1
|
Marks K, Ahn SJ, Rai N, Anfray A, Iadecola C, Anrather J. A minimally invasive thrombotic stroke model to study circadian rhythm in awake mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598243. [PMID: 38915621 PMCID: PMC11195071 DOI: 10.1101/2024.06.10.598243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Experimental stroke models in rodents are essential for mechanistic studies and therapeutic development. However, these models have several limitations negatively impacting their translational relevance. Here we aimed to develop a minimally invasive thrombotic stroke model through magnetic particle delivery that does not require craniotomy, is amenable to reperfusion therapy, can be combined with in vivo imaging modalities, and can be performed in awake mice. We found that the model results in reproducible cortical infarcts within the middle cerebral artery (MCA) with cytologic and immune changes similar to that observed with more invasive distal MCA occlusion models. Importantly, the injury produced by the model was ameliorated by tissue plasminogen activator (tPA) administration. We also show that MCA occlusion in awake animals results in bigger ischemic lesions independent of day/night cycle. Magnetic particle delivery had no overt effects on physiologic parameters and systemic immune biomarkers. In conclusion, we developed a novel stroke model in mice that fulfills many requirements for modeling human stroke.
Collapse
|
2
|
Wu S, Tabassum S, Payne CT, Hu H, Gusdon AM, Choi HA, Ren XS. Updates of the role of B-cells in ischemic stroke. Front Cell Neurosci 2024; 18:1340756. [PMID: 38550918 PMCID: PMC10972894 DOI: 10.3389/fncel.2024.1340756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/27/2024] [Indexed: 10/11/2024] Open
Abstract
Ischemic stroke is a major disease causing death and disability in the elderly and is one of the major diseases that seriously threaten human health and cause a great economic burden. In the early stage of ischemic stroke, neuronal structure is destroyed, resulting in death or damage, and the release of a variety of damage-associated pattern molecules induces an increase in neuroglial activation, peripheral immune response, and secretion of inflammatory mediators, which further exacerbates the damage to the blood-brain barrier, exacerbates cerebral edema, and microcirculatory impairment, triggering secondary brain injuries. After the acute phase of stroke, various immune cells initiate a protective effect, which is released step by step and contributes to the repair of neuronal cells through phenotypic changes. In addition, ischemic stroke induces Central Nervous System (CNS) immunosuppression, and the interaction between the two influences the outcome of stroke. Therefore, modulating the immune response of the CNS to reduce the inflammatory response and immune damage during stroke is important for the protection of brain function and long-term recovery after stroke, and modulating the immune function of the CNS is expected to be a novel therapeutic strategy. However, there are fewer studies on B-cells in brain function protection, which may play a dual role in the stroke process, and the understanding of this cell is still incomplete. We review the existing studies on the mechanisms of the role of B-cells, inflammatory response, and immune response in the development of ischemic stroke and provide a reference for the development of adjuvant therapeutic drugs for ischemic stroke targeting inflammatory injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuefang S. Ren
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
3
|
Woodell AS, Landoni E, Valdivia A, Buckley A, Ogunnaike EA, Dotti G, Hingtgen SD. Utilizing induced neural stem cell-based delivery of a cytokine cocktail to enhance chimeric antigen receptor-modified T-cell therapy for brain cancer. Bioeng Transl Med 2023; 8:e10538. [PMID: 38023712 PMCID: PMC10658508 DOI: 10.1002/btm2.10538] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 12/01/2023] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cell therapy has shown enormous clinical promise against blood cancers, yet efficacy against solid tumors remains a challenge. Here, we investigated the potential of a new combination cell therapy, where tumor-homing induced neural stem cells (iNSCs) are used to enhance CAR-T-cell therapy and achieve efficacious suppression of brain tumors. Using in vitro and in vivo migration assays, we found iNSC-secreted RANTES/IL-15 increased CAR-T-cell migration sixfold and expansion threefold, resulting in greater antitumor activity in a glioblastoma (GBM) tumor model. Furthermore, multimodal imaging showed iNSC delivery of RANTES/IL-15 in combination with intravenous administration of CAR-T cells reduced established orthotopic GBM xenografts 2538-fold within the first week, followed by durable tumor remission through 60 days post-treatment. By contrast, CAR-T-cell therapy alone only partially controlled tumor growth, with a median survival of only 19 days. Together, these studies demonstrate the potential of combined cell therapy platforms to improve the efficacy of CAR-T-cell therapy for brain tumors.
Collapse
Affiliation(s)
- Alex S. Woodell
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Elisa Landoni
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Andrew Buckley
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Edikan A. Ogunnaike
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Center for Nanotechnology in Drug Delivery, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Shawn D. Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
4
|
Sharp RC, Guenther DT, Farrer MJ. Experimental procedures for flow cytometry of wild-type mouse brain: a systematic review. Front Immunol 2023; 14:1281705. [PMID: 38022545 PMCID: PMC10646240 DOI: 10.3389/fimmu.2023.1281705] [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: 08/22/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The aim of this study was to systematically review the neuroimmunology literature to determine the average immune cell counts reported by flow cytometry in wild-type (WT) homogenized mouse brains. Background Mouse models of gene dysfunction are widely used to study age-associated neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. The importance of the neuroimmune system in these multifactorial disorders has become increasingly evident, and methods to quantify resident and infiltrating immune cells in the brain, including flow cytometry, are necessary. However, there appears to be no consensus on the best approach to perform flow cytometry or quantify/report immune cell counts. The development of more standardized methods would accelerate neuroimmune discovery and validation by meta-analysis. Methods There has not yet been a systematic review of 'neuroimmunology' by 'flow cytometry' via examination of the PROSPERO registry. A protocol for a systematic review was subsequently based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) using the Studies, Data, Methods, and Outcomes (SDMO) criteria. Literature searches were conducted in the Google Scholar and PubMed databases. From that search, 900 candidate studies were identified, and 437 studies were assessed for eligibility based on formal exclusion criteria. Results Out of the 437 studies reviewed, 58 were eligible for inclusion and comparative analysis. Each study assessed immune cell subsets within homogenized mouse brains and used flow cytometry. Nonetheless, there was considerable variability in the methods, data analysis, reporting, and results. Descriptive statistics have been presented on the study designs and results, including medians with interquartile ranges (IQRs) and overall means with standard deviations (SD) for specific immune cell counts and their relative proportions, within and between studies. A total of 58 studies reported the most abundant immune cells within the brains were TMEM119+ microglia, bulk CD4+ T cells, and bulk CD8+ T cells. Conclusion Experiments to conduct and report flow cytometry data, derived from WT homogenized mouse brains, would benefit from a more standardized approach. While within-study comparisons are valid, the variability in methods of counting of immune cell populations is too broad for meta-analysis. The inclusion of a minimal protocol with more detailed methods, controls, and standards could enable this nascent field to compare results across studies.
Collapse
Affiliation(s)
| | | | - Matthew J. Farrer
- Department of Neurology, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Nong J, Glassman PM, Myerson JW, Zuluaga-Ramirez V, Rodriguez-Garcia A, Mukalel A, Omo-Lamai S, Walsh LR, Zamora ME, Gong X, Wang Z, Bhamidipati K, Kiseleva RY, Villa CH, Greineder CF, Kasner SE, Weissman D, Mitchell MJ, Muro S, Persidsky Y, Brenner JS, Muzykantov VR, Marcos-Contreras OA. Targeted Nanocarriers Co-Opting Pulmonary Intravascular Leukocytes for Drug Delivery to the Injured Brain. ACS NANO 2023; 17:13121-13136. [PMID: 37432926 PMCID: PMC10373654 DOI: 10.1021/acsnano.2c08275] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 06/08/2023] [Indexed: 07/13/2023]
Abstract
Ex vivo-loaded white blood cells (WBC) can transfer cargo to pathological foci in the central nervous system (CNS). Here we tested affinity ligand driven in vivo loading of WBC in order to bypass the need for ex vivo WBC manipulation. We used a mouse model of acute brain inflammation caused by local injection of tumor necrosis factor alpha (TNF-α). We intravenously injected nanoparticles targeted to intercellular adhesion molecule 1 (anti-ICAM/NP). We found that (A) at 2 h, >20% of anti-ICAM/NP were localized to the lungs; (B) of the anti-ICAM/NP in the lungs >90% were associated with leukocytes; (C) at 6 and 22 h, anti-ICAM/NP pulmonary uptake decreased; (D) anti-ICAM/NP uptake in brain increased up to 5-fold in this time interval, concomitantly with migration of WBCs into the injured brain. Intravital microscopy confirmed transport of anti-ICAM/NP beyond the blood-brain barrier and flow cytometry demonstrated complete association of NP with WBC in the brain (98%). Dexamethasone-loaded anti-ICAM/liposomes abrogated brain edema in this model and promoted anti-inflammatory M2 polarization of macrophages in the brain. In vivo targeted loading of WBC in the intravascular pool may provide advantages of coopting WBC predisposed to natural rapid mobilization from the lungs to the brain, connected directly via conduit vessels.
