, Zorzella-Pezavento SFG, Lara VS, Sartori A. Should mast cells be considered therapeutic targets in multiple sclerosis? Neural Regen Res 2020; 15:1995-2007. [PMID: 32394947 PMCID: PMC7716037 DOI: 10.4103/1673-5374.282238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/19/2019] [Accepted: 01/16/2020] [Indexed: 12/23/2022] Open
Abstract
Mast cells are immune cells of the myeloid lineage that are found throughout the body, including the central nervous system. They perform many functions associated with innate and specific immunity, angiogenesis, and vascular homeostasis. Moreover, they have been implicated in a series of pathologies (e.g., hypersensitivity reactions, tumors, and inflammatory disorders). In this review, we propose that this cell could be a relevant therapeutic target in multiple sclerosis, which is a central nervous system degenerative disease. To support this proposition, we describe the general biological properties of mast cells, their contribution to innate and specific immunity, and the participation of mast cells in the various stages of multiple sclerosis and experimental autoimmune encephalomyelitis development. The final part of this review is dedicated to an overview of the available mast cells immunomodulatory drugs and their activity on multiple sclerosis and experimental autoimmune encephalomyelitis, including our own experience related to the effect of ketotifen fumarate on experimental autoimmune encephalomyelitis evolution.
Collapse
Affiliation(s)
- Karen Henriette Pinke
- Institute of Biosciences, Department of Microbiology and Immunology, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | | | - Vanessa Soares Lara
- Bauru School of Dentistry, Department of Surgery, Stomatology, Pathology and Radiology, University of São Paulo, Bauru, São Paulo, Brazil
| | - Alexandrina Sartori
- Institute of Biosciences, Department of Microbiology and Immunology, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
14
|
Fresegna D, Bullitta S, Musella A, Rizzo FR, De Vito F, Guadalupi L, Caioli S, Balletta S, Sanna K, Dolcetti E, Vanni V, Bruno A, Buttari F, Stampanoni Bassi M, Mandolesi G, Centonze D, Gentile A. Re-Examining the Role of TNF in MS Pathogenesis and Therapy. Cells 2020; 9:cells9102290. [PMID: 33066433 PMCID: PMC7602209 DOI: 10.3390/cells9102290] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a common neurological disorder of putative autoimmune origin. Clinical and experimental studies delineate abnormal expression of specific cytokines over the course of the disease. One major cytokine that has been shown to play a pivotal role in MS is tumor necrosis factor (TNF). TNF is a pleiotropic cytokine regulating many physiological and pathological functions of both the immune system and the central nervous system (CNS). Convincing evidence from studies in human and experimental MS have demonstrated the involvement of TNF in various pathological hallmarks of MS, including immune dysregulation, demyelination, synaptopathy and neuroinflammation. However, due to the complexity of TNF signaling, which includes two-ligands (soluble and transmembrane TNF) and two receptors, namely TNF receptor type-1 (TNFR1) and type-2 (TNFR2), and due to its cell- and context-differential expression, targeting the TNF system in MS is an ongoing challenge. This review summarizes the evidence on the pathophysiological role of TNF in MS and in different MS animal models, with a special focus on pharmacological treatment aimed at controlling the dysregulated TNF signaling in this neurological disorder.
Collapse
Affiliation(s)
- Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, 00166 Roma, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Francesca De Vito
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Silvia Caioli
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Sara Balletta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Krizia Sanna
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Mario Stampanoni Bassi
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, 00166 Roma, Italy
| | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
- Correspondence: ; Tel.: +39-06-7259-6010; Fax: +39-06-7259-6006
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
| |
Collapse
|
15
|
Ribatti D, Tamma R, Annese T. Mast cells and angiogenesis in multiple sclerosis. Inflamm Res 2020; 69:1103-1110. [PMID: 32808153 DOI: 10.1007/s00011-020-01394-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/06/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease, characterized by multiple demyelination of axons in both white and gray matter in the Central Nervous System (CNS). There is increasing evidence to support the notion that angiogenesis and chronic inflammation are mutually related. Different immune cells, including monocytes-macrophages, lymphocytes, neutrophils, mast cells (MCs) and dendritic cells are able to secrete an array of angiogenic cytokines, which promote growth, migration, and activation of endothelial cells. MCs play various roles in MS pathogenesis, influencing the innate immune response in peripheral tissues and in CNS. The aim of this review article is to discuss the role of MCs in MS pathogenesis with particular reference to the involvement of these inflammatory cells in the angiogenic processes occurring during MS.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy.
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
16
|
Song JH, Yu DH, Hwang TS, Seung BJ, Sur JH, Kim YJ, Jung DI. Expression of platelet-derived growth factor receptor-α/ß, vascular endothelial growth factor receptor-2, c-Abl, and c-Kit in canine granulomatous meningoencephalitis and necrotizing encephalitis. Vet Med Sci 2020; 6:965-974. [PMID: 32585777 PMCID: PMC7738704 DOI: 10.1002/vms3.314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Accepted: 05/27/2020] [Indexed: 01/21/2023] Open
Abstract
Background Given the active research on targeted therapy using tyrosine kinase (TK) inhibitors (TKIs) in the field of oncology, further studies have recently been conducted to evaluate their use in autoimmune disorders. Based on immunological investigations, previous studies have suggested that granulomatous meningoencephalomyelitis (GME) and necrotizing encephalomyelitis (NE) are similar to multiple sclerosis (MS), which is a human autoimmune demyelinating central nervous system disease. Objectives Considering this perspective, we hypothesized that canine GME and NE have significant expression of one or more TKs, which are associated with human MS pathogenesis. Methods To determine the possible use of conventional multi‐targeted TKIs as a treatment for canine GME and NE, we characterized the immunohistochemical expression of platelet‐derived growth factor receptor (PDGFR)‐α, PDGFR‐ß, vascular endothelial growth factor receptor (VEGFR)‐2, c‐Abl and c‐Kit in GME and NE samples. Results Histological samples from four dogs with GME and three with NE were retrieved. All samples stained positive for PDGFR‐ß (7/7 [100%]). PDGFR‐α and c‐Kit were expressed in 3/7 (42.8%) samples each. c‐Abl was identified in 2/7 (28.5%) samples; no sample showed VEGFR‐2 (0%) expression. Co‐expression of TKs was identified in 6/7 (85.7%) dogs. Conclusions All samples were positive for at least one or more of PDGFR‐α, PDGFR‐ß, c‐Kit and c‐Abl, which are known as the target TKs of conventional multi‐targeted TKIs. Their presence does suggest that these TKs may play a role in the pathogenesis of GME and NE. Therefore, multi‐targeted TKIs may provide benefits in the treatment of canine GME and NE by suppressing the activity of these TKs.
Collapse
Affiliation(s)
- Joong-Hyun Song
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Do-Hyeon Yu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Tae-Sung Hwang
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Byung-Joon Seung
- Department of Pathobiology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jung-Hyang Sur
- Department of Pathobiology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Young Joo Kim
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, 91766-1854, USA
| | - Dong-In Jung
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| |
Collapse
|
17
|
Byun S, Myung W, Kim H, Lee H. Association between diurnal temperature range and emergency department visits for multiple sclerosis: A time-stratified case-crossover study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 720:137565. [PMID: 32145628 DOI: 10.1016/j.scitotenv.2020.137565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 06/10/2023]
Abstract
Although multiple sclerosis (MS) has been the leading cause of neurologically-induced disability in young adults, risk factors for the relapse and acute aggravation of MS remain unclear. A few studies have suggested a possible role of temperature changes on the relapse and acute aggravation of MS. We investigated the association between short-term exposure to wide diurnal temperature ranges (DTRs) and acute exacerbation of MS requiring an emergency department (ED) visit. A total of 1265 patients visited EDs for acute aggravation of MS as the primary disease in Seoul between 2008 and 2014 from the national emergency database. We conducted a conditional logistic regression analysis of the time-stratified case-crossover design to compare DTRs on the ED visit days for MS and those on control days matched according to the day of the week, month, and year. We examined possible associations with other temperature-related variables (ambient temperature, between-day temperature change, and sunlight hours). Short-term exposure to wide DTRs immediately increased the risk of ED visits for MS. Especially, 2-day average (lag0-1) DTR levels on the day of and one day prior to ED visits exhibited the strongest association (an 8.81% [95% CI: 3.46%-14.44%] change in the odds ratio per 1 °C increase in the DTR). Other temperature-related variables were not associated with MS aggravation. Our results suggest that exposure to wider DTR may increase the risk of acute exacerbation of MS. Given the increasing societal burden of MS and the increasing temperature variability due to climate change, further studies are required.
