1
|
Gladen-Kolarsky N, Neff CJ, Hack W, Brandes MS, Wiedrick J, Meza-Romero R, Lockwood DR, Quinn JF, Offner H, Vandenbark AA, Gray NE. The CD74 inhibitor DRhQ improves short-term memory and mitochondrial function in 5xFAD mouse model of Aβ accumulation. Metab Brain Dis 2025; 40:95. [PMID: 39808341 DOI: 10.1007/s11011-024-01433-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025]
Abstract
Neuroinflammation and mitochondrial dysfunction are early events in Alzheimer's disease (AD) and contribute to neurodegeneration and cognitive impairment. Evidence suggests that the inflammatory axis mediated by macrophage migration inhibitory factor (MIF) binding to its receptor, CD74, plays an important role in many central nervous system (CNS) disorders such as AD. Our group has developed DRhQ, a novel CD74 binding construct which competitively inhibits MIF binding, blocks macrophage activation and migration into the CNS, enhances anti-inflammatory microglia cell numbers and reduces pro-inflammatory gene expression. Here, we evaluate its effects in amyloid-β (Aβ) overexpressing mice. 5xFAD mice and their wild type littermates were treated with DRhQ (100 µg) or vehicle for 4 weeks. DRhQ improved cognition and cortical mitochondrial function in both male and female 5xFAD mice. Aβ plaque burden in 5xFAD animals was not robustly impacted by DRhQ treatment in either the hippocampus or the cortex. Cortical microglial activation was similarly not apparently affected by DRhQ treatment, although in the hippocampus there was evidence of a reduction in activated microglia for female 5xFAD mice. Future studies are needed to confirm this possible sex-dependent response on microglial activation, as well as to optimize the dose and timing of DRhQ treatment and gain a better understanding of its mechanism of action in AD.
Collapse
Affiliation(s)
- Noah Gladen-Kolarsky
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Cody J Neff
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Wyatt Hack
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Mikah S Brandes
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Jack Wiedrick
- Biostatistics & Design Program, OHSU-PSU School of Public Health, Portland, OR, 97201, USA
| | - Roberto Meza-Romero
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Neuroimmunology Research, VA Portland Healthcare System, Portland, OR, 97239, USA
| | - Denesa R Lockwood
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Neuroimmunology Research, VA Portland Healthcare System, Portland, OR, 97239, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR, 97239, USA
| | - Halina Offner
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Neuroimmunology Research, VA Portland Healthcare System, Portland, OR, 97239, USA
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Neuroimmunology Research, VA Portland Healthcare System, Portland, OR, 97239, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA.
| |
Collapse
|
2
|
Zerimech S, Nguyen H, Vandenbark AA, Offner H, Baltan S. Novel therapeutic for multiple sclerosis protects white matter function in EAE mouse model. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1237078. [PMID: 37933270 PMCID: PMC10627517 DOI: 10.3389/fmmed.2023.1237078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease with prominent axon dysfunction. Our previous studies in an MS mouse model, experimental autoimmune encephalomyelitis (EAE), demonstrated that major histocompatibility complex Class II constructs can reverse clinical signs of EAE. These constructs block binding and downstream signaling of macrophage migration inhibitory factors (MIF-1/2) through CD74, thereby inhibiting phosphorylation of extracellular signal-regulated kinase (ERK) activation and tissue inflammation and promoting remyelination. To directly assess the effects of a novel third generation construct, DRhQ, on axon integrity in EAE, we compared axon conduction properties using electrophysiology on corpus callosum slices and optic nerves. By using two distinct white matter (WM) tracts, we aimed to assess the impact of the EAE and the benefit of DRhQ on myelinated and unmyelinated axons as well as to test the clinical value of DRhQ on demyelinating lesions in CC and optic myelitis. Our study found that EAE altered axon excitability, delayed axon conduction and slowed spatiotemporal summation correlated with diffuse astrocyte and microglia activation. Because MS predisposes patients to stroke, we also investigated and showed that vulnerability to WM ischemia is increased in the EAE MS mouse model. Treatment with DRhQ after the onset of EAE drastically inhibited microglial and astrocyte activation, improved functional integrity of the myelinated axons and enhanced recovery after ischemia. These results demonstrate that DRhQ administered after the onset of EAE promotes WM integrity and function, and reduces subsequent vulnerability to ischemic injury, suggesting important therapeutic potential for treatment of progressive MS.