Collapse
Affiliation(s)
- Jia Nong
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Patrick M. Glassman
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Pharmaceutical Sciences, Temple University
School of Pharmacy, Philadelphia, Pennsylvania 19140, United States
| | - Jacob W. Myerson
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Viviana Zuluaga-Ramirez
- Department
of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Alba Rodriguez-Garcia
- Department
of Pathology and Laboratory Medicine, Ovarian Cancer Research Center,
Perelman School of Medicine, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center
for Cellular Immunotherapies, Abramson Cancer Center, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alvin Mukalel
- Department
of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- Division
of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R. Walsh
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marco E. Zamora
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- School
of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Xijing Gong
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Division
of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Zhicheng Wang
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kartik Bhamidipati
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Raisa Y. Kiseleva
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H. Villa
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin Fred Greineder
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Scott E. Kasner
- Department
of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Drew Weissman
- Division
of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J. Mitchell
- Department
of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson
Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute
for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular
Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute
for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain
- Institute of Catalonia for Research and
Advanced Studies (ICREA), Barcelona, 08010, Spain
- Institute
for Bioscience and Biotechnology (IBBR), College Park, Maryland 20850, United States
| | - Yuri Persidsky
- Department
of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Jacob Samuel Brenner
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Division
of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R. Muzykantov
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A. Marcos-Contreras
- Department
of Systems Pharmacology and Translational Therapeutics, Perelman School
of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
6
|
Nong J, Glassman PM, Reyes-Esteves S, Descamps HC, Shuvaev VV, Kiseleva RY, Papp TE, Alameh MG, Tam YK, Mui BL, Omo-Lamai S, Zamora ME, Shuvaeva T, Arguiri E, Thaiss CA, Myerson JW, Weissman D, Kasner SE, Parhiz H, Muzykantov VR, Brenner JS, Marcos-Contreras OA. Targeting lipid nanoparticles to the blood brain barrier to ameliorate acute ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544645. [PMID: 37398465 PMCID: PMC10312645 DOI: 10.1101/2023.06.12.544645] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
After more than 100 failed drug trials for acute ischemic stroke (AIS), one of the most commonly cited reasons for the failure has been that drugs achieve very low concentrations in the at-risk penumbra. To address this problem, here we employ nanotechnology to significantly enhance drug concentration in the penumbra's blood-brain barrier (BBB), whose increased permeability in AIS has long been hypothesized to kill neurons by exposing them to toxic plasma proteins. To devise drug-loaded nanocarriers targeted to the BBB, we conjugated them with antibodies that bind to various cell adhesion molecules on the BBB endothelium. In the transient middle cerebral artery occlusion (tMCAO) mouse model, nanocarriers targeted with VCAM antibodies achieved the highest level of brain delivery, nearly 2 orders of magnitude higher than untargeted ones. VCAM-targeted lipid nanoparticles loaded with either a small molecule drug (dexamethasone) or mRNA (encoding IL-10) reduced cerebral infarct volume by 35% or 73%, respectively, and both significantly lowered mortality rates. In contrast, the drugs delivered without the nanocarriers had no effect on AIS outcomes. Thus, VCAM-targeted lipid nanoparticles represent a new platform for strongly concentrating drugs within the compromised BBB of penumbra, thereby ameliorating AIS. Graphical abstract Acute ischemic stroke induces upregulation of VCAM. We specifically targeted upregulated VCAM in the injured region of the brain with drug- or mRNA-loaded targeted nanocarriers. Nanocarriers targeted with VCAM antibodies achieved the highest brain delivery, nearly orders of magnitude higher than untargeted ones. VCAM-targeted nanocarriers loaded with dexamethasone and mRNA encoding IL-10 reduced infarct volume by 35% and 73%, respectively, and improved survival rates.
Collapse
|
7
|
Kim BH, Hadas E, Kelschenbach J, Chao W, Gu CJ, Potash MJ, Volsky DJ. CCL2 is required for initiation but not persistence of HIV infection mediated neurocognitive disease in mice. Sci Rep 2023; 13:6577. [PMID: 37085605 PMCID: PMC10121554 DOI: 10.1038/s41598-023-33491-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/13/2023] [Indexed: 04/23/2023] Open
Abstract
HIV enters the brain within days of infection causing neurocognitive impairment (NCI) in up to half of infected people despite suppressive antiretroviral therapy. The virus is believed to enter the brain in infected monocytes through chemotaxis to the major monocyte chemokine, CCL2, but the roles of CCL2 in established NCI are not fully defined. We addressed this question during infection of conventional and CCL2 knockout mice with EcoHIV in which NCI can be verified in behavioral tests. EcoHIV enters mouse brain within 5 days of infection, but NCI develops gradually with established cognitive disease starting 25 days after infection. CCL2 knockout mice infected by intraperitoneal injection of virus failed to develop brain infection and NCI. However, when EcoHIV was directly injected into the brain, CCL2 knockout mice developed NCI. Knockout of CCL2 or its principal receptor, CCR2, slightly reduced macrophage infection in culture. Treatment of mice prior to and during EcoHIV infection with the CCL2 transcriptional inhibitor, bindarit, prevented brain infection and NCI and reduced macrophage infection. In contrast, bindarit treatment of mice 4 weeks after infection affected neither brain virus burden nor NCI. Based on these findings we propose that HIV enters the brain mainly through infected monocytes but that resident brain cells are sufficient to maintain NCI. These findings suggest that NCI therapy must act within the brain.
Collapse
Affiliation(s)
- Boe-Hyun Kim
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Eran Hadas
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Jennifer Kelschenbach
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Wei Chao
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Chao-Jiang Gu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
- College of Life and Health Sciences, Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Mary Jane Potash
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - David J Volsky
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
8
|
Swan SL, Mehta N, Ilich E, Shen SH, Wilkinson DS, Anderson AR, Segura T, Sanchez-Perez L, Sampson JH, Bellamkonda RV. IL7 and IL7 Flt3L co-expressing CAR T cells improve therapeutic efficacy in mouse EGFRvIII heterogeneous glioblastoma. Front Immunol 2023; 14:1085547. [PMID: 36817432 PMCID: PMC9936235 DOI: 10.3389/fimmu.2023.1085547] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy in glioblastoma faces many challenges including insufficient CAR T cell abundance and antigen-negative tumor cells evading targeting. Unfortunately, preclinical studies evaluating CAR T cells in glioblastoma focus on tumor models that express a single antigen, use immunocompromised animals, and/or pre-treat with lymphodepleting agents. While lymphodepletion enhances CAR T cell efficacy, it diminishes the endogenous immune system that has the potential for tumor eradication. Here, we engineered CAR T cells to express IL7 and/or Flt3L in 50% EGFRvIII-positive and -negative orthotopic tumors pre-conditioned with non-lymphodepleting irradiation. IL7 and IL7 Flt3L CAR T cells increased intratumoral CAR T cell abundance seven days after treatment. IL7 co-expression with Flt3L modestly increased conventional dendritic cells as well as the CD103+XCR1+ population known to have migratory and antigen cross-presenting capabilities. Treatment with IL7 or IL7 Flt3L CAR T cells improved overall survival to 67% and 50%, respectively, compared to 9% survival with conventional or Flt3L CAR T cells. We concluded that CAR T cells modified to express IL7 enhanced CAR T cell abundance and improved overall survival in EGFRvIII heterogeneous tumors pre-conditioned with non-lymphodepleting irradiation. Potentially IL7 or IL7 Flt3L CAR T cells can provide new opportunities to combine CAR T cells with other immunotherapies for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Sheridan L Swan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Nalini Mehta
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Ekaterina Ilich
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Steven H Shen
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Daniel S Wilkinson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Alexa R Anderson
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Tatiana Segura
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States.,Clinical Science Departments of Neurology and Dermatology, Duke University, Durham, NC, United States
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Pathology, Duke University Medical Center, Durham, NC, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Ravi V Bellamkonda
- Department of Biology, Emory University, Atlanta, GA, United States.,Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
| |
Collapse
|
9
|
Yau ACY, Globisch MA, Onyeogaziri FC, Conze LL, Smith R, Jauhiainen S, Corada M, Orsenigo F, Huang H, Herre M, Olsson AK, Malinverno M, Sundell V, Rezai Jahromi B, Niemelä M, Laakso A, Garlanda C, Mantovani A, Lampugnani MG, Dejana E, Magnusson PU. Inflammation and neutrophil extracellular traps in cerebral cavernous malformation. Cell Mol Life Sci 2022; 79:206. [PMID: 35333979 PMCID: PMC8949649 DOI: 10.1007/s00018-022-04224-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Cerebral Cavernous Malformation (CCM) is a brain vascular disease with various neurological symptoms. In this study, we describe the inflammatory profile in CCM and show for the first time the formation of neutrophil extracellular traps (NETs) in rodents and humans with CCM. Through RNA-seq analysis of cerebellum endothelial cells from wild-type mice and mice with an endothelial cell-specific ablation of the Ccm3 gene (Ccm3iECKO), we show that endothelial cells from Ccm3iECKO mice have an increased expression of inflammation-related genes. These genes encode proinflammatory cytokines and chemokines, as well as adhesion molecules, which promote recruitment of inflammatory and immune cells. Similarly, immunoassays showed elevated levels of these cytokines and chemokines in the cerebellum of the Ccm3iECKO mice. Consistently, both flow cytometry and immunofluorescence analysis showed infiltration of different subsets of leukocytes into the CCM lesions. Neutrophils, which are known to fight against infection through different strategies, including the formation of NETs, represented the leukocyte subset within the most pronounced increase in CCM. Here, we detected elevated levels of NETs in the blood and the deposition of NETs in the cerebral cavernomas of Ccm3iECKO mice. Degradation of NETs by DNase I treatment improved the vascular barrier. The deposition of NETs in the cavernomas of patients with CCM confirms the clinical relevance of NETs in CCM.