Collapse
Affiliation(s)
- Seonjeong Byun
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, Republic of Korea; Department of Neuropsychiatry, Uijeongbu St. Mary's Hospital, Gyeonggi-do, Republic of Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Bundang-gu, Seongnam, Republic of Korea
| | - Ho Kim
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Hyewon Lee
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Bundang-gu, Seongnam, Republic of Korea; Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Shelestak J, Singhal N, Frankle L, Tomor R, Sternbach S, McDonough J, Freeman E, Clements R. Increased blood-brain barrier hyperpermeability coincides with mast cell activation early under cuprizone administration. PLoS One 2020; 15:e0234001. [PMID: 32511268 PMCID: PMC7279587 DOI: 10.1371/journal.pone.0234001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
The cuprizone induced animal model of demyelination is characterized by demyelination in many regions of the brain with high levels of demyelination in the corpus callosum as well as changes in neuronal function by 4–6 weeks of exposure. The model is used as a tool to study demyelination and subsequent degeneration as well as therapeutic interventions on these effects. Historically, the cuprizone model has been shown to contain no alterations to blood-brain barrier integrity, a key feature in many diseases that affect the central nervous system. Cuprizone is generally administered for 4–6 weeks to obtain maximal demyelination and degeneration. However, emerging evidence has shown that the effects of cuprizone on the brain may occur earlier than measurable gross demyelination. This study sought to investigate changes to blood-brain barrier permeability early in cuprizone administration. Results showed an increase in blood-brain barrier permeability and changes in tight junction protein expression as early as 3 days after beginning cuprizone treatment. These changes preceded glial morphological activation and demyelination known to occur during cuprizone administration. Increases in mast cell presence and activity were measured alongside the increased permeability implicating mast cells as a potential source for the blood-brain barrier disruption. These results provide further evidence of blood-brain barrier alterations in the cuprizone model and a target of therapeutic intervention in the prevention of cuprizone-induced pathology. Understanding how mast cells become activated under cuprizone and if they contribute to blood-brain barrier alterations may give further insight into how and when the blood-brain barrier is affected in CNS diseases. In summary, cuprizone administration causes an increase in blood-brain barrier permeability and this permeability coincides with mast cell activation.
Collapse
Affiliation(s)
- John Shelestak
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
- * E-mail:
| | - Naveen Singhal
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Lana Frankle
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Riely Tomor
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Sarah Sternbach
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Jennifer McDonough
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Ernest Freeman
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Robert Clements
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| |
Collapse
|
19
|
Vattathara JJ, Prakash O, Subhramanian S, Satheeshkumar MK, Xavier T, Anil M, Pillai GS, Anandakuttan A, Radhakrishnan S, Sivanarayanan TB, Akk U, Mohan CG, Menon KN. Substrate Specific Inhibitor Designed against the Immunomodulator GMF-beta Reversed the Experimental Autoimmune Encephalomyelitis. Sci Rep 2020; 10:3790. [PMID: 32123210 PMCID: PMC7051966 DOI: 10.1038/s41598-020-60710-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 02/14/2020] [Indexed: 01/16/2023] Open
Abstract
The concept of substrate inhibition to prevent its phosphorylation has potential in drug discovery and is envisioned to treat the autoimmune disorder multiple sclerosis (MS). Glia maturation factor-β (GMF-β) Ser83 phosphorylation by protein kinase A (PKA) is pivotal in the activation of GMF-β-p38MAPK-NFκB biochemical pathway towards proinflammatory response induction in experimental autoimmune encephalomyelitis (EAE). Using structure-based drug design, we identified the small molecule inhibitor 1-H-indazole-4yl methanol (GMFBI.1) that specifically blocked Ser83 phosphorylation site on GMF-β substrate. Using in vitro and in vivo techniques, molecular mechanism of action of GMFBI.1’s direct interaction with GMF-β substrate and prevention of its Ser83 phosphorylation was established. GMFBI.1 down regulated p38MAPK phosphorylation and NFκB expression essential for proinflammatory response. Further, GMFBI.1 administration at peak of EAE reversed clinical symptoms, immunopathology, proinflammatory cytokine response and up regulated the anti-inflammatory cytokines. Present strategy of substrate inhibition against the key immunomodulatory target has immense therapeutic potential in MS.
Collapse
Affiliation(s)
- Jane Jose Vattathara
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Ohm Prakash
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Sunitha Subhramanian
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Madathiparambil Kumaran Satheeshkumar
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Tessy Xavier
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Meenakshi Anil
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Gopal S Pillai
- Department of Ophthalmology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Anandkumar Anandakuttan
- Department of Neurology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Sureshkumar Radhakrishnan
- Department of Neurology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - T B Sivanarayanan
- Central Animal Laboratory, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Unni Akk
- Central Animal Laboratory, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India
| | - Chethampadi Gopi Mohan
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India.
| | - Krishnakumar N Menon
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi-682 041, Kerala, India.
| |
Collapse
|
20
|
Pinke KH, Zorzella-Pezavento SFG, de Campos Fraga-Silva TF, Mimura LAN, de Oliveira LRC, Ishikawa LLW, Fernandes AAH, Lara VS, Sartori A. Calming Down Mast Cells with Ketotifen: A Potential Strategy for Multiple Sclerosis Therapy? Neurotherapeutics 2020; 17:218-234. [PMID: 31463682 PMCID: PMC7007452 DOI: 10.1007/s13311-019-00775-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by extensive inflammation, demyelination, axonal loss and gliosis. Evidence indicates that mast cells contribute to immunopathogenesis of both MS and experimental autoimmune encephalomyelitis (EAE), which is the most employed animal model to study this disease. Considering the inflammatory potential of mast cells, their presence at the CNS and their stabilization by certain drugs, we investigated the effect of ketotifen fumarate (Ket) on EAE development. EAE was induced in C57BL/6 mice by immunization with MOG35-55 and the animals were injected daily with Ket from the seventh to the 17th day after disease induction. This early intervention with Ket significantly reduced disease prevalence and severity. The protective effect was concomitant with less NLRP3 inflammasome activation, rebalanced oxidative stress and also reduced T cell infiltration at the CNS. Even though Ket administration did not alter mast cell percentage at the CNS, it decreased the local CPA3 and CMA1 mRNA expression that are enzymes typically produced by these cells. Evaluation of the CNS-barrier permeability indicated that Ket clearly restored the permeability levels of this barrier. Ket also triggered an evident lymphadenomegaly due to accumulation of T cells that produced higher levels of encephalitogenic cytokines in response to in vitro stimulation with MOG. Altogether these findings reinforce the concept that mast cells are particularly relevant in MS immunopathogenesis and that Ket, a known stabilizer of their activity, has the potential to be used in MS control.
Collapse
Affiliation(s)
- Karen Henriette Pinke
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil.
| | - Sofia Fernanda Gonçalves Zorzella-Pezavento
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil
| | - Thais Fernanda de Campos Fraga-Silva
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil
| | - Luiza Ayumi Nishiyama Mimura
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil
| | - Larissa Ragozo Cardoso de Oliveira
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil
| | - Larissa Lumi Watanabe Ishikawa
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil
| | - Ana Angélica Henrique Fernandes
- Department of Chemistry and Biochemistry, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Vanessa Soares Lara
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, São Paulo, Brazil
| | - Alexandrina Sartori
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Rua Dr. Plinio Pinto e Silva, S/N, Distrito de Rubião Júnior, Botucatu, São Paulo, 18618-691, Brazil
| |
Collapse
|
21
|
Mast cells drive IgE-mediated disease but might be bystanders in many other inflammatory and neoplastic conditions. J Allergy Clin Immunol 2019; 144:S19-S30. [DOI: 10.1016/j.jaci.2019.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/11/2019] [Accepted: 07/08/2019] [Indexed: 01/05/2023]
|
22
|
Kanellopoulos JM, Delarasse C. Pleiotropic Roles of P2X7 in the Central Nervous System. Front Cell Neurosci 2019; 13:401. [PMID: 31551714 PMCID: PMC6738027 DOI: 10.3389/fncel.2019.00401] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022] Open
Abstract
The purinergic receptor P2X7 is expressed in neural and immune cells known to be involved in neurological diseases. Its ligand, ATP, is a signaling molecule that can act as a neurotransmitter in physiological conditions or as a danger signal when released in high amount by damaged/dying cells or activated glial cells. Thus, ATP is a danger-associated molecular pattern. Binding of ATP by P2X7 leads to the activation of different biochemical pathways, depending on the physiological or pathological environment. The aim of this review is to discuss various functions of P2X7 in the immune and central nervous systems. We present evidence that P2X7 may have a detrimental or beneficial role in the nervous system, in the context of neurological pathologies: epilepsy, Alzheimer’s disease, multiple sclerosis, amyotrophic lateral sclerosis, age-related macular degeneration and cerebral artery occlusion.