Collapse
Affiliation(s)
- Sarah Zerimech
- Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University, Portland, OR, United States
| | - Hung Nguyen
- Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University, Portland, OR, United States
| | - Arthur A. Vandenbark
- Neuroimmunology Research, VA Portland Healthcare System, Portland, OR, United States
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Halina Offner
- Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University, Portland, OR, United States
- Neuroimmunology Research, VA Portland Healthcare System, Portland, OR, United States
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Selva Baltan
- Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
3
|
Liu R, Song P, Gu X, Liang W, Sun W, Hua Q, Zhang Y, Qiu Z. Comprehensive Landscape of Immune Infiltration and Aberrant Pathway Activation in Ischemic Stroke. Front Immunol 2022; 12:766724. [PMID: 35140708 PMCID: PMC8818702 DOI: 10.3389/fimmu.2021.766724] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Ischemic stroke (IS) is a multifactorial disease caused by the interaction of multiple environmental and genetic risk factors, and it is the most common cause of disability. The immune microenvironment and inflammatory response participate in the whole process of IS occurrence and development. Therefore, the rational use of relevant markers or characteristic pathways in the immune microenvironment will become one of the important therapeutic strategies for the treatment of IS. We collected peripheral blood samples from 10 patients diagnosed with IS at the First Affiliated Hospital of Gannan Medical University and First Affiliated Hospital, Jinan" University, and from 10 normal people. The GSE16561 dataset was downloaded from the Gene Expression Omnibus (GEO) database. xCell, gene set enrichment analysis (GSEA), single-sample GSEA (ssGSEA) and immune-related gene analysis were used to evaluate the differences in the immune microenvironment and characteristic pathways between the IS and control groups of the two datasets. xCell analysis showed that the IS-24h group had significantly reduced central memory CD8+ T cell, effector memory CD8+ T cell, B cell and Th1 cell scores and significantly increased M1 macrophage and macrophage scores. GSEA showed that the IS-24h group had significantly increased inflammation-related pathway activity(myeloid leukocyte activation, positive regulation of tumor necrosis factor biosynthetic process, myeloid leukocyte migration and leukocyte chemotaxis), platelet-related pathway activity(platelet activation, signaling and aggregation; protein polymerization; platelet degranulation; cell-cell contact zone) and pathology-related pathway activity (ERBB signaling pathway, positive regulation of ERK1 and ERK2 cascade, vascular endothelial growth factor receptor signaling pathway, and regulation of MAP kinase activity). Immune-related signature analysis showed that the macrophage signature, antigen presentation-related signature, cytotoxicity-related signature, B cell-related signature and inflammation-related signature were significantly lower in the IS-24h group than in the control group. In this study, we found that there were significant differences in the immune microenvironment between the peripheral blood of IS patients and control patients, as shown by the IS group having significantly reduced CD8+ Tcm, CD8+ Tem, B cell and Th1 cell scores and significantly increased macrophage and M1 macrophage scores. Additionally, inflammation-related, pathological, and platelet-related pathway activities were significantly higher in the IS group than in the control group.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Neurology, Ganzhou People’s Hospital, Ganzhou, China
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Pingping Song
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Xunhu Gu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weidong Liang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wei Sun
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Qian Hua
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yusheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
- *Correspondence: Yusheng Zhang, ; Zhengang Qiu,
| | - Zhengang Qiu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Yusheng Zhang, ; Zhengang Qiu,
| |
Collapse
|
4
|
Var SR, Shetty AV, Grande AW, Low WC, Cheeran MC. Microglia and Macrophages in Neuroprotection, Neurogenesis, and Emerging Therapies for Stroke. Cells 2021; 10:3555. [PMID: 34944064 PMCID: PMC8700390 DOI: 10.3390/cells10123555] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022] Open
Abstract
Stroke remains the number one cause of morbidity in the United States. Within weeks to months after an ischemic event, there is a resolution of inflammation and evidence of neurogenesis; however, years following a stroke, there is evidence of chronic inflammation in the central nervous system, possibly by the persistence of an autoimmune response to brain antigens as a result of ischemia. The mechanisms underlying the involvement of macrophage and microglial activation after stroke are widely acknowledged as having a role in ischemic stroke pathology; thus, modulating inflammation and neurological recovery is a hopeful strategy for treating the long-term outcomes after ischemic injury. Current treatments fail to provide neuroprotective or neurorestorative benefits after stroke; therefore, to ameliorate brain injury-induced deficits, therapies must alter both the initial response to injury and the subsequent inflammatory process. This review will address differences in macrophage and microglia nomenclature and summarize recent work in elucidating the mechanisms of macrophage and microglial participation in antigen presentation, neuroprotection, angiogenesis, neurogenesis, synaptic remodeling, and immune modulating strategies for treating the long-term outcomes after ischemic injury.