Collapse
Affiliation(s)
- Anthony C Y Yau
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Maria Ascencion Globisch
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Favour Chinyere Onyeogaziri
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Lei L Conze
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Ross Smith
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Suvi Jauhiainen
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Monica Corada
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Fabrizio Orsenigo
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Melanie Herre
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Matteo Malinverno
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Veronica Sundell
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Behnam Rezai Jahromi
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Niemelä
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Maria Grazia Lampugnani
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy.,Mario Negri Institute for Pharmacological Research, 20157, Milan, Italy
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden.,Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden.
| |
Collapse
|
10
|
Fagan SG, Bechet S, Dev KK. Fingolimod Rescues Memory and Improves Pathological Hallmarks in the 3xTg-AD Model of Alzheimer's Disease. Mol Neurobiol 2022; 59:1882-1895. [PMID: 35031916 PMCID: PMC8882098 DOI: 10.1007/s12035-021-02613-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/19/2021] [Indexed: 10/26/2022]
Abstract
Therapeutic strategies for Alzheimer's disease (AD) have largely focused on the regulation of amyloid pathology while those targeting tau pathology, and inflammatory mechanisms are less explored. In this regard, drugs with multimodal and concurrent targeting of Aβ, tau, and inflammatory processes may offer advantages. Here, we investigate one such candidate drug in the triple transgenic 3xTg-AD mouse model of AD, namely the disease-modifying oral neuroimmunomodulatory therapeutic used in patients with multiple sclerosis, called fingolimod. In this study, administration of fingolimod was initiated after behavioral symptoms are known to emerge, at 6 months of age. Treatment continued to 12 months when behavioral tests were performed and thereafter histological and biochemical analysis was conducted on postmortem tissue. The results demonstrate that fingolimod reverses deficits in spatial working memory at 8 and 12 months of age as measured by novel object location and Morris water maze tests. Inflammation in the brain is alleviated as demonstrated by reduced Iba1-positive and CD3-positive cell number, less ramified microglial morphology, and improved cytokine profile. Finally, treatment with fingolimod was shown to reduce phosphorylated tau and APP levels in the hippocampus and cortex. These results highlight the potential of fingolimod as a multimodal therapeutic for the treatment of AD.
Collapse
Affiliation(s)
- Steven G Fagan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Sibylle Bechet
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
11
|
Lee HN, Manangeeswaran M, Lewkowicz AP, Engel K, Chowdhury M, Garige M, Eckhaus MA, Sourbier C, Ireland DD, Verthelyi D. NK cells require immune checkpoint receptor LILRB4/gp49B to control neurotropic Zika virus infections in mice. JCI Insight 2022; 7:151420. [PMID: 35132958 PMCID: PMC8855830 DOI: 10.1172/jci.insight.151420] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Immune cells express an array of inhibitory checkpoint receptors that are upregulated upon activation and limit tissue damage associated with excessive response to pathogens or allergens. Mouse leukocyte immunoglobulin like receptor B4 (LILRB4), also known as glycoprotein 49B (gp49B), is an inhibitory checkpoint receptor constitutively expressed in myeloid cells and upregulated in B cells, T cells, and NK cells upon activation. Here, we report that expression of LILRB4, which binds Zika virus (ZIKV), was increased in microglia and myeloid cells infiltrating the brains of neonatal mice with ZIKV-associated meningoencephalitis. Importantly, while C57BL/6 mice developed transient neurological symptoms but survived infection, mice lacking LILRB4/gp49B (LILRB4 KO) exhibited more severe signs of neurological disease and succumbed to disease. Their brains showed increased cellular infiltration but reduced control of viral burden. The reduced viral clearance was associated with altered NK cell function in the absence of LILRB4/gp49B. In naive animals, this manifested as reduced granzyme B responses to stimulation, but in ZIKV-infected animals, NK cells showed phenotypic changes that suggested altered maturation, diminished glucose consumption, reduced IFN-γ and granzyme B production, and impaired cytotoxicity. Together, our data reveal LILRB4/gp49B as an important regulator of NK cell function during viral infections.
Collapse
Affiliation(s)
- Ha-Na Lee
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Mohanraj Manangeeswaran
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Aaron P Lewkowicz
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Kaliroi Engel
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Monica Chowdhury
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Mamatha Garige
- Laboratory of Molecular Oncology, Division of Biotechnology Review and Research-I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Michael A Eckhaus
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Carole Sourbier
- Laboratory of Molecular Oncology, Division of Biotechnology Review and Research-I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Derek Dc Ireland
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| | - Daniela Verthelyi
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, and
| |
Collapse
|
12
|
Amu S, Malone K. Flow cytometry and stroke: from current methodology to future applications. Neural Regen Res 2022; 17:1748-1750. [PMID: 35017430 PMCID: PMC8820698 DOI: 10.4103/1673-5374.332138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Sylvie Amu
- Cancer Research , Ireland.,UCC, University College Cork, Cork, Ireland
| | - Kyle Malone
- Department of Pharmacology and Therapeutics, Western Gateway Building; School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Zhang L, Wei W, Ai X, Kilic E, Hermann DM, Venkataramani V, Bähr M, Doeppner TR. Extracellular vesicles from hypoxia-preconditioned microglia promote angiogenesis and repress apoptosis in stroke mice via the TGF-β/Smad2/3 pathway. Cell Death Dis 2021; 12:1068. [PMID: 34753919 PMCID: PMC8578653 DOI: 10.1038/s41419-021-04363-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/15/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022]
Abstract
Systemic transplantation of oxygen-glucose deprivation (OGD)-preconditioned primary microglia enhances neurological recovery in rodent stroke models, albeit the underlying mechanisms have not been sufficiently addressed. Herein, we analyzed whether or not extracellular vesicles (EVs) derived from such microglia are the biological mediators of these observations and which signaling pathways are involved in the process. Exposing bEnd.3 endothelial cells (ECs) and primary cortical neurons to OGD, the impact of EVs from OGD-preconditioned microglia on angiogenesis and neuronal apoptosis by the tube formation assay and TUNEL staining was assessed. Under these conditions, EV treatment stimulated both angiogenesis and tube formation in ECs and repressed neuronal cell injury. Characterizing microglia EVs by means of Western blot analysis and other techniques revealed these EVs to be rich in TGF-β1. The latter turned out to be a key compound for the therapeutic potential of microglia EVs, affecting the Smad2/3 pathway in both ECs and neurons. EV infusion in stroke mice confirmed the aforementioned in vitro results, demonstrating an activation of the TGF-β/Smad2/3 signaling pathway within the ischemic brain. Furthermore, enriched TGF-β1 in EVs secreted from OGD-preconditioned microglia stimulated M2 polarization of residing microglia within the ischemic cerebral environment, which may contribute to a regulation of an early inflammatory response in postischemic hemispheres. These observations are not only interesting from the mechanistic point of view but have an immediate therapeutic implication as well, since stroke mice treated with such EVs displayed a better functional recovery in the behavioral test analyses. Hence, the present findings suggest a new way of action of EVs derived from OGD-preconditioned microglia by regulating the TGF-β/Smad2/3 pathway in order to promote tissue regeneration and neurological recovery in stroke mice.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoyu Ai
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Ertugrul Kilic
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Vivek Venkataramani
- Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
14
|
Comparative Review of Microglia and Monocytes in CNS Phagocytosis. Cells 2021; 10:cells10102555. [PMID: 34685535 PMCID: PMC8534258 DOI: 10.3390/cells10102555] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023] Open
Abstract
Macrophages maintain tissue homeostasis by phagocytosing and removing unwanted materials such as dead cells and cell debris. Microglia, the resident macrophages of the central nervous system (CNS), are no exception. In addition, a series of recent studies have shown that microglia phagocytose the neuronal synapses that form the basis of neural circuit function. This discovery has spurred many neuroscientists to study microglia. Importantly, in the CNS parenchyma, not only microglia but also blood-derived monocytes, which essentially differentiate into macrophages after infiltration, exert phagocytic ability, making the study of phagocytosis in the CNS even more interesting and complex. In particular, in the diseased brain, the phagocytosis of tissue-damaging substances, such as myelin debris in multiple sclerosis (MS), has been shown to be carried out by both microglia and blood-derived monocytes. However, it remains largely unclear why blood-derived monocytes need to invade the parenchyma, where microglia are already abundant, to assist in phagocytosis. We will also discuss whether this phagocytosis can affect the fate of the phagocytosing cell itself as well as the substance being phagocytosed and the surrounding environment in addition to future research directions. In this review, we will introduce recent studies to answer a question that often arises when studying microglial phagocytosis: under what circumstances and to what extent blood-derived monocytes infiltrate the CNS and contribute to phagocytosis. In addition, the readers will learn how recent studies have experimentally distinguished between microglia and infiltrating monocytes. Finally, we aim to contribute to the progress of phagocytosis research by discussing the effects of phagocytosis on phagocytic cells.
Collapse
|
15
|
Zhang C, Tian R, Dreifus EM, Hashemi Shahraki A, Holt G, Cai R, Griswold A, Bejarano P, Jackson R, V Schally A, Mirsaeidi M. Activity of the growth hormone-releasing hormone antagonist MIA602 and its underlying mechanisms of action in sarcoidosis-like granuloma. Clin Transl Immunology 2021; 10:e1310. [PMID: 34257968 PMCID: PMC8256670 DOI: 10.1002/cti2.1310] [Citation(s) in RCA: 3] [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: 01/19/2021] [Revised: 04/09/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Growth hormone-releasing hormone (GHRH) is a potent stimulator of growth hormone (GH) secretion from the pituitary gland. Although GHRH is essential for the growth of immune cells, the regulatory effects of its antagonist in granulomatous disease remain unknown. METHODS Here, we report expression of GHRH receptor (R) in human tissue with sarcoidosis granuloma and demonstrate the anti-inflammatory effects of MIA602 (a GHRH antagonist) in two in vitro human granuloma models and an in vivo granuloma model using different methods including ELISA, immunohistochemistry, RNA-seq analysis and flow cytometry. RESULTS MIA602 decreases the levels of IL-2, IL-2R, IL-7, IL-12, IL-17A and TNF-α in an in vitro granuloma model. Further, we show that the anti-inflammatory effect of MIA602 appears to be mediated by a reduction in CD45+CD68+ cells in granulomatous tissue and upregulation in PD-1 expression in macrophages. Analysis of the expression of proteins involved in the mitochondrial stage of apoptosis showed that MIA602 increases the levels of caspase-3, BCL-xL/BAK dimer and MCl-1/Bak dimer in the granuloma. These findings indicate that MIA602 may not induce apoptosis. CONCLUSIONS Our findings further suggest that GHRH-R is potentially a clinical target for the treatment of granulomatous disease and that MIA602 may be used as a novel therapeutic agent for sarcoidosis.