Collapse
Affiliation(s)
| | - Cécile Delarasse
- Inserm, Sorbonne Université, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
23
|
Desbiens L, Lapointe C, Gendron L, Gharagozloo M, Vincent L, Pejler G, Gris D, D'Orléans-Juste P. Experimental Autoimmune Encephalomyelitis Potentiates Mouse Mast Cell Protease 4-Dependent Pressor Responses to Centrally or Systemically Administered Big Endothelin-1. J Pharmacol Exp Ther 2019; 370:437-446. [PMID: 31248979 DOI: 10.1124/jpet.118.256016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/25/2019] [Indexed: 01/03/2025] Open
Abstract
Multiple sclerosis is a neurodegenerative disease affecting predominantly female patients between 20 and 45 years of age. We previously reported the significant contribution of mouse mast cell protease 4 (mMCP-4) in the synthesis of endothelin-1 (ET-1) in healthy mice and in a murine model of experimental autoimmune encephalomyelitis (EAE). In the current study, the cardiovascular effects of ET-1 and big endothelin-1 (big-ET-1) administered systemically or intrathecally were assessed in the early preclinical phase of EAE in telemetry instrumented/conscious mice. Chymase-specific enzymatic activity was also measured in the lung, brain, and mast cell extracts in vitro. Finally, the impact of EAE immunization was studied on the pulmonary and brain mRNA expression of different genes of the endothelin pathway, interleukin-33 (IL-33), and monitoring of immunoreactive tumor necrosis factor-α (TNF-α). Systemically or intrathecally administered big-ET-1 triggered increases in blood pressure in conscious mice. One week post-EAE, the pressor responses to big-ET-1 were potentiated in wild-type (WT) mice but not in mMCP-4 knockout (KO) mice. EAE triggered mMCP-4-specific activity in cerebral homogenates and peritoneal mast cells. Enhanced pulmonary, but not cerebral preproendothelin-1 and IL-33 mRNA were found in KO mice and further increased 1 week post-EAE immunization, but not in WT animals. Finally, TNF-α levels were also increased in serum from mMCP-4 KO mice, but not WT, 1 week post-EAE. Our study suggests that mMCP-4 activity is enhanced both centrally and systemically in a mouse model of EAE.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Endothelin-1/administration & dosage
- Endothelin-1/pharmacology
- Gene Knockout Techniques
- Hemodynamics/drug effects
- Injections, Spinal
- Interleukin-33/deficiency
- Interleukin-33/genetics
- Lung/drug effects
- Lung/metabolism
- Mast Cells/drug effects
- Mast Cells/metabolism
- Mice
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Serine Endopeptidases/deficiency
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Louisane Desbiens
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Catherine Lapointe
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Louis Gendron
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Marjan Gharagozloo
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Laurence Vincent
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Gunnar Pejler
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Denis Gris
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| | - Pedro D'Orléans-Juste
- Department of Pharmacology and Physiology, Medical School, Université de Sherbrooke, Sherbrooke, Québec, Canada (L.D., C.L., L.G., M.G., L.V., D.G., P.D'.O.J.); Department of Medical Biochemistry and Microbiology, Uppsala University BMC, Uppsala, Sweden (G.P.); and Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden (G.P.)
| |
Collapse
|
24
|
Javed S, Mitchell K, Sidsworth D, Sellers SL, Reutens-Hernandez J, Massicotte HB, Egger KN, Lee CH, Payne GW. Inonotus obliquus attenuates histamine-induced microvascular inflammation. PLoS One 2019; 14:e0220776. [PMID: 31437163 PMCID: PMC6706056 DOI: 10.1371/journal.pone.0220776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 07/23/2019] [Indexed: 01/14/2023] Open
Abstract
Cell-to-cell communication is a key element of microvascular blood flow control, including rapidly carrying signals through the vascular endothelium in response to local stimuli. This cell-to-cell communication is negatively impacted during inflammation through the disruption of junctional integrity. Such disruption is associated with promoting the onset of cardiovascular diseases as a result of altered microvascular blood flow regulation. Therefore, understanding the mechanisms how inflammation drives microvascular dysfunction and compounds that mitigate such inflammation and dysfunction are of great interest for development. As such we aimed to investigate extracts of mushrooms as potential novel compounds. Using intravital microscopy, the medicinal mushroom, Inonotus obliquus was observed, to attenuate histamine-induced inflammation conducted vasodilation in second-order arterioles in the gluteus maximus muscle of C57BL/6 mice. Mast cell activation by C48/80 similarly disrupted endothelial junctions and conducted vasodilation but only histamine was blocked by the histamine antagonist, pyrilamine not C48/80 suggesting the importance of mast cell activation. Data presented here supports that histamine induced inflammation is a major disruptor of junctional integrity, and highlights the important anti-inflammatory properties of Inonotus obliquus focusing future assessment of mast cells as putative target for Inonotus obliquus.
Collapse
Affiliation(s)
- Sumreen Javed
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, Canada
| | - Kevin Mitchell
- Northern Medical Program, University of Northern British Columbia, Prince George, Canada
| | - Danielle Sidsworth
- Northern Medical Program, University of Northern British Columbia, Prince George, Canada
| | - Stephanie L. Sellers
- Centre for Heart Lung Innovation & Department of Radiology, University of British Columba & St. Paul’s Hospital, Vancouver, Canada
| | - Jennifer Reutens-Hernandez
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, Canada
| | - Hugues B. Massicotte
- Ecosystem Science and Management Program, University of Northern British Columbia, Prince George, Canada
| | - Keith N. Egger
- Ecosystem Science and Management Program, University of Northern British Columbia, Prince George, Canada
| | - Chow H. Lee
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, Canada
| | - Geoffrey W. Payne
- Northern Medical Program, University of Northern British Columbia, Prince George, Canada
- * E-mail:
| |
Collapse
|
25
|
Silva BA, Ferrari CC. Cortical and meningeal pathology in progressive multiple sclerosis: a new therapeutic target? Rev Neurosci 2019; 30:221-232. [PMID: 30048237 DOI: 10.1515/revneuro-2018-0017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/04/2018] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease that involves an intricate interaction between the central nervous system and the immune system. Nevertheless, its etiology is still unknown. MS exhibits different clinical courses: recurrent episodes with remission periods ('relapsing-remitting') that can evolve to a 'secondary progressive' form or persistent progression from the onset of the disease ('primary progressive'). The discovery of an effective treatment and cure has been hampered due to the pathological and clinical heterogeneity of the disease. Historically, MS has been considered as a disease exclusively of white matter. However, patients with progressive forms of MS present with cortical lesions associated with meningeal inflammation along with physical and cognitive disabilities. The pathogenesis of the cortical lesions has not yet been fully described. Animal models that represent both the cortical and meningeal pathologies will be critical in addressing MS pathogenesis as well as the design of specific treatments. In this review, we will address the state-of-the-art diagnostic and therapeutic alternatives and the development of strategies to discover new therapeutic approaches, especially for the progressive forms.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Potosi 4240 (C1199ABB), CABA, Buenos Aires, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435 (C1405BWE), Buenos Aires, Argentina, e-mail:
| | - Carina Cintia Ferrari
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Potosi 4240 (C1199ABB), CABA, Buenos Aires, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435 (C1405BWE), Buenos Aires, Argentina
| |
Collapse
|
26
|
Type 2 Inflammatory Responses in Autoimmune Demyelination of the Central Nervous System: Recent Advances. J Immunol Res 2019; 2019:4204512. [PMID: 31205957 PMCID: PMC6530110 DOI: 10.1155/2019/4204512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/17/2019] [Indexed: 12/28/2022] Open
Abstract
Type 2 immunity has long been confined to a restricted spectrum of responses, mostly including allergic reactions to innocuous environmental triggers. However, growing evidence suggests that cells and mediators typically associated with type 2 inflammation are involved in several physiopathological conditions, such as defense against toxic substances, anticancer immunity, and autoimmune diseases. In neuromyelitis optica, an autoimmune demyelinating disorder of the spinal cord and optic nerve, eosinophils extensively infiltrate lesions in the central nervous system (CNS) and promote tissue pathology in experimental models of this disease. Next-generation sequencing of CD4+ T cells isolated from a specific subtype of multiple sclerosis plaque has uncovered an unexpectedly Th2 profile of these cells. Even mast cells and other allergic mediators have been implicated in the modulation and/or effector mechanisms of autoimmune reactions against the CNS. In this review article, the most recent developments showing the involvement of type 2 inflammatory components in CNS autoimmunity are summarised and possible lines of further investigation are discussed.