Collapse
Affiliation(s)
- Susanna R. Var
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Anala V. Shetty
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
- Department of Biological Sciences, University of Minnesota Medical School, Minneapolis, MN 55108, USA
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Maxim C. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
5
|
Zhang D, Ren J, Luo Y, He Q, Zhao R, Chang J, Yang Y, Guo ZN. T Cell Response in Ischemic Stroke: From Mechanisms to Translational Insights. Front Immunol 2021; 12:707972. [PMID: 34335623 PMCID: PMC8320432 DOI: 10.3389/fimmu.2021.707972] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/01/2021] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke, caused by a sudden disruption of blood flow to the brain, is a leading cause of death and exerts a heavy burden on both patients and public health systems. Currently available treatments for ischemic stroke are very limited and are not feasible in many patients due to strict time windows required for their administration. Thus, novel treatment strategies are keenly required. T cells, which are part of the adaptive immune system, have gained more attention for its effects in ischemic stroke. Both preclinical and clinical studies have revealed the conflicting roles for T cells in post-stroke inflammation and as potential therapeutic targets. This review summarizes the mediators of T cell recruitment, as well as the temporal course of its infiltration through the blood-brain-barrier, choroid plexus, and meningeal pathways. Furthermore, we describe the mechanisms behind the deleterious and beneficial effects of T cells in the brain, in both antigen-dependent and antigen-independent manners, and finally we specifically focus on clinical and preclinical studies that have investigated T cells as potential therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Dianhui Zhang
- Stroke Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Jiaxin Ren
- Stroke Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Yun Luo
- Stroke Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China.,Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qianyan He
- Stroke Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Ruoyu Zhao
- Stroke Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Junlei Chang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yi Yang
- Stroke Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| | - Zhen-Ni Guo
- Neuroscience Center, Department of Neurology, First Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Cho JY, Matsukawa N. The unsolved mystery of hippocampal cholinergic neurostimulating peptide: A potent cholinergic regulator. Brain Circ 2021; 7:29-32. [PMID: 34084974 PMCID: PMC8057103 DOI: 10.4103/bc.bc_14_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 11/27/2022] Open
Abstract
Cholinergic efferent networks located from the medial septal nucleus to the hippocampus play a pivotal role in learning and memory outcomes by generating regular theta rhythms that enhance information retention. Hippocampal cholinergic neurostimulating peptide (HCNP), derived from the N-terminus of HCNP precursor protein (HCNP-pp), promotes the synthesis of acetylcholine in the medial septal nuclei. HCNP-pp deletion significantly reduced theta power in CA1 possibly due to lower levels of choline acetyltransferase-positive axons in CA1 stratum oriens, suggesting cholinergic disruptions in the septo-hippocampal system. This review also explores HCNP as a potent cholinergic regulator in the septo-hippocampal network while also examining the limitations of our understanding of the neurostimulating peptide.