Collapse
Affiliation(s)
- Chongxu Zhang
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
| | - Runxia Tian
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
| | | | | | - Gregory Holt
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
- Division of Pulmonary and Critical CareUniversity of MiamiMiamiFLUSA
| | - Renzhi Cai
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
| | - Anthony Griswold
- School of MedicineJohn P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFLUSA
| | | | - Robert Jackson
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
- School of MedicineUniversity of MiamiMiamiFLUSA
| | - Andrew V Schally
- Polypeptide and Cancer InstituteVeterans Affairs Medical CenterMiamiFLUSA
- Department of PathologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Mehdi Mirsaeidi
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
- Division of Pulmonary and Critical CareUniversity of MiamiMiamiFLUSA
| |
Collapse
|
16
|
Yu X, Zhou G, Shao B, Zhou H, Xu C, Yan F, Wang L, Chen G, Li J, Fu X. Gut Microbiota Dysbiosis Induced by Intracerebral Hemorrhage Aggravates Neuroinflammation in Mice. Front Microbiol 2021; 12:647304. [PMID: 34025607 PMCID: PMC8137318 DOI: 10.3389/fmicb.2021.647304] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/25/2021] [Indexed: 01/20/2023] Open
Abstract
Intracerebral hemorrhage (ICH) induces a strong hematoma-related neuroinflammatory reaction and alters peripheral immune homeostasis. Recent research has found that gut microbiota plays a role in neurodegeneration and autoimmune diseases by regulating immune homeostasis and neuroinflammation. Therefore, we investigated the relationship between ICH, microbiota alteration, and immune responses after hematoma-induced acute brain injury. In our study, we used a mouse model of ICH, and 16S ribosomal RNA sequencing showed that ICH causes gut microbiota dysbiosis, which in turn affects ICH outcome through immune-mediated mechanisms. There was prominent reduced species diversity and microbiota overgrowth in the dysbiosis induced by ICH, which may reduce intestinal motility and increase gut permeability. In addition, recolonizing ICH mice with a normal health microbiota ameliorates functional deficits and neuroinflammation after ICH. Meanwhile, cell-tracking studies have demonstrated the migration of intestinal lymphocytes to the brain after ICH. In addition, therapeutic transplantation of fecal microbiota improves intestinal barrier damage. These results support the conclusion that the gut microbiome is a target of ICH-induced systemic alteration and is considered to have a substantial impact on ICH outcome.
Collapse
Affiliation(s)
- Xiaobo Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guoyang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurosurgery, The First People's Hospital of Wenling, Wenling, China
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Park E, Lyon JG, Alvarado-Velez M, Betancur MI, Mokarram N, Shin JH, Bellamkonda RV. Enriching neural stem cell and anti-inflammatory glial phenotypes with electrical stimulation after traumatic brain injury in male rats. J Neurosci Res 2021; 99:1864-1884. [PMID: 33772860 PMCID: PMC8360147 DOI: 10.1002/jnr.24834] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) by an external physical impact results in compromised brain function via undesired neuronal death. Following the injury, resident and peripheral immune cells, astrocytes, and neural stem cells (NSCs) cooperatively contribute to the recovery of the neuronal function after TBI. However, excessive pro‐inflammatory responses of immune cells, and the disappearance of endogenous NSCs at the injury site during the acute phase of TBI, can exacerbate TBI progression leading to incomplete healing. Therefore, positive outcomes may depend on early interventions to control the injury‐associated cellular milieu in the early phase of injury. Here, we explore electrical stimulation (ES) of the injury site in a rodent model (male Sprague–Dawley rats) to investigate its overall effect on the constituent brain cell phenotype and composition during the acute phase of TBI. Our data showed that a brief ES for 1 hr on day 2 of TBI promoted anti‐inflammatory phenotypes of microglia as assessed by CD206 expression and increased the population of NSCs and Nestin+ astrocytes at 7 days post‐TBI. Also, ES effectively increased the number of viable neurons when compared to the unstimulated control group. Given the salience of microglia and neural stem cells for healing after TBI, our results strongly support the potential benefit of the therapeutic use of ES during the acute phase of TBI to regulate neuroinflammation and to enhance neuroregeneration.
Collapse
Affiliation(s)
- Eunyoung Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Melissa Alvarado-Velez
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha I Betancur
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Nassir Mokarram
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
18
|
Fernández A, Quintana E, Velasco P, Moreno-Jimenez B, de Andrés B, Gaspar ML, Liste I, Vilar M, Mira H, Cano E. Senescent accelerated prone 8 (SAMP8) mice as a model of age dependent neuroinflammation. J Neuroinflammation 2021; 18:75. [PMID: 33736657 PMCID: PMC7977588 DOI: 10.1186/s12974-021-02104-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aging and age-related diseases are strong risk factors for the development of neurodegenerative diseases. Neuroinflammation (NIF), as the brain's immune response, plays an important role in aged associated degeneration of central nervous system (CNS). There is a need for well characterized animal models that will allow the scientific community to understand and modulate this process. METHODS We have analyzed aging-phenotypical and inflammatory changes of brain myeloid cells (bMyC) in a senescent accelerated prone aged (SAMP8) mouse model, and compared with their senescence resistant control mice (SAMR1). We have performed morphometric methods to evaluate the architecture of cellular prolongations and determined the appearance of Iba1+ clustered cells with aging. To analyze specific constant brain areas, we have performed stereology measurements of Iba1+ cells in the hippocampal formation. We have isolated bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch), and analyzed their response to systemic lipopolysaccharide (LPS)-driven inflammation. RESULTS Aged 10 months old SAMP8 mice present many of the hallmarks of aging-dependent neuroinflammation when compared with their SAMR1 control, i.e., increase of protein aggregates, presence of Iba1+ clusters, but not an increase in the number of Iba1+ cells. We have further observed an increase of main inflammatory mediator IL-1β, and an augment of border MHCII+Iba1+ cells. Isolated CD45+ bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch) have been analyzed, showing that there is not a significant increase of CD45+ cells from the periphery. Our data support that aged-driven pro-inflammatory cytokine interleukin 1 beta (IL-1β) transcription is enhanced in CD45+BP cells. Furthermore, LPS-driven systemic inflammation produces inflammatory cytokines mainly in border bMyC, sensed to a lesser extent by the BP bMyC, showing that IL-1β expression is further augmented in aged SAMP8 compared to control SAMR1. CONCLUSION Our data validate the SAMP8 model to study age-associated neuroinflammatory events, but careful controls for age and strain are required. These animals show morphological changes in their bMyC cell repertoires associated to age, corresponding to an increase in the production of pro-inflammatory cytokines such as IL-1β, which predispose the brain to an enhanced inflammatory response after LPS-systemic challenge.
Collapse
Affiliation(s)
- Andrés Fernández
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Elena Quintana
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Patricia Velasco
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Belén Moreno-Jimenez
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Belén de Andrés
- Unidad de Inmunobiología, Instituto de Salud Carlos II, Madrid, Spain
| | | | - Isabel Liste
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Marçal Vilar
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eva Cano
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain.
| |
Collapse
|
19
|
Zhang L, Graf I, Kuang Y, Zheng X, Haupt M, Majid A, Kilic E, Hermann DM, Psychogios MN, Weber MS, Ochs J, Bähr M, Doeppner TR. Neural Progenitor Cell-Derived Extracellular Vesicles Enhance Blood-Brain Barrier Integrity by NF-κB (Nuclear Factor-κB)-Dependent Regulation of ABCB1 (ATP-Binding Cassette Transporter B1) in Stroke Mice. Arterioscler Thromb Vasc Biol 2020; 41:1127-1145. [PMID: 33327747 PMCID: PMC7901534 DOI: 10.1161/atvbaha.120.315031] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: Extracellular vesicles (EVs) derived from neural progenitor cells enhance poststroke neurological recovery, albeit the underlying mechanisms remain elusive. Since previous research described an enhanced poststroke integrity of the blood-brain barrier (BBB) upon systemic transplantation of neural progenitor cells, we examined if neural progenitor cell-derived EVs affect BBB integrity and which cellular mechanisms are involved in the process. Approach and Results: Using in vitro models of primary brain endothelial cell (EC) cultures as well as co-cultures of brain ECs (ECs) and astrocytes exposed to oxygen glucose deprivation, we examined the effects of EVs or vehicle on microvascular integrity. In vitro data were confirmed using a mouse transient middle cerebral artery occlusion model. Cultured ECs displayed increased ABCB1 (ATP-binding cassette transporter B1) levels when exposed to oxygen glucose deprivation, which was reversed by treatment with EVs. The latter was due to an EV-induced inhibition of the NF-κB (nuclear factor-κB) pathway. Using a BBB co-culture model of ECs and astrocytes exposed to oxygen glucose deprivation, EVs stabilized the BBB and ABCB1 levels without affecting the transcellular electrical resistance of ECs. Likewise, EVs yielded reduced Evans blue extravasation, decreased ABCB1 expression as well as an inhibition of the NF-κB pathway, and downstream matrix metalloproteinase 9 (MMP-9) activity in stroke mice. The EV-induced inhibition of the NF-κB pathway resulted in a poststroke modulation of immune responses. Conclusions: Our findings suggest that EVs enhance poststroke BBB integrity via ABCB1 and MMP-9 regulation, attenuating inflammatory cell recruitment by inhibition of the NF-κB pathway.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany
| | - Irina Graf
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany
| | - Yaoyun Kuang
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany
| | - Xuan Zheng
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany
| | - Matteo Haupt
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (A.M.)
| | - Ertugrul Kilic
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Turkey (E.K., T.R.D.)