Collapse
|
27
|
Jones MK, Nair A, Gupta M. Mast Cells in Neurodegenerative Disease. Front Cell Neurosci 2019; 13:171. [PMID: 31133804 PMCID: PMC6524694 DOI: 10.3389/fncel.2019.00171] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide, yet there are currently no effective treatments. Because risk of neurodegenerative disease substantially increases with age, greater life expectancy with a concomitant aging population means more individuals will be affected in the coming decades. Thus, there is an urgent need for understanding the mechanisms driving neurodegenerative diseases in order to develop improved treatment strategies. Inflammation in the nervous system, termed “neuroinflammation,” has become increasingly recognized as being associated with neurodegenerative diseases. Early attention focused primarily on morphological changes in astrocytes and microglia; however, brain and CNS resident mast cells are now receiving attention as a result of being “first responders” to injury. Mast cells also exert profound effects on their microenvironment and neighboring cells including behavior and/or activation of astrocytes, microglia, and neurons, which, in turn, are implicated in neuroinflammation, neurogenesis and neurodegeneration. Mast cells also affect disruption/permeability of the blood brain barrier enabling toxin and immune cell entry exacerbating an inflammatory microenvironment. Here, we discuss the roles of mast cells in neuroinflammation and neurodegeneration with a focus on development and progression of four prominent neurodegenerative diseases: Alzheimer’s Disease, Parkinson’s Disease, Amyotrophic Lateral Sclerosis, and Huntington’s Disease.
Collapse
Affiliation(s)
- Michael K Jones
- Department of Medicine, Vascular Biology Center, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Archana Nair
- Department of Ophthalmology, New York University, New York, NY, United States
| | - Mihir Gupta
- Department of Neurosurgery, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
28
|
Japanese encephalitis virus neuropenetrance is driven by mast cell chymase. Nat Commun 2019; 10:706. [PMID: 30742008 PMCID: PMC6370868 DOI: 10.1038/s41467-019-08641-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/16/2019] [Accepted: 01/23/2019] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis virus (JEV) is a leading cause of viral encephalitis. However, the mechanisms of JEV penetration of the blood-brain-barrier (BBB) remain poorly understood. Mast cells (MCs) are granulated innate immune sentinels located perivascularly, including at the BBB. Here we show that JEV activates MCs, leading to the release of granule-associated proteases in vivo. MC-deficient mice display reduced BBB permeability during JEV infection compared to congenic wild-type (WT) mice, indicating that enhanced vascular leakage in the brain during JEV infection is MC-dependent. Moreover, MCs promoted increased JEV infection in the central nervous system (CNS), enhanced neurological deficits, and reduced survival in vivo. Mechanistically, chymase, a MC-specific protease, enhances JEV-induced breakdown of the BBB and cleavage of tight-junction proteins. Chymase inhibition reversed BBB leakage, reduced brain infection and neurological deficits during JEV infection, and prolonged survival, suggesting chymase is a novel therapeutic target to prevent JEV encephalitis. How Japanese encephalitis virus (JEV) penetrates the blood-brain barrier (BBB) remains unclear. Here, using a genetic mouse model and a virulent JEV strain, the authors show that perivascular mast cells (MC) mediate JEV neuroinvasion and identify the MC-protease chymase as a potential therapeutic target.
Collapse
|
29
|
Mast Cells, Basophils, and Mastocytosis. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00023-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
30
|
Ocak U, Ocak PE, Wang A, Zhang JH, Boling W, Wu P, Mo J, Zhang T, Huang L. Targeting mast cell as a neuroprotective strategy. Brain Inj 2018; 33:723-733. [PMID: 30554528 DOI: 10.1080/02699052.2018.1556807] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Mast cells (MCs) are perivascularly located immune cells of haematopoietic origin. Emerging evidences suggest that the activation of MCs play important roles in the pathogenesis of blood brain barrier disruption, neuroinflammation, and neurodegeneration. Objectives: In this review, we aimed to discuss the detrimental effects of MCs in response to various types of brain injury, as well as the therapeutic potential and neuroprotective effects of targeting the activation and degranulation of MCs, particularly in the management of the acute phase. Methods: An extensive online literature search was conducted through Pubmed/Central on March 2018. Then, we comprehensively summarized the effects of the activation of brain MCs in acute brain injury along with current pharmacological strategies targeting at the activation of MCs. Results: The review of the current literature indicated that the activation and degranulation of brain MCs significantly contribute to the acute pathological process following different types of brain injury including focal and global cerebral ischaemia, intracerebral haemorrhage, subarachnoid haemorrhage, and traumatic brain injury. Conclusions: Brain MCs significantly contribute to the acute pathological processes following brain injury. In that regard, targeting brain MCs may provide a novel strategy for neuroprotection.
Collapse
Affiliation(s)
- Umut Ocak
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA
| | - Pinar Eser Ocak
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA
| | - Annie Wang
- b Department of Anesthesiology , Loma Linda University School of Medicine , Loma Linda , CA , USA
| | - John H Zhang
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA.,b Department of Anesthesiology , Loma Linda University School of Medicine , Loma Linda , CA , USA.,c Department of Neurosurgery , Loma Linda University School of Medicine , Loma Linda , CA , USA
| | - Warren Boling
- c Department of Neurosurgery , Loma Linda University School of Medicine , Loma Linda , CA , USA
| | - Pei Wu
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA.,d Department of Neurosurgery , The First Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang , China
| | - Jun Mo
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA.,e Department of Neurosurgery, The Fourth Affiliated Hospital , School of Medicine, Zhejiang University , Yiwu , Zhejiang , China
| | - Tongyu Zhang
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA.,d Department of Neurosurgery , The First Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang , China
| | - Lei Huang
- a Department of Basic Sciences, Division of Physiology , Loma Linda University School of Medicine , Loma Linda , CA , USA.,c Department of Neurosurgery , Loma Linda University School of Medicine , Loma Linda , CA , USA
| |
Collapse
|
31
|
Le Foll B, French L. Transcriptomic Characterization of the Human Habenula Highlights Drug Metabolism and the Neuroimmune System. Front Neurosci 2018; 12:742. [PMID: 30429765 PMCID: PMC6220030 DOI: 10.3389/fnins.2018.00742] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022] Open
Abstract
Due to size and accessibility, most information about the habenula is derived from rodent studies. To better understand the molecular signature of the habenula we characterized the genes that have high expression in the habenula. We compared anatomical expression profiles of three normal adult human brains and four fetal brains. We used gene set enrichment analyses to determine if genes annotated to specific molecular functions, cellular components, and biological processes are enriched in the habenula. We also tested gene sets related to depression and addiction to determine if they uniquely involve the habenula. As expected, we observed high habenular expression of GPR151, nicotinic cholinergic receptors, and cilia-associated genes (medial division). Genes identified in genetic studies of smoking and associated with nicotine response were enriched in the habenula. Genes associated with major depressive disorder did not have enriched expression in the habenula but genes negatively correlated with hedonic well-being were, providing a link to anhedonia. We observed enrichment of genes associated with diseases that are comorbid with addictions (hematopoiesis, thrombosis, liver cirrhosis, pneumonia, and pulmonary fibrosis) and depression (rheumatoid arthritis, multiple sclerosis, and kidney disease). These inflammatory diseases mark a neuroimmune signature that is supported by genes associated with mast cells, acute inflammatory response, and leukocyte migration. We also found enrichment of cytochrome p450 genes suggesting the habenula is uniquely sensitive to endogenous and xenobiotic compounds. Our results suggest the habenula receives negative reward signals from immune and drug processing molecules. This is consistent with the habenular role in the "anti-reward" system and suggests it may be a key bridge between autoimmune disorders, drug use, and psychiatric diseases.