Collapse
Affiliation(s)
- Justin Y Cho
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | | |
Collapse
|
7
|
Gonzales-Portillo BM, Lee JY, Vandenbark AA, Offner H, Borlongan CV. Major histocompatibility complex Class II-based therapy for stroke. Brain Circ 2021; 7:37-40. [PMID: 34084976 PMCID: PMC8057100 DOI: 10.4103/bc.bc_16_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 11/04/2022] Open
Abstract
This review discusses the potential of major histocompatibility complex (MHC) Class II constructs as stroke therapeutics. We focus on the delivery of MHC Class II construct, DRmQ, as a safe and effective treatment for ischemic stroke. DRmQ was observed to attenuate behavioral deficits and decrease microglia activation and proinflammatory cytokines, illustrating its ability to mitigate the secondary cell death following stroke. Similar anti-neuroinflammation treatments, such as transplantation of mesenchymal stem cells and mitochondrial transfers, are briefly discussed to provide further support that sequestration of inflammation stands as a robust therapeutic target for stroke.
Collapse
Affiliation(s)
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Arthur A. Vandenbark
- Department of Veterans Affairs, Veterans Affairs Portland Health Care System, Portland, OR, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular Microbiology and Immunology and Anaesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Department of Anaesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Cesario V. Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
8
|
Loftis JM, Navis T, Taylor J, Hudson R, Person U, Lattal KM, Vandenbark AA, Shirley R, Huckans M. Partial MHC/neuroantigen peptide constructs attenuate methamphetamine-seeking and brain chemokine (C-C motif) ligand 2 levels in rats. Eur J Pharmacol 2020; 880:173175. [PMID: 32416183 DOI: 10.1016/j.ejphar.2020.173175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/09/2020] [Accepted: 05/06/2020] [Indexed: 11/28/2022]
Abstract
There are no medications that target the neurotoxic effects or reduce the use of methamphetamine. Recombinant T-cell receptor ligand (RTL) 1000 [a partial major histocompatibility complex (pMHC) class II construct with a tethered myelin peptide], addresses the neuroimmune effects of methamphetamine addiction by competitively inhibiting the disease-promoting activity of macrophage migration inhibitory factor to CD74, a key pathway involved in several chronic inflammatory conditions, including substance use disorders. We previously reported that RTL constructs improve learning and memory impairments and central nervous system (CNS) inflammation induced by methamphetamine in mouse models. The present study in Lewis rats evaluated the effects of RTL1000 on maintenance of self-administration and cue-induced reinstatement using operant behavioral methods. Post-mortem brain and serum samples were evaluated for the levels of inflammatory factors. Rats treated with RTL1000 displayed significantly fewer presses on the active lever as compared to rats treated with vehicle during the initial extinction session, indicating more rapid extinction in the presence of RTL1000. Immunoblotting of rat brain sections revealed reduced levels of the pro-inflammatory chemokine (C-C motif) ligand 2 (CCL2) in the frontal cortex of rats treated with RTL1000, as compared to vehicle. Post hoc analysis identified a positive association between the levels of CCL2 detected in the frontal cortex and the number of lever presses during the first extinction session. Taken together, results suggest that RTL1000 may block downstream inflammatory effects of methamphetamine exposure and facilitate reduced drug seeking-potentially offering a new strategy for the treatment of methamphetamine-induced CNS injury and neuropsychiatric impairments.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA; Methamphetamine Research Center, Portland, OR, USA.