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (D.M.H.)
| | | | - Martin S Weber
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany.,Department of Neuropathology (M.S.W., J.O.), University Medical Center Göttingen, Germany
| | - Jasmin Ochs
- Department of Neuropathology (M.S.W., J.O.), University Medical Center Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany
| | - Thorsten R Doeppner
- Department of Neurology (L.Z., I.G., Y.K., X.Z., M.H., M.S.W., M.B., T.R.D.), University Medical Center Göttingen, Germany.,Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Turkey (E.K., T.R.D.)
| |
Collapse
|
20
|
Enam SF, Kader SR, Bodkin N, Lyon JG, Calhoun M, Azrak C, Tiwari PM, Vanover D, Wang H, Santangelo PJ, Bellamkonda RV. Evaluation of M2-like macrophage enrichment after diffuse traumatic brain injury through transient interleukin-4 expression from engineered mesenchymal stromal cells. J Neuroinflammation 2020; 17:197. [PMID: 32563258 PMCID: PMC7306141 DOI: 10.1186/s12974-020-01860-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/29/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Appropriately modulating inflammation after traumatic brain injury (TBI) may prevent disabilities for the millions of those inflicted annually. In TBI, cellular mediators of inflammation, including macrophages and microglia, possess a range of phenotypes relevant for an immunomodulatory therapeutic approach. It is thought that early phenotypic modulation of these cells will have a cascading healing effect. In fact, an anti-inflammatory, "M2-like" macrophage phenotype after TBI has been associated with neurogenesis, axonal regeneration, and improved white matter integrity (WMI). There already exist clinical trials seeking an M2-like bias through mesenchymal stem/stromal cells (MSCs). However, MSCs do not endogenously synthesize key signals that induce robust M2-like phenotypes such as interleukin-4 (IL-4). METHODS To enrich M2-like macrophages in a clinically relevant manner, we augmented MSCs with synthetic IL-4 mRNA to transiently express IL-4. These IL-4 expressing MSCs (IL-4 MSCs) were characterized for expression and functionality and then delivered in a modified mouse TBI model of closed head injury. Groups were assessed for functional deficits and MR imaging. Brain tissue was analyzed through flow cytometry, multi-plex ELISA, qPCR, histology, and RNA sequencing. RESULTS We observed that IL-4 MSCs indeed induce a robust M2-like macrophage phenotype and promote anti-inflammatory gene expression after TBI. However, here we demonstrate that acute enrichment of M2-like macrophages did not translate to improved functional or histological outcomes, or improvements in WMI on MR imaging. To further understand whether dysfunctional pathways underlie the lack of therapeutic effect, we report transcriptomic analysis of injured and treated brains. Through this, we discovered that inflammation persists despite acute enrichment of M2-like macrophages in the brain. CONCLUSION The results demonstrate that MSCs can be engineered to induce a stronger M2-like macrophage response in vivo. However, they also suggest that acute enrichment of only M2-like macrophages after diffuse TBI cannot orchestrate neurogenesis, axonal regeneration, or improve WMI. Here, we also discuss our modified TBI model and methods to assess severity, behavioral studies, and propose that IL-4 expressing MSCs may also have relevance in other cavitary diseases or in improving biomaterial integration into tissues.
Collapse
Affiliation(s)
- Syed Faaiz Enam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Nicholas Bodkin
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mark Calhoun
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Cesar Azrak
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Pooja Munnilal Tiwari
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Daryll Vanover
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Haichen Wang
- Department of Neurology, Duke University, Durham, NC, USA
| | - Philip J Santangelo
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | |
Collapse
|
21
|
Li M, Meng N, Guo X, Niu X, Zhao Z, Wang W, Xie X, Lv P. Dl-3-n-Butylphthalide Promotes Remyelination and Suppresses Inflammation by Regulating AMPK/SIRT1 and STAT3/NF-κB Signaling in Chronic Cerebral Hypoperfusion. Front Aging Neurosci 2020; 12:137. [PMID: 32581761 PMCID: PMC7296049 DOI: 10.3389/fnagi.2020.00137] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Demyelination in vascular dementia (VD) is partly attributable to inflammation induced by chronic cerebral hypoperfusion (CCH). Remyelination contributes to the recovery of cognitive impairment by inducing the proliferation and differentiation of oligodendrocyte progenitor cells. It was previously reported that Dl-3-n-butylphthalide (NBP) promotes cognitive improvement. However, whether NBP can stimulate remyelination and suppress inflammation after CCH remains unclear. To answer this question, the present study investigated the effects of NBP on remyelination in a rat model of CCH established by bilateral carotid artery occlusion. Functional recovery was evaluated with the Morris water maze (MWM) test, and myelin integrity, regeneration of mature oligodendrocytes, and inhibition of astrocyte proliferation were assessed by immunohistochemistry and histologic analysis. Additionally, activation of 5′ AMP-activated protein kinase (AMPK)/Sirtuin (SIRT)1 and Signal transducer and activator of transcription (STAT)3/nuclear factor (NF)-κB signaling pathways was evaluated by western blotting. The results showed that NBP treatment improved memory and learning performance in CCH rats, which was accompanied by increased myelin integrity and oligodendrocyte regeneration, and reduced astrocyte proliferation and inflammation. Additionally, NBP induced the activation of AMPK/SIRT1 signaling while inhibiting the STAT3/NF-κB pathway. These results indicate that NBP alleviates cognitive impairment following CCH by promoting remyelination and suppressing inflammation via modulation of AMPK/SIRT1 and STAT3/NF-κB signaling.
Collapse
Affiliation(s)
- Meixi Li
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Nan Meng
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Xin Guo
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiaoli Niu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhongmin Zhao
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Wei Wang
- Department of Neurology, Hebei Medical University, Shijiazhuang, China
| | - Xiaohua Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
22
|
Peng L, Wang Y, Fei S, Wei C, Tong F, Wu G, Ma H, Dong X. The effect of combining Endostar with radiotherapy on blood vessels, tumor-associated macrophages, and T cells in brain metastases of Lewis lung cancer. Transl Lung Cancer Res 2020; 9:745-760. [PMID: 32676336 PMCID: PMC7354151 DOI: 10.21037/tlcr-20-500] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Combining Endostar (ES) with radiotherapy (RT) has shown a promising therapeutic effect on non-small cell lung carcinoma with brain metastases (BMs) in clinical practice. However, the specific mechanism is not yet fully understood. The present study aimed to investigate the effects of ES on blood vessels, tumor-associated macrophages (TAMs), and T cells in a tumor microenvironment treated with RT. Methods BM models were established by stereotactic and intracarotid injection of luciferase-Lewis lung cancer (LLC) cells into female C57BL mice. The animals were randomly divided into 4 groups: normal saline (NS), ES, RT, and ES plus radiotherapy (ES + RT) groups. Tumor size was determined with the IVIS imaging system. Tumor specimens were stained with CD34 and α-SMA to investigate tumor vascular changes. The proportions of TAMs, CD4+ T cells, and CD8+ T cells in tumor tissues were determined by flow cytometry and immunofluorescence. The expressions of hypoxia-inducible factor 1α (HIF-1α) and CXCR4 were deduced using western blotting and immunohistochemistry (IHC). Results ES + RT significantly suppressed tumor growth compared to the other 3 groups. RT decreased M1 and increased M2 in microglial cells and bone marrow-derived macrophages (BMDMs) relative to NS, while ES had the opposite effect. The ratio of CD8+T/CD4+T was increased in the ES + RT group compared to the other 3 groups. Tumor vascular maturity (α-SMA+/CD34+) was increased while HIF-1α was significantly suppressed in the ES + RT group. CXCR4 expression, which is involved in TAM recruitment, increased following RT, whereas, ES attenuated its expression. Conclusions Our findings suggest that ES can promote the normalization of tumor blood vessels and increase the anti-tumor immune-related immune cells infiltrating the tumor following RT treatment.
Collapse
Affiliation(s)
- Ling Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ying Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shihong Fei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunhua Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fan Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
23
|
TRIM32 Drives Pathogenesis in Streptococcal Toxic Shock-Like Syndrome and Streptococcus suis Meningitis by Regulating Innate Immune Responses. Infect Immun 2020; 88:IAI.00957-19. [PMID: 31988176 DOI: 10.1128/iai.00957-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Streptococcus suis is an emerging zoonotic agent that causes streptococcal toxic shock-like syndrome (STSLS) and meningitis in humans, with high mortality and morbidity. The pathogenesis of both STSLS and central nervous system (CNS) infections caused by S. suis is not well understood. TRIM32, a member of the tripartite motif (TRIM) protein family, has been reported to regulate host inflammatory responses. In this study, we showed that TRIM32 deficiency significantly reduced the level of bacteremia and the production of proinflammatory cytokines following severe S. suis infection, protecting infected mice from STSLS. The influence of TRIM32 gene deletion on a range of processes known to be involved in S. suis meningitis was also examined. Both levels of bacterial loads and indications of brain hemorrhage were reduced in infected Trim32- / - mice compared with infected wild-type (WT) controls. We also found that TRIM32 deficiency increased the permeability of the blood-brain barrier (BBB) and the recruitment of inflammatory monocytes during the early course of S. suis infection, potentially limiting the development of S. suis meningitis. Our results suggest that TRIM32 sensitizes S. suis-induced infection via innate immune response regulation.
