1
|
Ocón B, Xiang M, Bi Y, Tan S, Brulois K, Ayesha A, Kunte M, Zhou C, LaJevic M, Lazarus N, Mengoni F, Sharma T, Montgomery S, Hooper JE, Huang M, Handel T, Dawson JRD, Kufareva I, Zabel BA, Pan J, Butcher EC. A lymphocyte chemoaffinity axis for lung, non-intestinal mucosae and CNS. Nature 2024; 635:736-745. [PMID: 39293486 PMCID: PMC11887596 DOI: 10.1038/s41586-024-08043-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Tissue-selective chemoattractants direct lymphocytes to epithelial surfaces to establish local immune environments, regulate immune responses to food antigens and commensal organisms, and protect from pathogens. Homeostatic chemoattractants for small intestines, colon and skin are known1,2, but chemotropic mechanisms selective for respiratory tract and other non-intestinal mucosal tissues remain poorly understood. Here we leveraged diverse omics datasets to identify GPR25 as a lymphocyte receptor for CXCL17, a chemoattractant cytokine whose expression by epithelial cells of airways, upper gastrointestinal and squamous mucosae unifies the non-intestinal mucosal tissues and distinguishes them from intestinal mucosae. Single-cell transcriptomic analyses show that GPR25 is induced on innate lymphocytes before emigration to the periphery, and is imprinted in secondary lymphoid tissues on activated B and T cells responding to immune challenge. GPR25 characterizes B and T tissue resident memory cells and regulatory T lymphocytes in non-intestinal mucosal tissues and lungs in humans and mediates lymphocyte homing to barrier epithelia of the airways, oral cavity, stomach, and biliary and genitourinary tracts in mouse models. GPR25 is also expressed by T cells in cerebrospinal fluid and CXCL17 by neurons, suggesting a role in central nervous system (CNS) immune regulation. We reveal widespread imprinting of GPR25 on regulatory T cells, suggesting a mechanistic link to population genetics evidence that GPR25 is protective in autoimmunity3,4. Our results define a GPR25-CXCL17 chemoaffinity axis with the potential to integrate immunity and tolerance at non-intestinal mucosae and the CNS.
Collapse
Affiliation(s)
- Borja Ocón
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Menglan Xiang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Yuhan Bi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Serena Tan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin Brulois
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Aiman Ayesha
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Manali Kunte
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Catherine Zhou
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Melissa LaJevic
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Nicole Lazarus
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Francesca Mengoni
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Tanya Sharma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Stephen Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jody E Hooper
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mian Huang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Tracy Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - John R D Dawson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Junliang Pan
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Eugene C Butcher
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
2
|
Witt LT, Greenfield KG, Knoop KA. Streptococcus agalactiae and Escherichia coli induce distinct effector γδ T cell responses during neonatal sepsis. iScience 2024; 27:109669. [PMID: 38646164 PMCID: PMC11033170 DOI: 10.1016/j.isci.2024.109669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/22/2024] [Accepted: 04/02/2024] [Indexed: 04/23/2024] Open
Abstract
Neonates born prematurely are vulnerable to life-threatening conditions such as bacterial sepsis. Streptococcus agalactiae (GBS) and Escherichia coli are frequent causative pathogens of neonatal sepsis, however, it remains unclear if these pathogens induce differential immune responses. We find that γδ T cells rapidly respond to single-organism GBS and E. coli bloodstream infections in neonatal mice. Furthermore, GBS and E. coli induce distinct cytokine production from IFN-γ and IL-17 producing γδ T cells, respectively. We also find that IL-17 production during E. coli infection is driven by γδTCR signaling, whereas IFN-γ production during GBS infection occurs independently of γδTCR signaling. The divergent effector responses of γδ T cells during GBS and E. coli infections impart distinctive neuroinflammatory phenotypes on the neonatal brain. Thus, the neonatal adaptive immune system differentially responds to distinct bacterial stimuli, resulting in unique neuroinflammatory phenotypes.