| | - Tommy Navis
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Jonathan Taylor
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Rebekah Hudson
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Ulziibat Person
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Arthur A Vandenbark
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Renee Shirley
- Virogenomics BioDevelopment, Inc., Portland, OR, USA
| | - Marilyn Huckans
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA; Methamphetamine Research Center, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
9
|
Lee JY, Castelli V, Bonsack B, Coats AB, Navarro-Torres L, Garcia-Sanchez J, Kingsbury C, Nguyen H, Vandenbark AA, Meza-Romero R, Offner H, Borlongan CV. A Novel Partial MHC Class II Construct, DRmQ, Inhibits Central and Peripheral Inflammatory Responses to Promote Neuroprotection in Experimental Stroke. Transl Stroke Res 2020; 11:831-836. [PMID: 31797249 PMCID: PMC10166182 DOI: 10.1007/s12975-019-00756-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 12/23/2022]
Abstract
Recognizing that the pathologic progression of stroke is closely associated with aberrant immune responses, in particular the activation of peripheral leukocytes, namely T cells, we hypothesized that finding a treatment designed to inhibit neuroantigen-specific T cells and block cytotoxic monocytes and macrophages may render therapeutic effects in stroke. We previously reported that subcutaneous administration of partial MHC class II constructs promote behavioral and histological effects in stroke mice by centrally promoting a protective M2 macrophage/microglia phenotype in the CNS and peripherally reversing stroke-associated splenic atrophy. Here, we employed a second species using adult Sprague-Dawley rats exposed to the middle cerebral artery occlusion stroke model and observed similar therapeutic effects with a mouse partial MHC class II construct called DRmQ, as evidenced by reductions in stroke-induced motor deficits, infarcts, and peri-infarct cell loss and neuroinflammation. More importantly, we offered further evidence of peripheral sequestration of inflammation at the level of the spleen, which was characterized by attenuation of stroke-induced spleen weight reduction and TNF-ɑ and IL-6 upregulation. Collectively, these results satisfy the Stroke Therapy Academic Industry Roundtable criteria of testing a novel therapeutic in a second species and support the use of partial MHC class II constructs as a stroke therapeutic designed to sequester both central and peripheral inflammation responses in an effort to retard, or even halt, the neuroinflammation that exacerbates the secondary cell death in stroke.
Collapse
Affiliation(s)
- Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Vanessa Castelli
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Alexandreya B Coats
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Lisset Navarro-Torres
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Julian Garcia-Sanchez
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Chase Kingsbury
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Hung Nguyen
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710, SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology and Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Roberto Meza-Romero
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710, SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology and Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710, SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology and Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
10
|
Brown J, Kingsbury C, Lee J, Vandenbark AA, Meza‐Romero R, Offner H, Borlongan CV. Spleen participation in partial MHC class II construct neuroprotection in stroke. CNS Neurosci Ther 2020; 26:663-669. [PMID: 32237074 PMCID: PMC7298973 DOI: 10.1111/cns.13369] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Pathological progression of stroke in the peripheral and central nervous systems (PNS and CNS) is characterized by multiple converging signalling pathways that exacerbate neuroinflammation-mediated secondary cell death. This creates a need for a novel type of immunotherapy capable of simultaneously lowering the synergistic inflammatory responses in the PNS and CNS, specifically the spleen and brain. Previously, we demonstrated that partial major histocompatibility complex (MHC) class II constructs can be administered subcutaneously to promote histological and behavioural effects that alleviate common symptoms found in a murine model of transient stroke. This MHC class II manipulates T cell cytokine expression in both PNS and CNS, resulting in dampened inflammation. In our long-standing efforts towards translational research, we recently demonstrated that a potent next generation mouse-based partial MHC class II construct named DRmQ (DRa1L50Q -mMOG-35-55) similarly induces neuroprotection in stroke rats, replicating the therapeutic effects of the human homolog as DRhQ (DRa1L50Q -human (h)MOG-35-55) in stroke mice. Our preclinical studies showed that DRmQ reduces motor deficits, infarct volume and peri-infarct cell loss by targeting inflammation in this second species. Moreover, we provided mechanistic support in both animal studies that partial MHC class II constructs effectively modulate the spleen, an organ which plays a critical role in modulating secondary cell death. Together, these preclinical studies satisfy testing the constructs in two stroke models, which is a major criterion of the Stroke Therapy Academic Industry Roundtable (STAIR) criteria and a key step in effectively translating this drug to the clinic. Additional translational studies, including dose-response and larger animal models may be warranted to bring MHC class II constructs closer to the clinic.