Collapse
Affiliation(s)
- Bernard Le Foll
- Addictions Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Family & Community Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Leon French
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
32
|
Schubert N, Lisenko K, Auerbach C, Weitzmann A, Ghouse SM, Muhandes L, Haase C, Häring T, Schulze L, Voehringer D, Gunzer F, Müller W, Feyerabend TB, Rodewald HR, Dudeck A, Roers A. Unimpaired Responses to Vaccination With Protein Antigen Plus Adjuvant in Mice With Kit-Independent Mast Cell Deficiency. Front Immunol 2018; 9:1870. [PMID: 30210490 PMCID: PMC6123530 DOI: 10.3389/fimmu.2018.01870] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022] Open
Abstract
Innate inflammatory responses are crucial for induction and regulation of T cell and antibody responses. Mast cell (MC)-deficient Kit mutant mice showed impaired adaptive immunity, suggesting that MCs provide essential adjuvant activities, and pharmacological MC activation was proposed as a new adjuvant principle. However, the Kit mutations result in complex alterations of the immune system in addition to MC deficiency. We revisited the role of MCs in vaccination responses using Mcpt5-Cre R26DTA/DTA and Cpa3Cre/+ mice that lack connective tissue MCs or all MCs, respectively, but feature an otherwise normal immune system. These animals showed no impairment of T and B cell responses to intradermal vaccination with protein antigen plus complete Freund’s adjuvant. Moreover, we demonstrate that the adjuvant effects of the MC secretagogue c48/80 in intradermal or mucosal immunization are independent of the presence of MCs. We hence find no evidence for a regulation by MCs of adaptive immune responses to protein antigens. The finding that immunological MC functions differ from those suggested by experiments in Kit mutants, emphasizes the importance of rigorous tests in Kit-independent MC-deficiency models.
Collapse
Affiliation(s)
- Nadja Schubert
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Katharina Lisenko
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Christian Auerbach
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, University of Technology Dresden, Dresden, Germany
| | - Anke Weitzmann
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Shanawaz Mohammed Ghouse
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Lina Muhandes
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Christa Haase
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Tobias Häring
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Livia Schulze
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Florian Gunzer
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, University of Technology Dresden, Dresden, Germany
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Anne Dudeck
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany.,Medical Faculty, Institute for Molecular and Clinical Immunology, Otto von Guericke University, Magdeburg, Germany
| | - Axel Roers
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| |
Collapse
|
33
|
Tatemoto K, Nozaki Y, Tsuda R, Kaneko S, Tomura K, Furuno M, Ogasawara H, Edamura K, Takagi H, Iwamura H, Noguchi M, Naito T. Endogenous protein and enzyme fragments induce immunoglobulin E-independent activation of mast cells via a G protein-coupled receptor, MRGPRX2. Scand J Immunol 2018; 87:e12655. [PMID: 29484687 DOI: 10.1111/sji.12655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
Mast cells play a central role in inflammatory and allergic reactions by releasing inflammatory mediators through 2 main pathways, immunoglobulin E-dependent and E-independent activation. In the latter pathway, mast cells are activated by a diverse range of basic molecules (collectively known as basic secretagogues) through Mas-related G protein-coupled receptors (MRGPRs). In addition to the known basic secretagogues, here, we discovered several endogenous protein and enzyme fragments (such as chaperonin-10 fragment) that act as bioactive peptides and induce immunoglobulin E-independent mast cell activation via MRGPRX2 (previously known as MrgX2), leading to the degranulation of mast cells. We discuss the possibility that MRGPRX2 responds various as-yet-unidentified endogenous ligands that have specific characteristics, and propose that MRGPRX2 plays an important role in regulating inflammatory responses to endogenous harmful stimuli, such as protein breakdown products released from damaged or dying cells.
Collapse
Affiliation(s)
- K Tatemoto
- Department of Molecular Physiology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Y Nozaki
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - R Tsuda
- Department of Molecular Physiology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - S Kaneko
- Department of Molecular Physiology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - K Tomura
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - M Furuno
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - H Ogasawara
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - K Edamura
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - H Takagi
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - H Iwamura
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - M Noguchi
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| | - T Naito
- Pharmaceutical Frontier Research Laboratories, Japan Tobacco Inc., Yokohama, Japan
| |
Collapse
|
34
|
Russi AE, Walker-Caulfield ME, Brown MA. Mast cell inflammasome activity in the meninges regulates EAE disease severity. Clin Immunol 2018; 189:14-22. [DOI: 10.1016/j.clim.2016.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/16/2022]
|
35
|
Brown MA, Weinberg RB. Mast Cells and Innate Lymphoid Cells: Underappreciated Players in CNS Autoimmune Demyelinating Disease. Front Immunol 2018; 9:514. [PMID: 29619025 PMCID: PMC5871669 DOI: 10.3389/fimmu.2018.00514] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis, are autoimmune CNS inflammatory diseases. As a result of a breakdown in the relatively impermeable blood–brain barrier (BBB) in affected individuals, myelin-specific CD4+ and CD8+ T cells gain entry into the immune privileged CNS and initiate myelin, oligodendrocyte, and nerve axon destruction. However, despite the absolute requirement for T cells, there is increasing evidence that innate immune cells also play critical amplifying roles in disease pathogenesis. By modulating the character and magnitude of the myelin-reactive T cell response and regulating BBB integrity, innate cells affect both disease initiation and progression. Two classes of innate cells, mast cells and innate lymphoid cells (ILCs), have been best studied in models of allergic and gastrointestinal inflammatory diseases. Yet, there is emerging evidence that these cell types also exert a profound influence in CNS inflammatory disease. Both cell types are residents within the meninges and can be activated early in disease to express a wide variety of disease-modifying cytokines and chemokines. In this review, we discuss how mast cells and ILCs can have either disease-promoting or -protecting effects on MS and other CNS inflammatory diseases and how sex hormones may influence this outcome. These observations suggest that targeting these cells and their unique mediators can be exploited therapeutically.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Rebecca B Weinberg
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
36
|
Brown MA. Studies of Mast Cells: Adventures in Serendipity. Front Immunol 2018; 9:520. [PMID: 29593744 PMCID: PMC5859373 DOI: 10.3389/fimmu.2018.00520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/28/2018] [Indexed: 01/14/2023] Open
Abstract
Like many of us who had the great fortune to work with Bill Paul, my science life was immeasurably altered by my interactions with him. Although intimidating at first because of his stature in the immunology world, it was soon clear that he not only truly cared about the specific research we were doing together, but he wished to convey to his trainees an approach to science that was open, always questioning, and infinitely fun. His enthusiasm was infectious and after my training with him, despite stresses due to funding and publishing hurdles, I never regretted the path I took. My research took a sharp turn from the studies of adaptive immunity I had planned on pursuing after my fellowship with Bill to a life long quest to understand the wonders of the mast cell, a relatively rare innate immune cell. This came about because Bill’s curiosity and expectation of the unexpected allowed him to view, in retrospect, a rather mundane observation we made together involving a non-physiological transformed mast cell line as something that might be really interesting. I have never forgotten that lesson: Look at the data with an eye on the big picture. Sometimes the unexpected is more interesting than predicted results. His example in this regard was incredibly important when as an independent investigator a mistake in mouse sex determination led to unexpected and very confusing data. Yet, these data ultimately revealed a role for mast cells in male-specific protection in experimental autoimmune encephalomyelitis, the mouse model of multiple sclerosis. Bill’s influence in immunology is far-reaching and will continue to be felt as those of us who train our own students and post-doctoral fellows pass on his wisdom and approach to scientific research.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
37
|
Barclay W, Shinohara ML. Inflammasome activation in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Brain Pathol 2018; 27:213-219. [PMID: 27997058 DOI: 10.1111/bpa.12477] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/14/2016] [Indexed: 12/19/2022] Open
Abstract
The aptly named inflammasomes are powerful signaling complexes that sense inflammatory signals under a myriad of conditions, including those from infections and endogenous sources. The inflammasomes promote inflammation by maturation and release of the pro-inflammatory cytokines, IL-1β and IL-18. Several inflammasomes have been identified so far, but this review focuses mainly on the NLRP3 inflammasome. By still ill-defined activation mechanisms, a sensor molecule, NLRP3 (NACHT, LRR and PYD domains-containing protein 3), responds to danger signals and rapidly recruits ASC (apoptosis-associated speck-like protein containing a CARD) and pro-caspase-1 to form a large oligomeric signaling platform-the inflammasome. Involvement of the NLRP3 inflammasome in infections, metabolic disorders, autoinflammation, and autoimmunity, underscores its position as a central player in sensing microbial and damage signals and coordinating pro-inflammatory immune responses. Indeed, evidence in patients with multiple sclerosis (MS) suggests inflammasome activation occurs during disease. Experiments with the mouse model of MS, experimental autoimmune encephalomyelitis (EAE), specifically describe the NLRP3 inflammasome as critical and necessary to disease development. This review discusses recent studies in EAE and MS which describe associations of inflammasome activation with promotion of T cell pathogenicity, infiltration of cells into the central nervous system (CNS) and direct neurodegeneration during EAE and MS.