Collapse
Affiliation(s)
- Lila T. Witt
- Department of Immunology, Mayo Clinic, Rochester MN 55901, USA
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55901, USA
| | | | - Kathryn A. Knoop
- Department of Immunology, Mayo Clinic, Rochester MN 55901, USA
- Department of Pediatrics, Mayo Clinic, Rochester, MN 55901, USA
| |
Collapse
|
3
|
Zheng J, Wang L, Zhao S, Zhang W, Chang Y, Bosco DB, Huang T, Dheer A, Gao S, Xu S, Ayasoufi K, Al-Kharboosh R, Qi F, Xie M, Johnson AJ, Dong H, Quiñones-Hinojosa A, Wu LJ. TREM2 mediates MHCII-associated CD4+ T-cell response against gliomas. Neuro Oncol 2024; 26:811-825. [PMID: 37941134 PMCID: PMC11066911 DOI: 10.1093/neuonc/noad214] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Myeloid cells comprise up to 50% of the total tumor mass in glioblastoma (GBM) and have been implicated in promoting tumor progression and immunosuppression. Modulating the response of myeloid cells to the tumor has emerged as a promising new approach for cancer treatment. In this regard, we focus on the Triggering Receptor Expressed on Myeloid Cells 2 (TREM2), which has recently emerged as a novel immune modulator in peripheral tumors. METHODS We studied the TREM2 expression profile in various patient tumor samples and conducted single-cell transcriptomic analysis in both GBM patients and the GL261 mouse glioma model. We utilized multiple mouse glioma models and employed state-of-the-art techniques such as invivo 2-photon imaging, spectrum flow cytometry, and in vitro co-culture assays to study TREM2 function in myeloid cell-mediated phagocytosis of tumor cells, antigen presentation, and response of CD4+ T cells within the tumor hemispheres. RESULTS Our research revealed significantly elevated levels of TREM2 expression in brain tumors compared to other types of tumors in patients. TREM2 was predominantly localized in tumor-associated myeloid cells and was highly expressed in nearly all microglia, as well as various subtypes of macrophages. Surprisingly, in preclinical glioma models, TREM2 deficiency did not confer a beneficial effect; instead, it accelerated glioma progression. Through detailed investigations, we determined that TREM2 deficiency impaired the ability of tumor-myeloid cells to phagocytose tumor cells and led to reduced expression of MHCII. This deficiency further significantly decreased the presence of CD4+ T cells within the tumor hemispheres. CONCLUSIONS Our study unveiled a previously unrecognized protective role of tumor-myeloid TREM2. Specifically, we found that TREM2 enhances the phagocytosis of tumor cells and promotes an immune response by facilitating MHCII-associated CD4+ T-cell responses against gliomas.
Collapse
Affiliation(s)
- Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Wenjing Zhang
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Yuzhou Chang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tao Huang
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shan Gao
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Shengze Xu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Rawan Al-Kharboosh
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, Florida, USA
| | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
4
|
Witt LT, Greenfield KG, Knoop KA. Streptococcus agalactiae and Escherichia coli Induce Distinct Effector γδ T Cell Responses During Neonatal Sepsis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.02.560561. [PMID: 37873122 PMCID: PMC10592965 DOI: 10.1101/2023.10.02.560561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Neonates born prematurely are highly vulnerable to life-threatening conditions such as bacterial sepsis. Streptococcus agalactiae, also known as group B Streptococcus (GBS) and Escherichia coli are frequent causative pathogens of neonatal sepsis, however, it remains unclear if distinct sepsis pathogens induce differential adaptive immune responses. In the present study, we find that γδ T cells in neonatal mice rapidly respond to single-organism GBS and E. coli bloodstream infections and that these pathogens induce distinct activation and cytokine production from IFN-γ and IL-17 producing γδ T cells, respectively. We also report differential reliance on γδTCR signaling to elicit effector cytokine responses during neonatal sepsis, with IL-17 production during E. coli infection being driven by γδTCR signaling, and IFN-γ production during GBS infection occurring independently of γδTCR signaling. Furthermore, we report that the divergent effector responses of γδ T cells during GBS and E. coli infections impart distinctive neuroinflammatory phenotypes on the neonatal brain. The present study reveals that the neonatal adaptive immune system differentially responds to distinct bacterial stimuli, resulting in unique neuroinflammatory phenotypes.
Collapse
Affiliation(s)
- Lila T Witt
- Department of Immunology, Mayo Clinic, Rochester MN, USA 55901
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic
| | | | - Kathryn A Knoop
- Department of Immunology, Mayo Clinic, Rochester MN, USA 55901
- Department of Pediatrics, Mayo Clinic
| |
Collapse
|
5
|
Fain CE, Zheng J, Jin F, Ayasoufi K, Wu Y, Lilley MT, Dropik AR, Wolf DM, Rodriguez RC, Aibaidula A, Tritz ZP, Bouchal SM, Pewe LL, Urban SL, Chen Y, Chang SY, Hansen MJ, Kachergus JM, Shi J, Thompson EA, Jensen HE, Harty JT, Parney IF, Sun J, Wu LJ, Johnson AJ. Discrete class I molecules on brain endothelium differentially regulate neuropathology in experimental cerebral malaria. Brain 2024; 147:566-589. [PMID: 37776513 PMCID: PMC11734323 DOI: 10.1093/brain/awad319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/15/2023] [Accepted: 08/31/2023] [Indexed: 10/02/2023] Open
Abstract
Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T-cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell major histocompatibility complex (MHC) class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class I-like molecules, which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies, we interrogated human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observed that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T-cell infiltration, targeted cell death and regional blood-brain barrier disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T-cell activation, attenuated T-cell interaction with brain vasculature, lessened targeted cell death, preserved blood-brain barrier integrity and prevention of ECM and the death of the animal. We were able to show that these events were brain-specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria.