Collapse
Affiliation(s)
- John Brown
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Chase Kingsbury
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Jea‐Young Lee
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Arthur A. Vandenbark
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Roberto Meza‐Romero
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Halina Offner
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| |
Collapse
|
11
|
Vandenbark AA, Meza-Romero R, Benedek G, Offner H. A novel neurotherapeutic for multiple sclerosis, ischemic injury, methamphetamine addiction, and traumatic brain injury. J Neuroinflammation 2019; 16:14. [PMID: 30683115 PMCID: PMC6346590 DOI: 10.1186/s12974-018-1393-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023] Open
Abstract
Neurovascular, autoimmune, and traumatic injuries of the central nervous system (CNS) all have in common an initial acute inflammatory response mediated by influx across the blood-brain barrier of activated mononuclear cells followed by chronic and often progressive disability. Although some anti-inflammatory therapies can reduce cellular infiltration into the initial lesions, there are essentially no effective treatments for the progressive phase. We here review the successful treatment of animal models for four separate neuroinflammatory and neurodegenerative CNS conditions using a single partial MHC class II construct called DRa1-hMOG-35-55 or its newest iteration, DRa1(L50Q)-hMOG-35-55 (DRhQ) that can be administered without a need for class II tissue type matching due to the conserved DRα1 moiety of the drug. These constructs antagonize the cognate TCR and bind with high affinity to their cell-bound CD74 receptor on macrophages and dendritic cells, thereby competitively inhibiting downstream signaling and pro-inflammatory effects of macrophage migration inhibitory factor (MIF) and its homolog, d-dopachrome tautomerase (D-DT=MIF-2) that bind to identical residues of CD74 leading to progressive disease. These effects suggest the existence of a common pathogenic mechanism involving a chemokine-driven influx of activated monocytes into the CNS tissue that can be reversed by parenteral injection of the DRa1-MOG-35-55 constructs that also induce anti-inflammatory macrophages and microglia within the CNS. Due to their ability to block this common pathway, these novel drugs appear to be prime candidates for therapy of a wide range of neuroinflammatory and neurodegenerative CNS conditions.
Collapse
Affiliation(s)
- Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA. .,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA. .,Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Roberto Meza-Romero
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Gil Benedek
- Present Address: Tissue Typing and Immunogenetics Laboratory, Hadassah Medical Center, Jerusalem, Israel
| | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.,Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| |
Collapse
|
12
|
Wan X, Pei W, Shahzad KA, Zhang L, Song S, Jin X, Wang L, Zhao C, Shen C. A Tolerogenic Artificial APC Durably Ameliorates Experimental Autoimmune Encephalomyelitis by Directly and Selectively Modulating Myelin Peptide–Autoreactive CD4+and CD8+T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1194-1210. [DOI: 10.4049/jimmunol.1800108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/19/2018] [Indexed: 01/19/2023]
|
13
|
Liu DD, Chu SF, Chen C, Yang PF, Chen NH, He X. Research progress in stroke-induced immunodepression syndrome (SIDS) and stroke-associated pneumonia (SAP). Neurochem Int 2018; 114:42-54. [DOI: 10.1016/j.neuint.2018.01.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
|
14
|
Yang L, Liu Z, Ren H, Zhang L, Gao S, Ren L, Chai Z, Meza-Romero R, Benedek G, Vandenbark AA, Offner H, Li M. DRα1-MOG-35-55 treatment reduces lesion volumes and improves neurological deficits after traumatic brain injury. Metab Brain Dis 2017; 32:1395-1402. [PMID: 28303450 PMCID: PMC5600636 DOI: 10.1007/s11011-017-9991-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/08/2017] [Indexed: 10/20/2022]
Abstract
Traumatic brain injury (TBI) results in severe neurological impairments without effective treatments. Inflammation appears to be an important contributor to key pathogenic events such as secondary brain injury following TBI and therefore serves as a promising target for novel therapies. We have recently demonstrated the ability of a molecular construct comprised of the human leukocyte antigen (HLA)-DRα1 domain linked covalently to mouse (m)MOG-35-55 peptide (DRα1-MOG-35-55 construct) to reduce CNS inflammation and tissue injury in animal models of multiple sclerosis and ischemic stroke. The aim of the current study was to determine if DRα1-MOG-35-55 treatment of a fluid percussion injury (FPI) mouse model of TBI could reduce the lesion size and improve disease outcome measures. Neurodeficits, lesion size, and immune responses were determined to evaluate the therapeutic potential and mechanisms of neuroprotection induced by DRα1-MOG-35-55 treatment. The results demonstrated that daily injections of DRα1-MOG-35-55 given after FPI significantly reduced numbers of infiltrating CD74+ and CD86+ macrophages and increased numbers of CD206+ microglia in the brain concomitant with smaller lesion sizes and improvement in neurodeficits. Conversely, DRα1-MOG-35-55 treatment of TBI increased numbers of circulating CD11b+ monocytes and their expression of CD74 but had no detectable effect on cell numbers or marker expression in the spleen. These results demonstrate that DRα1-MOG-35-55 therapy can reduce CNS inflammation and significantly improve histological and clinical outcomes after TBI. Future studies will further examine the potential of DRα1-MOG-35-55 for treatment of TBI.