Collapse
Affiliation(s)
- William Barclay
- Department of Immunology, Duke University Medical School, Durham, NC
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical School, Durham, NC.,Department of Molecular Genetics and Microbiology, Duke University Medical School, Durham, NC
| |
Collapse
|
38
|
Fang H, Zhang Y, Li N, Wang G, Liu Z. The Autoimmune Skin Disease Bullous Pemphigoid: The Role of Mast Cells in Autoantibody-Induced Tissue Injury. Front Immunol 2018; 9:407. [PMID: 29545809 PMCID: PMC5837973 DOI: 10.3389/fimmu.2018.00407] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/14/2018] [Indexed: 01/09/2023] Open
Abstract
Bullous pemphigoid (BP) is an autoimmune and inflammatory skin disease associated with subepidermal blistering and autoantibodies directed against the hemidesmosomal components BP180 and BP230. Animal models of BP were developed by passively transferring anti-BP180 IgG into mice, which recapitulates the key features of human BP. By using these in vivo model systems, key cellular and molecular events leading to the BP disease phenotype are identified, including binding of pathogenic IgG to its target, complement activation of the classical pathway, mast cell degranulation, and infiltration and activation of neutrophils. Proteinases released by infiltrating neutrophils cleave BP180 and other hemidesmosome-associated proteins, causing DEJ separation. Mast cells and mast cell-derived mediators including inflammatory cytokines and proteases are increased in lesional skin and blister fluids of BP. BP animal model evidence also implicates mast cells in the pathogenesis of BP. However, recent studies questioned the pathogenic role of mast cells in autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, and epidermolysis bullosa acquisita. This review highlights the current knowledge on BP pathophysiology with a focus on a potential role for mast cells in BP and mast cell-related critical issues needing to be addressed in the future.
Collapse
Affiliation(s)
- Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Zhang
- Department of Dermatology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Dermatology, The Second Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Ning Li
- Department of Dermatology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhi Liu
- Department of Dermatology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
39
|
Diverse exocytic pathways for mast cell mediators. Biochem Soc Trans 2018; 46:235-247. [PMID: 29472369 DOI: 10.1042/bst20170450] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/23/2017] [Accepted: 01/04/2018] [Indexed: 12/14/2022]
Abstract
Mast cells play pivotal roles in innate and adaptive immunities but are also culprits in allergy, autoimmunity, and cardiovascular diseases. Mast cells respond to environmental changes by initiating regulated exocytosis/secretion of various biologically active compounds called mediators (e.g. proteases, amines, and cytokines). Many of these mediators are stored in granules/lysosomes and rely on intricate degranulation processes for release. Mast cell stabilizers (e.g. sodium cromoglicate), which prevent such degranulation processes, have therefore been clinically employed to treat asthma and allergic rhinitis. However, it has become increasingly clear that different mast cell diseases often involve multiple mediators that rely on overlapping but distinct mechanisms for release. This review illustrates existing evidence that highlights the diverse exocytic pathways in mast cells. We also discuss strategies to delineate these pathways so as to identify unique molecular components which could serve as new drug targets for more effective and specific treatments against mast cell-related diseases.
Collapse
|
40
|
Elieh-Ali-Komi D, Cao Y. Role of Mast Cells in the Pathogenesis of Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Clin Rev Allergy Immunol 2018; 52:436-445. [PMID: 28025778 DOI: 10.1007/s12016-016-8595-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune disorder of the central nervous system (CNS), characterized by recurrent episodes of inflammatory demyelination and consequent axonal deterioration. The hallmark of the disease is the demyelinated plaque, a hypocellular area characterized by formation of astrocytic scars and infiltration of mononuclear cells. Recent studies have revealed that both innate and adaptive immune cells contribute to the pathogenesis of MS and its experimental autoimmune encephalomyelitis (EAE) model. Here, we review the current understanding of the role of mast cells in the pathogenesis of MS and EAE. Mast cells may act at the early stage that promote demyelination through interactions among mast cells, neurons, and other immune cells to mediate neuroinflammation. Studies from EAE model suggest that mast cells regulate adaptive autoimmune responses, present myelin antigens to T cells, disrupt the blood-brain barrier, and permit the entry of inflammatory cells and mediators into the CNS. Depletion or limiting mast cells could be a new promising therapeutic target for MS and EAE.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Immunology Research Center, Department of Immunology, and Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yonghao Cao
- Center for Synthetic Biology Engineering Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China. .,Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
41
|
Podbielska M, O'Keeffe J, Hogan EL. Autoimmunity in multiple sclerosis: role of sphingolipids, invariant NKT cells and other immune elements in control of inflammation and neurodegeneration. J Neurol Sci 2017; 385:198-214. [PMID: 29406905 DOI: 10.1016/j.jns.2017.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system. It is classified as being an autoimmune response in the genetically susceptible individual to a persistent but unidentified antigen(s). Both the adaptive and the innate immune systems are likely to contribute significantly to MS pathogenesis. This review summarizes current understanding of the characteristics of MS autoimmunity in the initiation and progression of the disease. In particular we find it timely to classify the autoimmune responses by focusing on the immunogenic features of myelin-derived lipids in MS including molecular mimicry; on alterations of bioactive sphingolipids mediators in MS; and on functional roles for regulatory effector cells, including innate lymphocyte populations, like the invariant NKT (iNKT) cells which bridge adaptive and innate immune systems. Recent progress in identifying the nature of sphingolipids recognition for iNKT cells in immunity and the functional consequences of the lipid-CD1d interaction opens new avenues of access to the pathogenesis of demyelination in MS as well as design of lipid antigen-specific therapeutics.
Collapse
Affiliation(s)
- Maria Podbielska
- Department of Neurology and Neurosurgery, Medical University of South Carolina Charleston, SC, USA; Laboratory of Signal Transduction Molecules, Ludwik Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland.
| | - Joan O'Keeffe
- Department of Biopharmaceutical & Medical Science, School of Science & Computing, Galway-Mayo Institute of Technology, Galway, Ireland
| | - Edward L Hogan
- Department of Neurology and Neurosurgery, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|
42
|
Gao YY, Liu QM, Liu B, Xie CL, Cao MJ, Yang XW, Liu GM. Inhibitory Activities of Compounds from the Marine Actinomycete Williamsia sp. MCCC 1A11233 Variant on IgE-Mediated Mast Cells and Passive Cutaneous Anaphylaxis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:10749-10756. [PMID: 29148756 DOI: 10.1021/acs.jafc.7b04314] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The compounds of the deep-sea-derived marine Williamsia sp. MCCC 1A11233 (CDMW) were isolated, which are secondary metabolites of the actinomycetes. In this study, seven kinds of CDMW were found to decrease degranulation and histamine release in immunoglobulin E (IgE)-mediated rat basophilic leukemia (RBL)-2H3 cells. The production of cytokines (tumor necrosis factor-α, interleukin-4) was inhibited by these CDMW in RBL-2H3 cells, and their chemical structures were established mainly based on detailed analysis of their NMR spectra. CDMW-3, CDMW-5, and CDMW-15 were further demonstrated to block mast cell-dependent passive cutaneous anaphylaxis in IgE-sensitized mice. Bone marrow mononuclear cells (BMMCs) were established to clarify the effect of CDMW-3, CDMW-5, and CDMW-15 on mast cells. The seven kinds of CDMW decreased the degranulation and histamine release of BMMCs. Furthermore, flow cytometry results indicated that CDMW-3, CDMW-5, and CDMW-15 increased the annexin+ cell population of BMMCs. In conclusion, CDMW-3, CDMW-5, and CDMW-15 have obvious antiallergic activity due to induction of the apoptosis of mast cells.