Collapse
Affiliation(s)
- Cori E Fain
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Jiaying Zheng
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905USA
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | | | - Yue Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Meredith T Lilley
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Abigail R Dropik
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Delaney M Wolf
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | | | - Abudumijiti Aibaidula
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905USA
| | - Zachariah P Tritz
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Samantha M Bouchal
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Lecia L Pewe
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Stina L Urban
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Yin Chen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Su-Youne Chang
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905USA
| | | | | | - Ji Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224USA
| | - Hadley E Jensen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | - John T Harty
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905USA
| | - Jie Sun
- Department of Medicine, University of Virginia, Charlottesville, VA 22903USA
| | - Long-Jun Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905USA
| |
Collapse
|
6
|
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: 1] [Impact Index Per Article: 0.5] [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
|
7
|
Ayasoufi K, Wolf DM, Namen SL, Jin F, Tritz ZP, Pfaller CK, Zheng J, Goddery EN, Fain CE, Gulbicki LR, Borchers AL, Reesman RA, Yokanovich LT, Maynes MA, Bamkole MA, Khadka RH, Hansen MJ, Wu LJ, Johnson AJ. Brain resident memory T cells rapidly expand and initiate neuroinflammatory responses following CNS viral infection. Brain Behav Immun 2023; 112:51-76. [PMID: 37236326 PMCID: PMC10527492 DOI: 10.1016/j.bbi.2023.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
The contribution of circulating verses tissue resident memory T cells (TRMs) to clinical neuropathology is an enduring question due to a lack of mechanistic insights. The prevailing view is TRMs are protective against pathogens in the brain. However, the extent to which antigen-specific TRMs induce neuropathology upon reactivation is understudied. Using the described phenotype of TRMs, we found that brains of naïve mice harbor populations of CD69+ CD103- T cells. Notably, numbers of CD69+ CD103- TRMs rapidly increase following neurological insults of various origins. This TRM expansion precedes infiltration of virus antigen-specific CD8 T cells and is due to proliferation of T cells within the brain. We next evaluated the capacity of antigen-specific TRMs in the brain to induce significant neuroinflammation post virus clearance, including infiltration of inflammatory myeloid cells, activation of T cells in the brain, microglial activation, and significant blood brain barrier disruption. These neuroinflammatory events were induced by TRMs, as depletion of peripheral T cells or blocking T cell trafficking using FTY720 did not change the neuroinflammatory course. Depletion of all CD8 T cells, however, completely abrogated the neuroinflammatory response. Reactivation of antigen-specific TRMs in the brain also induced profound lymphopenia within the blood compartment. We have therefore determined that antigen-specific TRMs can induce significant neuroinflammation, neuropathology, and peripheral immunosuppression. The use of cognate antigen to reactivate CD8 TRMs enables us to isolate the neuropathologic effects induced by this cell type independently of other branches of immunological memory, differentiating this work from studies employing whole pathogen re-challenge. This study also demonstrates the capacity for CD8 TRMs to contribute to pathology associated with neurodegenerative disorders and long-term complications associated with viral infections. Understanding functions of brain TRMs is crucial in investigating their role in neurodegenerative disorders including MS, CNS cancers, and long-term complications associated with viral infections including COVID-19.