Collapse
|
24
|
Amarilla AA, Santos-Junior NN, Figueiredo ML, Luiz JPM, Fumagalli MJ, Colón DF, Lippi V, Alfonso HL, Lima-Junior DS, Trabuco AC, Spinieli RL, Desidera AC, Leite-Panissi CRA, Lauretti F, Mendoza SES, Silva CLA, Rego EM, Galvao-Lima LJ, Bassi GS, Penharvel Martíns SLB, Manrique WG, Alves-Filho JC, Cunha FQ, Peng NYG, Modhiran N, Setoh YX, Khromykh AA, Figueiredo LTM, Aquino VH. CCR2 Plays a Protective Role in Rocio Virus-Induced Encephalitis by Promoting Macrophage Infiltration Into the Brain. J Infect Dis 2020; 219:2015-2025. [PMID: 30715407 PMCID: PMC7107438 DOI: 10.1093/infdis/jiz029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/24/2019] [Indexed: 12/15/2022] Open
Abstract
Rocio virus (ROCV) is a highly neuropathogenic mosquito-transmitted flavivirus responsible for an unprecedented outbreak of human encephalitis during 1975–1976 in Sao Paulo State, Brazil. Previous studies have shown an increased number of inflammatory macrophages in the central nervous system (CNS) of ROCV-infected mice, implying a role for macrophages in the pathogenesis of ROCV. Here, we show that ROCV infection results in increased expression of CCL2 in the blood and in infiltration of macrophages into the brain. Moreover, we show, using CCR2 knockout mice, that CCR2 expression is essential for macrophage infiltration in the brain during ROCV infection and that the lack of CCR2 results in increased disease severity and mortality. Thus, our findings show the protective role of CCR2-mediated infiltration of macrophages in the brain during ROCV infection.
Collapse
Affiliation(s)
- Alberto A Amarilla
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil.,Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | | | - Mario Luis Figueiredo
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Joao Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | | | - David F Colón
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Veronica Lippi
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Helda Liz Alfonso
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Djalma S Lima-Junior
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, Ribeirao Preto, SP, Brazil
| | - Amanda C Trabuco
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Richard L Spinieli
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Amanda C Desidera
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Christie R A Leite-Panissi
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Silvia Elena Sánchez Mendoza
- Division of Hematology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil.,Division of Clinical Oncology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil
| | | | - Eduardo Magalhaes Rego
- Division of Hematology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil.,Division of Clinical Oncology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil
| | - Leonardo J Galvao-Lima
- Department of Immunology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Gabriel S Bassi
- Department of Pharmacology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sandra L B Penharvel Martíns
- Department of Surgery and Anatomy, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Wilson Gomez Manrique
- Animal Health Laboratory, Veterinary Medicine Course, Federal University of Rondonia - UNIR, Rolim de Moura, RO, Brazil
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Nias Y G Peng
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Naphak Modhiran
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Yin Xiang Setoh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Alexander A Khromykh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Luiz T M Figueiredo
- Virology Research Center, Ribeirao Preto, SP, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Victor H Aquino
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
25
|
Inflammatory Monocytes and Neutrophils Regulate Streptococcus suis-Induced Systemic Inflammation and Disease but Are Not Critical for the Development of Central Nervous System Disease in a Mouse Model of Infection. Infect Immun 2020; 88:IAI.00787-19. [PMID: 31818962 PMCID: PMC7035915 DOI: 10.1128/iai.00787-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
Streptococcus suis is an important porcine bacterial pathogen and zoonotic agent responsible for sudden death, septic shock, and meningitis. These pathologies are a consequence of elevated bacterial replication leading to exacerbated and uncontrolled inflammation, a hallmark of the S. suis systemic and central nervous system (CNS) infections. Monocytes and neutrophils are immune cells involved in various functions, including proinflammatory mediator production. Streptococcus suis is an important porcine bacterial pathogen and zoonotic agent responsible for sudden death, septic shock, and meningitis. These pathologies are a consequence of elevated bacterial replication leading to exacerbated and uncontrolled inflammation, a hallmark of the S. suis systemic and central nervous system (CNS) infections. Monocytes and neutrophils are immune cells involved in various functions, including proinflammatory mediator production. Moreover, monocytes are composed of two main subsets: shorter-lived inflammatory monocytes and longer-lived patrolling monocytes. However, regardless of their presence in blood and the fact that S. suis-induced meningitis is characterized by infiltration of monocytes and neutrophils into the CNS, their role during the S. suis systemic and CNS diseases remains unknown. Consequently, we hypothesized that monocytes and neutrophils participate in S. suis infection via bacterial clearance and inflammation. Results demonstrated that inflammatory monocytes and neutrophils regulate S. suis-induced systemic disease via their role in inflammation required for bacterial burden control. In the CNS, inflammatory monocytes contributed to exacerbation of S. suis-induced local inflammation, while neutrophils participated in bacterial burden control. However, development of clinical CNS disease was independent of both cell types, indicating that resident immune cells are mostly responsible for S. suis-induced CNS inflammation and clinical disease and that inflammatory monocyte and neutrophil infiltration is a consequence of the induced inflammation. In contrast, the implication of patrolling monocytes was minimal throughout the S. suis infection. Consequently, this study demonstrates that while inflammatory monocytes and neutrophils modulate S. suis-induced systemic inflammation and disease, they are not critical for CNS disease development.
Collapse
|
26
|
Selective targeting of nanomedicine to inflamed cerebral vasculature to enhance the blood-brain barrier. Proc Natl Acad Sci U S A 2020; 117:3405-3414. [PMID: 32005712 DOI: 10.1073/pnas.1912012117] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug targeting to inflammatory brain pathologies such as stroke and traumatic brain injury remains an elusive goal. Using a mouse model of acute brain inflammation induced by local tumor necrosis factor alpha (TNFα), we found that uptake of intravenously injected antibody to vascular cell adhesion molecule 1 (anti-VCAM) in the inflamed brain is >10-fold greater than antibodies to transferrin receptor-1 and intercellular adhesion molecule 1 (TfR-1 and ICAM-1). Furthermore, uptake of anti-VCAM/liposomes exceeded that of anti-TfR and anti-ICAM counterparts by ∼27- and ∼8-fold, respectively, achieving brain/blood ratio >300-fold higher than that of immunoglobulin G/liposomes. Single-photon emission computed tomography imaging affirmed specific anti-VCAM/liposome targeting to inflamed brain in mice. Intravital microscopy via cranial window and flow cytometry showed that in the inflamed brain anti-VCAM/liposomes bind to endothelium, not to leukocytes. Anti-VCAM/LNP selectively accumulated in the inflamed brain, providing de novo expression of proteins encoded by cargo messenger RNA (mRNA). Anti-VCAM/LNP-mRNA mediated expression of thrombomodulin (a natural endothelial inhibitor of thrombosis, inflammation, and vascular leakage) and alleviated TNFα-induced brain edema. Thus VCAM-directed nanocarriers provide a platform for cerebrovascular targeting to inflamed brain, with the goal of normalizing the integrity of the blood-brain barrier, thus benefiting numerous brain pathologies.
Collapse
|
27
|
Calvo B, Rubio F, Fernández M, Tranque P. Dissociation of neonatal and adult mice brain for simultaneous analysis of microglia, astrocytes and infiltrating lymphocytes by flow cytometry. IBRO Rep 2020; 8:36-47. [PMID: 32215337 PMCID: PMC7090101 DOI: 10.1016/j.ibror.2019.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022] Open
Abstract
Recovery of neural cells is higher with 30 % Percoll gradient than 30–70 %. Papain enhances combined extraction of microglia, astrocytes and lymphocytes. Dispase II potentiates papain action only in adult brain. Mechanical dissociation isolates neonatal and adult astrocytes better than enzymes. Papain + dispase II alows cell cytometry quantification of glial activation by LPS.
The technical difficulty to isolate microglia, astrocytes and infiltrating immune cells from mouse brain is nowadays a limiting factor in the study of neuroinflammation. Brain isolation requirements are cell-type and animal-age dependent, but current brain dissociation procedures are poorly standardized. This lack of comprehensive studies hampers the selection of optimized methodologies. Thus, we present here a comparative analysis of dissociation methods and Percoll-based separation to identify the most efficient procedure for the combined isolation of healthy microglia, astrocytes and infiltrated leukocytes; distinguishing neonatal and adult mouse brain. Gentle mechanical dissociation and DNase I incubation was supplemented with papain or collagenase II. Dispase II digestion was also used alone or in combination. In addition, cell separation efficiency of 30 % and 30–70 % Percoll gradients was compared. In these experiments, cell yield and integrity of freshly dissociated cells was measured by flow cytometry. We found that papain digestion in combination with dispase II followed by 30 % Percoll separation is the most balanced method to obtain a mixture of microglia, astrocytes and infiltrated immune cells; while addition of dispase II was not an advantage for neonatal brain. These dissociation conditions allowed flow cytometry detection of a slight glial activation triggered by sublethal LPS injection. In conclusion, the enzymes and Percoll density gradients tested here affected differently resting microglia, activated microglia/macrophages, astrocytes and infiltrated lymphocytes. Also, newborn and adult brain showed contrasting reactions to digestion. Our study highlights the strength of flow cytometry for the simultaneous analysis of neuroimmune cell populations once extraction is optimized.