Collapse
Affiliation(s)
- Yuan-Yuan Gao
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University , 43 Yindou Road, Xiamen 361021, Fujian, P. R. China
| | - Qing-Mei Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University , 43 Yindou Road, Xiamen 361021, Fujian, P. R. China
| | - Bo Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University , 43 Yindou Road, Xiamen 361021, Fujian, P. R. China
| | - Chun-Lan Xie
- Key Laboratory of Marine Biogenetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration , 184 Daxue Road, Xiamen 361005, P. R. China
| | - Min-Jie Cao
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University , 43 Yindou Road, Xiamen 361021, Fujian, P. R. China
| | - Xian-Wen Yang
- Key Laboratory of Marine Biogenetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration , 184 Daxue Road, Xiamen 361005, P. R. China
| | - Guang-Ming Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University , 43 Yindou Road, Xiamen 361021, Fujian, P. R. China
| |
Collapse
|
43
|
Immunoregulatory effect of mast cells influenced by microbes in neurodegenerative diseases. Brain Behav Immun 2017; 65:68-89. [PMID: 28676349 DOI: 10.1016/j.bbi.2017.06.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/17/2017] [Accepted: 06/30/2017] [Indexed: 02/06/2023] Open
Abstract
When related to central nervous system (CNS) health and disease, brain mast cells (MCs) can be a source of either beneficial or deleterious signals acting on neural cells. We review the current state of knowledge about molecular interactions between MCs and glia in neurodegenerative diseases such as Multiple Sclerosis, Alzheimer's disease, Amyotrophic Lateral Sclerosis, Parkinson's disease, Epilepsy. We also discuss the influence on MC actions evoked by the host microbiota, which has a profound effect on the host immune system, inducing important consequences in neurodegenerative disorders. Gut dysbiosis, reduced intestinal motility and increased intestinal permeability, that allow bacterial products to circulate and pass through the blood-brain barrier, are associated with neurodegenerative disease. There are differences between the microbiota of neurologic patients and healthy controls. Distinguishing between cause and effect is a challenging task, and the molecular mechanisms whereby remote gut microbiota can alter the brain have not been fully elucidated. Nevertheless, modulation of the microbiota and MC activation have been shown to promote neuroprotection. We review this new information contributing to a greater understanding of MC-microbiota-neural cells interactions modulating the brain, behavior and neurodegenerative processes.
Collapse
|
44
|
Lin CC, Edelson BT. New Insights into the Role of IL-1β in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2017; 198:4553-4560. [PMID: 28583987 DOI: 10.4049/jimmunol.1700263] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/23/2017] [Indexed: 01/05/2023]
Abstract
Multiple sclerosis (MS), and its animal model experimental autoimmune encephalomyelitis, are neuroinflammatory diseases driven by autoreactive pathogenic TH cells that elicit demyelination and axonal damage. How TH cells acquire pathogenicity and communicate with myeloid cells and cells of the CNS remain unclear. IL-1β is recognized to play an important role in experimental autoimmune encephalomyelitis (EAE) and perhaps MS. Clinical EAE is significantly attenuated in IL-1R-deficient and IL-1β-deficient mice, and IL-1β is found in the blood, cerebrospinal fluid, and CNS lesions of MS patients. In this article, we focus on new reports that elucidate the cellular sources of IL-1β and its actions during EAE, in both lymphoid tissues and within the CNS. Several immune cell types serve as critical producers of IL-1β during EAE, with this cytokine inducing response in both hematopoietic and nonhematopoietic cells. These findings from the EAE model should inspire efforts toward investigating the therapeutic potential of IL-1 blockade in MS.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
45
|
Kempuraj D, Selvakumar GP, Zaheer S, Thangavel R, Ahmed ME, Raikwar S, Govindarajan R, Iyer S, Zaheer A. Cross-Talk between Glia, Neurons and Mast Cells in Neuroinflammation Associated with Parkinson's Disease. J Neuroimmune Pharmacol 2017; 13:100-112. [PMID: 28952015 DOI: 10.1007/s11481-017-9766-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023]
Abstract
Parkinson's disease (PD) is a progressive movement disorder characterized by neuroinflammation and dopaminergic neurodegeneration in the brain. 1-methyl-4-phenylpyridinium (MPP+), a metabolite of the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces the release of inflammatory mediators from glial cells and neurons. Glia maturation factor (GMF), a brain proinflammatory protein, MPP+, and mast cell-derived inflammatory mediators induce neurodegeneration which eventually leads to PD. However, the precise mechanisms underlying interaction between glial cells, neurons and mast cells in PD still remain elusive. In the present study, mouse bone marrow-derived mast cells (BMMCs) and mouse fetal brain-derived mixed glia/neurons, astrocytes and neurons were incubated with MPP+, GMF and mast cell-derived inflammatory mediators mouse mast cell protease-6 (MMCP-6), MMCP-7 or tryptase/brain-specific serine protease-4 (tryptase/BSSP-4). Inflammatory mediators released from these cells in the culture medium were quantitated by enzyme-linked immunosorbent assay. Neurodegeneration was quantified by measuring total neurite outgrowth following microtubule-associated protein-2 immunocytochemistry. MPP+-induced significant neurodegeneration with reduced total neurite outgrowth. MPP+induced the release of tryptase/BSSP-4 from the mouse mast cells, and tryptase/BSSP-4 induced chemokine (C-C motif) ligand 2 (CCL2) release from astrocytes and glia/neurons. Overall our results suggest that MPP+, GMF, MMCP-6 or MMCP-7 stimulate glia/neurons, astrocytes or neurons to release CCL2 and matrix metalloproteinase-3. Additionally, CD40L expression is increased in BMMCs after incubation with MPP+ in a co-culture system consisting of BMMCs and glia/neurons. We propose that mast cell interaction with glial cells and neurons during neuroinflammation can be explored as a new therapeutic target for PD.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Govindhasamy Pushpavathi Selvakumar
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Ramasamy Thangavel
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Mohammad Ejaz Ahmed
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Sudhanshu Raikwar
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Raghav Govindarajan
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Shankar Iyer
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA
| | - Asgar Zaheer
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, 65201, USA. .,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, 65201, USA.
| |
Collapse
|
46
|
McLeod JJA, Caslin HL, Spence AJ, Kolawole EM, Qayum AA, Paranjape A, Taruselli M, Haque TT, Kiwanuka KN, Elford HL, Ryan JJ. Didox (3,4-dihydroxybenzohydroxamic acid) suppresses IgE-mediated mast cell activation through attenuation of NFκB and AP-1 transcription. Cell Immunol 2017; 322:41-48. [PMID: 28964543 DOI: 10.1016/j.cellimm.2017.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/06/2017] [Accepted: 09/21/2017] [Indexed: 01/11/2023]
Abstract
Mast cell activation via the high-affinity IgE receptor (FcεRI) elicits production of inflammatory mediators central to allergic disease. As a synthetic antioxidant and a potent ribonucleotide reductase (RNR) inhibitor, Didox (3,4-dihyroxybenzohydroxamic acid) has been tested in clinical trials for cancer and is an attractive therapeutic for inflammatory disease. We found that Didox treatment of mouse bone marrow-derived mast cells (BMMC) reduced IgE-stimulated degranulation and cytokine production, including IL-6, IL-13, TNF and MIP-1a (CCL3). These effects were consistent using BMMC of different genetic backgrounds and peritoneal mast cells. While the RNR inhibitor hydroxyurea had little or no effect on IgE-mediated function, high concentrations of the antioxidant N-acetylcysteine mimicked Didox-mediated suppression. Furthermore, Didox increased expression of the antioxidant genes superoxide dismutase and catalase, and suppressed DCFH-DA fluorescence, indicating reduced reactive oxygen species production. Didox effects were not due to changes in FcεRI expression or cell viability, suggesting it inhibits signaling required for inflammatory cytokine production. In support of this, we found that Didox reduced FcεRI-mediated AP-1 and NFκB transcriptional activity. Finally, Didox suppressed mast cell-dependent, IgE-mediated passive systemic anaphylaxis in vivo. These data demonstrate the potential use for Didox asa means of antagonizing mast cell responses in allergic disease.