Collapse
Affiliation(s)
| | - Delaney M Wolf
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Shelby L Namen
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Zachariah P Tritz
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Christian K Pfaller
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, United States; Paul-Ehrlich-Institut, Langen, Germany
| | - Jiaying Zheng
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Emma N Goddery
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Cori E Fain
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | | | - Anna L Borchers
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | | | - Lila T Yokanovich
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Mark A Maynes
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Michael A Bamkole
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Roman H Khadka
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Michael J Hansen
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Long-Jun Wu
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Molecular Medicine, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States.
| |
Collapse
|
8
|
Chen H, Liao Y, Zhang X, Shen H, Shang D, He Z, Zhou W, Song Z. Age- and sex-related differences of periodontal bone resorption, cognitive function, and immune state in APP/PS1 murine model of Alzheimer's disease. J Neuroinflammation 2023; 20:153. [PMID: 37370108 PMCID: PMC10294321 DOI: 10.1186/s12974-023-02790-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 04/24/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The existence of an interconnected mechanism between cognitive disorders and periodontitis has been confirmed by mounting evidence. However, the role of age or sex differences in this mechanism has been less studied. This study aims to investigate sex and age differences in the characterization of periodontal bone tissue, immune state and cognitive function in amyloid precursor protein/presenilin 1(APP/PS1) murine model of Alzheimer's disease (AD). METHODS Three- and twelve-month-old male and female APP/PS1 transgenic mice and wild-type (WT) littermates were used in this study. The Morris water maze (MWM) was used to assess cognitive function. The bone microarchitecture of the posterior maxillary alveolar bone was evaluated by microcomputed tomography (micro-CT). Pathological changes in periodontal bone tissue were observed by histological chemistry. The proportions of helper T cells1 (Th1), Th2, Th17 and regulatory T cells (Tregs) in the peripheral blood mononuclear cells (PBMCs) and brain samples were assessed by flow cytometry. RESULTS The learning ability and spatial memory of 12-month-old APP/PS1 mice was severely damaged. The changes in cognitive function were only correlated with age and genotype, regardless of sex. The 12-month-old APP/PS1 female mice exhibited markedly periodontal bone degeneration, evidenced by the decreased bone volume/total volume (BV/TV), trabecular thickness (Tb.Th), and bone mineral density (BMD), and the increased trabecular separation (Tb.Sp). The altered periodontal bone microarchitecture was associated with genotype, age and females. The flow cytometry data showed the increased Th1 and Th17 cells and the decreased Th2 cells in the brain and PBMC samples of 12-month-old APP/PS1 mice, compared to age- and sex-matched WT mice. However, there was no statistical correlation between age or sex and this immune state. CONCLUSIONS Our data emphasize that age and sex are important variables to consider in evaluating periodontal bone tissue of APP/PS1 mice, and the cognitive impairment is more related to age. In addition, immune dysregulation (Th1, Th2, and Th17 cells) was found in the brain tissue and PBMCs of APP/PS1 mice, but this alteration of immune state was not statistically correlated with sex or age.
Collapse
Affiliation(s)
- Huiwen Chen
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yue Liao
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xu Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Hui Shen
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Dihua Shang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhiyan He
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Wei Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China.
| | - Zhongchen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
9
|
Tritz ZP, Ayasoufi K, Wolf DM, Owens CA, Malo CS, Himes BT, Fain CE, Goddery EN, Yokanovich LT, Jin F, Hansen MJ, Parney IF, Wang C, Moynihan KD, Irvine DJ, Wittrup KD, Marcano RMD, Vile RG, Johnson AJ. Anti-PD-1 and Extended Half-life IL2 Synergize for Treatment of Murine Glioblastoma Independent of Host MHC Class I Expression. Cancer Immunol Res 2023; 11:763-776. [PMID: 36921098 PMCID: PMC10239322 DOI: 10.1158/2326-6066.cir-22-0570] [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: 07/25/2022] [Revised: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults, responsible for approximately 225,000 deaths per year. Despite preclinical successes, most interventions have failed to extend patient survival by more than a few months. Treatment with anti-programmed cell death protein 1 (anti-PD-1) immune checkpoint blockade (ICB) monotherapy has been beneficial for malignant tumors such as melanoma and lung cancers but has yet to be effectively employed in GBM. This study aimed to determine whether supplementing anti-PD-1 ICB with engineered extended half-life IL2, a potent lymphoproliferative cytokine, could improve outcomes. This combination therapy, subsequently referred to as enhanced checkpoint blockade (ECB), delivered intraperitoneally, reliably cures approximately 50% of C57BL/6 mice bearing orthotopic GL261 gliomas and extends median survival of the treated cohort. In the CT2A model, characterized as being resistant to CBI, ECB caused a decrease in CT2A tumor volume in half of measured animals similar to what was observed in GL261-bearing mice, promoting a trending survival increase. ECB generates robust immunologic responses, features of which include secondary lymphoid organ enlargement and increased activation status of both CD4 and CD8 T cells. This immunity is durable, with long-term ECB survivors able to resist GL261 rechallenge. Through employment of depletion strategies, ECB's efficacy was shown to be independent of host MHC class I-restricted antigen presentation but reliant on CD4 T cells. These results demonstrate ECB is efficacious against the GL261 glioma model through an MHC class I-independent mechanism and supporting further investigation into IL2-supplemented ICB therapies for tumors of the central nervous system.