Collapse
Key Words
- ANOVA, one-way analysis of variance
- Astrocytes
- CNS, Central Nervous System
- CaCl2, calcium chloride
- EBSS, Earle's Balanced Salt Solution
- EDTA, ethylenediaminetetraacetic acid
- FACS, Fluorescence-activated cell sorter
- FSC, forward-scattered light
- Flow cytometry
- Glia reactivity
- HBSS, Hank's Balanced Salt Solution
- LD, lethal dose
- LPS, lipopolysaccharide
- Lymphocytes
- MgCl2, magnesium chloride
- MgSO4, magnesium sulfate
- Microglia
- Neuroimmunity
- PBS, phosphate-buffered saline
- RT, room temperature
- SIP, stock solution of isotonic Percoll
- SSC, side-scattered light
- i.p, intraperitoneal injection
Collapse
Affiliation(s)
- Belén Calvo
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Felipe Rubio
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Miriam Fernández
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Pedro Tranque
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| |
Collapse
|
28
|
Joy MT, Ben Assayag E, Shabashov-Stone D, Liraz-Zaltsman S, Mazzitelli J, Arenas M, Abduljawad N, Kliper E, Korczyn AD, Thareja NS, Kesner EL, Zhou M, Huang S, Silva TK, Katz N, Bornstein NM, Silva AJ, Shohami E, Carmichael ST. CCR5 Is a Therapeutic Target for Recovery after Stroke and Traumatic Brain Injury. Cell 2020; 176:1143-1157.e13. [PMID: 30794775 DOI: 10.1016/j.cell.2019.01.044] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 10/05/2018] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.
Collapse
Affiliation(s)
- Mary T Joy
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Einor Ben Assayag
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dalia Shabashov-Stone
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sigal Liraz-Zaltsman
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel; The Joseph Sagol Neuroscience Center, Sheba Medical Center, Israel; Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono, Israel
| | - Jose Mazzitelli
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Marcela Arenas
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Nora Abduljawad
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Efrat Kliper
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Amos D Korczyn
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nikita S Thareja
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Efrat L Kesner
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miou Zhou
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Shan Huang
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Tawnie K Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Noomi Katz
- Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono, Israel
| | - Natan M Bornstein
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Esther Shohami
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
29
|
Morimoto K, Nakajima K. Role of the Immune System in the Development of the Central Nervous System. Front Neurosci 2019; 13:916. [PMID: 31551681 PMCID: PMC6735264 DOI: 10.3389/fnins.2019.00916] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/16/2019] [Indexed: 01/01/2023] Open
Abstract
The central nervous system (CNS) and the immune system are both intricate and highly organized systems that regulate the entire body, with both sharing certain common features in developmental mechanisms and operational modes. It is known that innate immunity-related molecules, such as cytokines, toll-like receptors, the complement family, and acquired immunity-related molecules, such as the major histocompatibility complex and antibody receptors, are also expressed in the brain and play important roles in brain development. Moreover, although the brain has previously been regarded as an immune-privileged site, it is known to contain lymphatic vessels. Not only microglia but also lymphocytes regulate cognition and play a vital role in the formation of neuronal circuits. This review provides an overview of the function of immune cells and immune molecules in the CNS, with particular emphasis on their effect on neural developmental processes.
Collapse
Affiliation(s)
- Keiko Morimoto
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Zhou S, Liu G, Guo J, Kong F, Chen S, Wang Z. Pro-inflammatory Effect of Downregulated CD73 Expression in EAE Astrocytes. Front Cell Neurosci 2019; 13:233. [PMID: 31191254 PMCID: PMC6549520 DOI: 10.3389/fncel.2019.00233] [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: 11/07/2018] [Accepted: 05/09/2019] [Indexed: 01/31/2023] Open
Abstract
CD73, an ectonucleotidase, participates in the regulation of immune responses by controlling the conversion of extracellular AMP to adenosine. In this study, we investigated whether any type of brain cells, especially neuroglia cells, exhibit altered CD73 expression, localization or activity upon experimental autoimmune uveitis (EAU) induction and whether altered CD73 manipulates the activation of effector T cells that interact with such cell types. First, the amount of cell membrane-exposed CD73 was detected by flow cytometry in various types of brain cells collected from either naïve or EAE mice. Compared to that in astrocytes from naïve control mice, the amount of membrane-bound CD73 was significantly decreased in astrocytes from EAE mice, while no significant differences were detected in other cell types. Thereafter, wild-type and CD73-/- astrocytes were used to study whether CD73 influences the function of inflammatory astrocytes, such as the production of cytokines/chemokines and the activation of effector T cells that interact with astrocytes. The results indicated that the addition of exogenous AMP significantly inhibited cytokine/chemokine production by wild type astrocytes but had no effect on CD73-/- astrocytes and that the effect of AMP was almost completely blocked by the addition of either a CD73 inhibitor (APCP) or an adenosine receptor A1 subtype (ARA1) antagonist (DPCPX). Although the addition of AMP did not affect CD73-/- astrocytes, the addition of adenosine successfully inhibited their cytokine/chemokine production. The antigen-specific interaction of astrocytes with invading CD4 cells caused CD73 downregulation in astrocytes from mice that underwent EAE induction. Collectively, our findings support the conclusion that, upon EAE induction, likely due to an interaction with invading CD4+ cells, astrocytes lose most of their membrane-localized CD73; this inhibits the generation of adenosine in the local microenvironment. As adenosine has anti-inflammatory effects on astrocytes and CNS-infiltrating effector T cells in EAE, the downregulation of CD73 in astrocytes may be considered a pro-inflammatory process for facilitating the pathogenesis of EAE.
Collapse
Affiliation(s)
- Shumin Zhou
- Clinical Laboratory, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Guoping Liu
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| | - Jie Guo
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| | - Fanqiang Kong
- Clinical Laboratory, General Hospital of Tianjin Medical University, Tianjin, China
| | - Song Chen
- Department of Ophthalmology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
31
|
Inhibition of MALT1 Decreases Neuroinflammation and Pathogenicity of Virulent Rabies Virus in Mice. J Virol 2018; 92:JVI.00720-18. [PMID: 30158289 DOI: 10.1128/jvi.00720-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/05/2018] [Indexed: 12/15/2022] Open
Abstract
Rabies virus is a neurovirulent RNA virus, which causes about 59,000 human deaths each year. Treatment for rabies does not exist due to incomplete understanding of the pathogenesis. MALT1 mediates activation of several immune cell types and is involved in the proliferation and survival of cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, leading to the expression of immunoregulatory genes. Here, we examined the impact of genetic or pharmacological MALT1 inhibition in mice on disease development after infection with the virulent rabies virus strain CVS-11. Morbidity and mortality were significantly delayed in Malt1 -/- compared to Malt1 +/+ mice, and this effect was associated with lower viral load, proinflammatory gene expression, and infiltration and activation of immune cells in the brain. Specific deletion of Malt1 in T cells also delayed disease development, while deletion in myeloid cells, neuronal cells, or NK cells had no effect. Disease development was also delayed in mice treated with the MALT1 protease inhibitor mepazine and in knock-in mice expressing a catalytically inactive MALT1 mutant protein, showing an important role of MALT1 proteolytic activity. The described protective effect of MALT1 inhibition against infection with a virulent rabies virus is the precise opposite of the sensitizing effect of MALT1 inhibition that we previously observed in the case of infection with an attenuated rabies virus strain. Together, these data demonstrate that the role of immunoregulatory responses in rabies pathogenicity is dependent on virus virulence and reveal the potential of MALT1 inhibition for therapeutic intervention.IMPORTANCE Rabies virus is a neurotropic RNA virus that causes encephalitis and still poses an enormous challenge to animal and public health. Efforts to establish reliable therapeutic strategies have been unsuccessful and are hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protease that mediates the activation of several innate and adaptive immune cells in response to multiple receptors, and therapeutic MALT1 targeting is believed to be a valid approach for autoimmunity and MALT1-addicted cancers. Here, we study the impact of MALT1 deficiency on brain inflammation and disease development in response to infection of mice with the highly virulent CVS-11 rabies virus. We demonstrate that pharmacological or genetic MALT1 inhibition decreases neuroinflammation and extends the survival of CVS-11-infected mice, providing new insights in the biology of MALT1 and rabies virus infection.
Collapse
|
32
|
Zhang Y, Zhang H, Ma W, Liu K, Zhao M, Zhao Y, Lu X, Zhang F, Li X, Gao GF, Liu WJ. Evaluation of Zika Virus-specific T-cell Responses in Immunoprivileged Organs of Infected Ifnar1-/- Mice. J Vis Exp 2018:58110. [PMID: 30394402 PMCID: PMC6235543 DOI: 10.3791/58110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Zika virus (ZIKV) can induce inflammation in immunoprivileged organs (e.g., the brain and testis), leading to the Guillain-Barré syndrome and damaging the testes. During an infection with the ZIKV, immune cells have been shown to infiltrate into the tissues. However, the cellular mechanisms that define the protection and/or immunopathogenesis of these immune cells during a ZIKV infection are still largely unknown. Herein, we describe methods to evaluate the virus-specific T-cell functionality in these immunoprivileged organs of ZIKV-infected mice. These methods include a) a ZIKV infection and vaccine inoculation in Ifnar1-/- mice; b) histopathology, immunofluorescence, and immunohistochemistry assays to detect the virus infection and inflammation in the brain, testes, and spleen; c) the preparation of a tetramer of ZIKV-derived T-cell epitopes; d) the detection of ZIKV-specific T cells in the monocytes isolated from the brain, testes, and spleen. Using these approaches, it is possible to detect the antigen-specific T cells that have infiltrated into the immunoprivileged organs and to evaluate the functions of these T cells during the infection: potential immune protection via virus clearance and/or immunopathogenesis to exacerbate the inflammation. These findings may also help to clarify the contribution of T cells induced by the immunization against ZIKV.