Collapse
Affiliation(s)
| | - Heather L Caslin
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Andrew J Spence
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Elizabeth M Kolawole
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Amina Abdul Qayum
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Anuya Paranjape
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Marcela Taruselli
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Tamara T Haque
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Kasalina N Kiwanuka
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Howard L Elford
- Molecules for Health, Inc, Richmond, VA 23219, United States
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA, 23284, United States.
| |
Collapse
|
47
|
Shrestha B, Jiang X, Ge S, Paul D, Chianchiano P, Pachter JS. Spatiotemporal resolution of spinal meningeal and parenchymal inflammation during experimental autoimmune encephalomyelitis. Neurobiol Dis 2017; 108:159-172. [PMID: 28844788 DOI: 10.1016/j.nbd.2017.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/10/2017] [Accepted: 08/18/2017] [Indexed: 01/14/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) induced by active immunization of C57BL/6 mice with peptide from myelin oligodendrocyte protein (MOG35-55), is a neuroinflammatory, demyelinating disease widely recognized as an animal model of multiple sclerosis (MS). Typically, EAE presents with an ascending course of paralysis, and inflammation that is predominantly localized to the spinal cord. Recent studies have further indicated that inflammation - in both MS and EAE - might initiate within the meninges and propagate from there to the underlying parenchyma. However, the patterns of inflammation within the respective meningeal and parenchymal compartments along the length of the spinal cord, and the progression with which these patterns develop during EAE, have yet to be detailed. Such analysis could hold key to identifying factors critical for spreading, as well as constraining, inflammation along the neuraxis. To address this issue, high-resolution 3-dimensional (3D) confocal microscopy was performed to visualize, in detail, the sequence of leukocyte infiltration at distinct regions of the spinal cord. High quality virtual slide scanning for imaging the entire spinal cord using epifluorescence was further conducted to highlight the directionality and relative degree of inflammation. Meningeal inflammation was found to precede parenchymal inflammation at all levels of the spinal cord, but did not develop equally or simultaneously throughout the subarachnoid space (SAS) of the meninges. Instead, meningeal inflammation was initially most obvious in the caudal SAS, from which it progressed to the immediate underlying parenchyma, paralleling the first signs of clinical disease in the tail and hind limbs. Meningeal inflammation could then be seen to extend in the caudal-to-rostral direction, followed by a similar, but delayed, trajectory of parenchymal inflammation. To additionally determine whether the course of ascending paralysis and leukocyte infiltration during EAE is reflected in differences in inflammatory gene expression by meningeal and parenchymal microvessels along the spinal cord, laser capture microdissection (LCM) coupled with gene expression profiling was performed. Expression profiles varied between these respective vessel populations at both the cervical and caudal levels of the spinal cord during disease progression, and within each vessel population at different levels of the cord at a given time during disease. These results reinforce a significant role for the meninges in the development and propagation of central nervous system inflammation associated with MS and EAE.
Collapse
Affiliation(s)
- Bandana Shrestha
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Xi Jiang
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Shujun Ge
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Debayon Paul
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Peter Chianchiano
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Joel S Pachter
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| |
Collapse
|
48
|
Hendriksen E, van Bergeijk D, Oosting RS, Redegeld FA. Mast cells in neuroinflammation and brain disorders. Neurosci Biobehav Rev 2017; 79:119-133. [DOI: 10.1016/j.neubiorev.2017.05.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 12/13/2022]
|
49
|
Kempuraj D, Thangavel R, Selvakumar GP, Zaheer S, Ahmed ME, Raikwar SP, Zahoor H, Saeed D, Natteru PA, Iyer S, Zaheer A. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2017; 11:216. [PMID: 28790893 PMCID: PMC5522882 DOI: 10.3389/fncel.2017.00216] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/05/2017] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammatory response is primarily a protective mechanism in the brain. However, excessive and chronic inflammatory responses can lead to deleterious effects involving immune cells, brain cells and signaling molecules. Neuroinflammation induces and accelerates pathogenesis of Parkinson’s disease (PD), Alzheimer’s disease (AD) and Multiple sclerosis (MS). Neuroinflammatory pathways are indicated as novel therapeutic targets for these diseases. Mast cells are immune cells of hematopoietic origin that regulate inflammation and upon activation release many proinflammatory mediators in systemic and central nervous system (CNS) inflammatory conditions. In addition, inflammatory mediators released from activated glial cells induce neurodegeneration in the brain. Systemic inflammation-derived proinflammatory cytokines/chemokines and other factors cause a breach in the blood brain-barrier (BBB) thereby allowing for the entry of immune/inflammatory cells including mast cell progenitors, mast cells and proinflammatory cytokines and chemokines into the brain. These peripheral-derived factors and intrinsically generated cytokines/chemokines, α-synuclein, corticotropin-releasing hormone (CRH), substance P (SP), beta amyloid 1–42 (Aβ1–42) peptide and amyloid precursor proteins can activate glial cells, T-cells and mast cells in the brain can induce additional release of inflammatory and neurotoxic molecules contributing to chronic neuroinflammation and neuronal death. The glia maturation factor (GMF), a proinflammatory protein discovered in our laboratory released from glia, activates mast cells to release inflammatory cytokines and chemokines. Chronic increase in the proinflammatory mediators induces neurotoxic Aβ and plaque formation in AD brains and neurodegeneration in PD brains. Glial cells, mast cells and T-cells can reactivate each other in neuroinflammatory conditions in the brain and augment neuroinflammation. Further, inflammatory mediators from the brain can also enter into the peripheral system through defective BBB, recruit immune cells into the brain, and exacerbate neuroinflammation. We suggest that mast cell-associated inflammatory mediators from systemic inflammation and brain could augment neuroinflammation and neurodegeneration in the brain. This review article addresses the role of some atypical inflammatory mediators that are associated with mast cell inflammation and their activation of glial cells to induce neurodegeneration.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Govindhasamy P Selvakumar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Mohammad E Ahmed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Prashant A Natteru
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Shankar Iyer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Asgar Zaheer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| |
Collapse
|
50
|
Musio S, Costanza M, Poliani PL, Fontana E, Cominelli M, Abolafio G, Steinman L, Pedotti R. Treatment with anti-FcεRIα antibody exacerbates EAE and T-cell immunity against myelin. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 4:e342. [PMID: 28616446 PMCID: PMC5462602 DOI: 10.1212/nxi.0000000000000342] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/09/2017] [Indexed: 12/25/2022]
Abstract
Objective: To investigate the effects of targeting the high-affinity receptor for immunoglobulin E (FcεRI), that plays a central role in allergic responses and is constitutively expressed on mast cells and basophils, in clinical disease and autoimmune T-cell response in experimental MS. Methods: Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein 35–55. Anti-FcεRI α-chain antibody was administered intraperitoneally. CNS immunohistochemistry, flow cytometry analysis of immune cell populations, IgE and histamine serum concentration, immune cell proliferation, and cytokine measurement were performed. In BALB/c mice, EAE was induced by immunization with myelin proteolipid protein 185–206. Results: Treatment with anti-FcεRIα antibody resulted in exacerbation of EAE and increased CNS inflammation in C57BL/6 mice. Treated mice displayed long-lasting complete depletion of basophils in the blood stream and peripheral lymphoid organs and increased antigen-induced immune cell proliferation and production of interferon-γ, interleukin (IL)-17, IL-6, and granulocyte-macrophage colony-stimulating factor. In BALB/c mice, which are T-helper (Th) 2 prone and resistant to EAE, treatment with anti-FcεRIα antibody restored susceptibility to EAE. Conclusion: Our observations that anti-FcεRIα antibody increases Th1 and Th17 responses against myelin antigen and exacerbates EAE suggest that FcεRI, basophils, and possibly other FcεRI-bearing cells that might be affected by this antibody play important roles in influencing the severity of CNS autoimmunity.
Collapse
Affiliation(s)
- Silvia Musio
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Massimo Costanza
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Pietro Luigi Poliani
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Elena Fontana
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Manuela Cominelli
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Gabriella Abolafio
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Lawrence Steinman
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| | - Rosetta Pedotti
- Department of Clinical Neuroscience (S.M., M. Costanza, R.P.), Foundation Neurological Institute IRCCS C. Besta, Milan; Department of Molecular and Translational Medicine (P.L.P., E.F., M. Cominelli), Pathology Unit, University of Brescia; Department of Experimental Oncology and Molecular Medicine (G.A.), Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy; and Department of Neurology and Neurological Sciences (L.S.), Stanford University School of Medicine, CA
| |
Collapse
|