Collapse
Affiliation(s)
- Liu Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
- St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Zhijia Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Lei Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Siman Gao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Li Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhi Chai
- "2011"Collaborative Innovation Center/Neurobiology Research Center, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, 030619, China
| | - Roberto Meza-Romero
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
- Tykeson MS Research Laboratory, Department of Neurology UHS-46, Oregon Health & Science University, Portland, OR, USA
| | - Gil Benedek
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
- Tykeson MS Research Laboratory, Department of Neurology UHS-46, Oregon Health & Science University, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
- Tykeson MS Research Laboratory, Department of Neurology UHS-46, Oregon Health & Science University, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA.
- Tykeson MS Research Laboratory, Department of Neurology UHS-46, Oregon Health & Science University, Portland, OR, USA.
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
| | - Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA.
| |
Collapse
|
15
|
Abstract
To achieve success in developing more effective treatments for stroke, we need a better understanding in all aspects of stroke including prevention, diagnosis, treatment, and post-stroke recovery and complications. The objective of this special issue is to bring to the readership of Neurochemistry International the latest developments and knowledge in a broad spectrum of areas of stroke research in both review and original research articles. Topics include neuroprotective diets, biomarkers used to aid clinical management, neurodegenerative as well as neuroprotective effects of the immune system, potential therapeutic targets, engineered growth factors that promote endogenous neuroregeneration, mechanisms of cerebral small vessel disease, and post stroke epilepsy.
Collapse
|
16
|
Yang L, Kong Y, Ren H, Li M, Wei CJ, Shi E, Jin WN, Hao J, Vandenbark AA, Offner H. Upregulation of CD74 and its potential association with disease severity in subjects with ischemic stroke. Neurochem Int 2016; 107:148-155. [PMID: 27884769 DOI: 10.1016/j.neuint.2016.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 10/20/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a key cytokine/chemokine in the activation and recruitment of inflammatory T lymphocytes known to exacerbate experimental stroke severity. MIF effects are mediated through its primary cellular receptor, CD74, the MHC class II invariant chain present on all class II expressing cells, including monocytes, macrophages and dendritic cells (DC). We demonstrated previously that partial MHC class II/peptide constructs (pMHC) can effectively treat mice with experimental stroke, in part through their ability to competitively inhibit MIF/CD74 interactions and downstream signaling. However, the role of MIF and CD74 in human ischemic stroke is not yet well established. To evaluate the therapeutic potential for pMHC, we assessed MIF and CD74 expression levels and their association with disease outcome in subjects with ischemic stroke. MIF levels were assessed in blood plasma by ELISA and CD74 expression was quantified by flow cytometry and qRT-PCR in peripheral blood mononuclear cells (PBMCs) obtained from subjects with ischemic stroke and age and sex-matched healthy controls (HC). MIF levels were increased in plasma and the number of CD74+ cells and CD74 mRNA expression levels were significantly increased in PBMC of subjects with ischemic stroke versus HC, mainly on CD4+ T cells, monocytes and DC. Greater increases of CD74+ cells were seen in subjects with cortical vs. subcortical infarcts and the number of CD74+ cells in blood correlated strongly with infarct size and neurological outcomes. However, differences in MIF and CD74 expression were not affected by age, gender or lesion laterality. Increased CD74 expression levels may serve as a useful biomarker for worse stroke severity and predicted outcomes in subjects with ischemic stroke and provide a rationale for potential future treatment with pMHC constructs.
Collapse
Affiliation(s)
- Liu Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Kong
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chang-Juan Wei
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Elaine Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei-Na Jin
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Arthur A Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|