Collapse
Affiliation(s)
- Yongli Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention
| | - Hangjie Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention
| | - Wenqiang Ma
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University
| | - Kefang Liu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention
| | - Min Zhao
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences
| | - Yingze Zhao
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention
| | - Xuancheng Lu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention
| | - Fuping Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences
| | - Xiangdong Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University;
| | - George F Gao
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention; Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences;
| | - William J Liu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention;
| |
Collapse
|
33
|
MALT1 Controls Attenuated Rabies Virus by Inducing Early Inflammation and T Cell Activation in the Brain. J Virol 2018; 92:JVI.02029-17. [PMID: 29367251 PMCID: PMC5874405 DOI: 10.1128/jvi.02029-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022] Open
Abstract
MALT1 is involved in the activation of immune responses, as well as in the proliferation and survival of certain cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, further promoting the expression of immunoregulatory genes. Deregulated MALT1 activity has been associated with autoimmunity and cancer, implicating MALT1 as a new therapeutic target. Although MALT1 deficiency has been shown to protect against experimental autoimmune encephalomyelitis, nothing is known about the impact of MALT1 on virus infection in the central nervous system. Here, we studied infection with an attenuated rabies virus, Evelyn-Rotnycki-Abelseth (ERA) virus, and observed increased susceptibility with ERA virus in MALT1−/− mice. Indeed, after intranasal infection with ERA virus, wild-type mice developed mild transient clinical signs with recovery at 35 days postinoculation (dpi). Interestingly, MALT1−/− mice developed severe disease requiring euthanasia at around 17 dpi. A decreased induction of inflammatory gene expression and cell infiltration and activation was observed in MALT1−/− mice at 10 dpi compared to MALT1+/+ infected mice. At 17 dpi, however, the level of inflammatory cell activation was comparable to that observed in MALT1+/+ mice. Moreover, MALT1−/− mice failed to produce virus-neutralizing antibodies. Similar results were obtained with specific inactivation of MALT1 in T cells. Finally, treatment of wild-type mice with mepazine, a MALT1 protease inhibitor, also led to mortality upon ERA virus infection. These data emphasize the importance of early inflammation and activation of T cells through MALT1 for controlling the virulence of an attenuated rabies virus in the brain. IMPORTANCE Rabies virus is a neurotropic virus which can infect any mammal. Annually, 59,000 people die from rabies. Effective therapy is lacking and hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protein involved in innate and adaptive immunity and is an interesting therapeutic target because MALT1-deregulated activity has been associated with autoimmunity and cancers. The role of MALT1 in viral infection is, however, largely unknown. Here, we study the impact of MALT1 on virus infection in the brain, using the attenuated ERA rabies virus in different models of MALT1-deficient mice. We reveal the importance of MALT1-mediated inflammation and T cell activation to control ERA virus, providing new insights in the biology of MALT1 and rabies virus infection.
Collapse
|
34
|
Good ME, Eucker SA, Li J, Bacon HM, Lang SM, Butcher JT, Johnson TJ, Gaykema RP, Patel MK, Zuo Z, Isakson BE. Endothelial cell Pannexin1 modulates severity of ischemic stroke by regulating cerebral inflammation and myogenic tone. JCI Insight 2018; 3:96272. [PMID: 29563335 PMCID: PMC5926909 DOI: 10.1172/jci.insight.96272] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/16/2018] [Indexed: 12/24/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality in the US; however, there currently exists only one effective acute pharmacological therapeutic intervention. Purinergic signaling has been shown to regulate vascular function and pathological processes, including inflammation and arterial myogenic reactivity, and plays a role in ischemic stroke outcome. Purinergic signaling requires extracellular purines; however, the mechanism of purine release from cells is unclear. Pannexin1 (Panx1) channels are potentially novel purine release channels expressed throughout the vascular tree that couples regulated purine release with purinergic signaling. Therefore, we examined the role of smooth muscle and endothelial cell Panx1, using conditional cell type-specific transgenic mice, in cerebral ischemia/reperfusion injury outcomes. Deletion of endothelial cell Panx1, but not smooth muscle cell Panx1, significantly reduced cerebral infarct volume after ischemia/reperfusion. Infiltration of leukocytes into brain tissue and development of cerebral myogenic tone were both significantly reduced when mice lacked endothelial Panx1. Panx1 regulation of myogenic tone was unique to the cerebral circulation, as mesenteric myogenic reactivity and blood pressure were independent of endothelial Panx1. Overall, deletion of endothelial Panx1 mitigated cerebral ischemic injury by reducing inflammation and myogenic tone development, indicating that endothelial Panx1 is a possible novel target for therapeutic intervention of ischemic stroke.
Collapse
Affiliation(s)
- Miranda E Good
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Stephanie A. Eucker
- Division of Emergency Medicine, Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Jun Li
- Department of Anesthesiology and
| | - Hannah M. Bacon
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Susan M. Lang
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Joshua T. Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Tyler J. Johnson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | | | | | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
35
|
Lasarte-Cia A, Lozano T, Pérez-González M, Gorraiz M, Iribarren K, Hervás-Stubbs S, Sarobe P, Rabal O, Cuadrado-Tejedor M, García-Osta A, Casares N, Lasarte JJ. Immunomodulatory Properties of Carvone Inhalation and Its Effects on Contextual Fear Memory in Mice. Front Immunol 2018; 9:68. [PMID: 29422905 PMCID: PMC5788902 DOI: 10.3389/fimmu.2018.00068] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/10/2018] [Indexed: 12/15/2022] Open
Abstract
A complex network of interactions exists between the immune, the olfactory, and the central nervous system (CNS). Inhalation of different fragrances can affect immunological reactions in response to an antigen but also may have effects on the CNS and cognitive activity. We performed an exploratory study of the immunomodulatory ability of a series of compounds representing each of the 10 odor categories or clusters described previously. We evaluated the impact of each particular odor on the immune response after immunization with the model antigen ovalbumin in combination with the TLR3 agonist poly I:C. We found that some odors behave as immunostimulatory agents, whereas others might be considered as potential immunosuppressant odors. Interestingly, the immunomodulatory capacity was, in some cases, strain-specific. In particular, one of the fragrances, carvone, was found to be immunostimulatory in BALB/c mice and immunosuppressive in C57BL/6J mice, facilitating or impairing viral clearance, respectively, in a model of a viral infection with a recombinant adenovirus. Importantly, inhalation of the odor improved the memory capacity in BALB/c mice in a fear-conditioning test, while it impaired this same capacity in C57BL/6J mice. The improvement in memory capacity in BALB/c was associated with higher CD3+ T cell infiltration into the hippocampus and increased local expression of mRNA coding for IL-1β, TNF-α, and IL-6 cytokines. In contrast, the memory impairment in C57BL/6 was associated with a reduction in CD3 numbers and an increase in IFN-γ. These data suggest an association between the immunomodulatory capacity of smells and their impact on the cognitive functions of the animals. These results highlight the potential of studying odors as therapeutic agents for CNS-related diseases.
Collapse
Affiliation(s)
- Aritz Lasarte-Cia
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Teresa Lozano
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marta Pérez-González
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Anatomy Department, School of Medicine, University of Navarra, Pamplona, Spain
| | - Marta Gorraiz
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Kristina Iribarren
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pablo Sarobe
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Obdulia Rabal
- Small Molecule Discovery Platform, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Anatomy Department, School of Medicine, University of Navarra, Pamplona, Spain
| | - Ana García-Osta
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Noelia Casares
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Juan José Lasarte
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
36
|
Weise G, Pösel C, Möller K, Kranz A, Didwischus N, Boltze J, Wagner DC. High-dosage granulocyte colony stimulating factor treatment alters monocyte trafficking to the brain after experimental stroke. Brain Behav Immun 2017; 60:15-26. [PMID: 27524669 DOI: 10.1016/j.bbi.2016.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/26/2016] [Accepted: 08/10/2016] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke elicits a prompt inflammatory response that is characterized by a well-timed recruitment of peripheral immune cells to the brain. Among these, monocytes play a particularly important, but multifaceted role and have been increasingly recognized to affect stroke outcome. Granulocyte colony stimulating factor (GCSF) is known for its immunosuppressive actions on mononuclear cells, but previous studies in the stroke field were mainly confined to its neuroprotective actions. Herein, we investigated whether GCSF affects post-stroke inflammation in a mouse model of focal brain ischemia by modulating monocyte responses. Treatment with GCSF was controlled by vehicle injection, sham surgery and naive animals. Despite a significant monocytosis, high-dosage GCSF reduced the number of brain-infiltrating monocytes/macrophages four days after stroke. Lower numbers of mononuclear phagocytes in the brain were associated with smaller cerebral edema and improved motor outcome after stroke. GCSF treatment over 72h, but not 24h diminished integrin expression on circulating Ly6C+ inflammatory monocytes. In vitro experiments further revealed that GCSF strongly promotes interleukin (IL)-10 secretion by activated mononuclear cells. Blockade of the IL-10 receptor partly reversed GCSF-induced downregulation of integrin surface expression. Overall, our results suggest that high-dosage GCSF mitigates monocyte infiltration after stroke, likely by attenuating integrin-mediated adhesion to the brain endothelium in an IL-10-dependent manner. Lower amounts of mononuclear cells in the brain translate to less severe brain edema and functional impairment and thus support a harmful role of Ly6C+ inflammatory monocytes in the acute stage of stroke.
Collapse
Affiliation(s)
- Gesa Weise
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; University of Leipzig, Department of Neurology, Leipzig, Germany.
| | - Claudia Pösel
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Karoline Möller
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Alexander Kranz
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Nadine Didwischus
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Institute for Biology, Human Biology, University of Leipzig, Leipzig, Germany
| | - Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Fraunhofer Research Institution of Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany; Massachusetts General Hospital and Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA, USA
| | - Daniel-Christoph Wagner
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Institute of Pathology, University Medical Center Mainz, Germany
| |
Collapse
|