Collapse
Affiliation(s)
| | | | | | | | - Courtney S. Malo
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | - Benjamin T. Himes
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN
| | - Cori E. Fain
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | - Emma N. Goddery
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | | | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN
| | | | - Ian F. Parney
- Mayo Clinic Department of Immunology, Rochester, MN
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN
| | - Chensu Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Kelly D. Moynihan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Howard Hughes Medical Institute, Chevy Chase, MD
| | - K. Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | - Richard G. Vile
- Mayo Clinic Department of Immunology, Rochester, MN
- Mayo Clinic Department of Molecular Medicine, Rochester, MN
| | - Aaron J. Johnson
- Mayo Clinic Department of Immunology, Rochester, MN
- Mayo Clinic Department of Molecular Medicine, Rochester, MN
- Mayo Clinic Department of Neurology, Rochester, MN
| |
Collapse
|
10
|
Zheng J, Wang L, Zhao S, Zhang W, Chang Y, Dheer A, Gao S, Xu S, Ayasoufi K, Al-kharboosh R, Xie M, Johnson AJ, Dong H, Quiñones-Hinojosa A, Wu LJ. TREM2 mediates MHCII-associated CD4 + T cell response against gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.05.535697. [PMID: 37066234 PMCID: PMC10104080 DOI: 10.1101/2023.04.05.535697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) was recently highlighted as a novel immune suppressive marker in peripheral tumors. The aim of this study was to characterize TREM2 expression in gliomas and investigate its contribution in glioma progression by using Trem2-/- mouse line. Our results showed that higher TREM2 expression was correlated with poor prognosis in glioma patients. Unexpectedly, TREM2 deficiency did not have a beneficial effect in a pre-clinical model of glioma. The increased TREM2 expression in glioma was likely due to increased myeloid cell infiltration, as evidenced by our single-cell analysis showing that almost all microglia and macrophages in gliomas were TREM2+. Furthermore, we found that deficiency of TREM2 impaired tumor-myeloid phagocytosis and MHCII presentation, and significantly reduced CD4+ T cells in tumor hemispheres. Our results revealed a previously unrecognized protective role of tumor-myeloid TREM2 in promoting MHCII-associated CD4+ T cell response against gliomas.
Collapse
Affiliation(s)
- Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Wenjing Zhang
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Yuzhou Chang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shan Gao
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Shengze Xu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Rawan Al-kharboosh
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, Florida, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
11
|
Faulhaber LD, Phuong AQ, Hartsuyker KJ, Cho Y, Mand KK, Harper SD, Olson AK, Garden GA, Shih AY, Gust J. Brain capillary obstruction during neurotoxicity in a mouse model of anti-CD19 chimeric antigen receptor T-cell therapy. Brain Commun 2021; 4:fcab309. [PMID: 35169706 PMCID: PMC8833245 DOI: 10.1093/braincomms/fcab309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/19/2021] [Accepted: 12/30/2021] [Indexed: 01/13/2023] Open
Abstract
Immunotherapy for haematologic malignancies with CD19-directed chimeric antigen receptor T cells has been highly successful at eradicating cancer but is associated with acute neurotoxicity in ∼40% of patients. This neurotoxicity correlates with systemic cytokine release syndrome, endothelial activation and disruption of endothelial integrity, but it remains unclear how these mechanisms interact and how they lead to neurologic dysfunction. We hypothesized that dysfunction of the neurovascular unit is a key step in the development of neurotoxicity. To recapitulate the interaction of the intact immune system with the blood-brain barrier, we first developed an immunocompetent mouse model of chimeric antigen receptor T-cell treatment-associated neurotoxicity. We treated wild-type mice with cyclophosphamide lymphodepletion followed by escalating doses of murine CD19-directed chimeric antigen receptor T cells. Within 3-5 days after chimeric antigen receptor T-cell infusion, these mice developed systemic cytokine release and abnormal behaviour as measured by daily neurologic screening exams and open-field testing. Histologic examination revealed widespread brain haemorrhages, diffuse extravascular immunoglobulin deposition, loss of capillary pericyte coverage and increased prevalence of string capillaries. To measure any associated changes in cerebral microvascular blood flow, we performed in vivo two-photon imaging through thinned-skull cranial windows. Unexpectedly, we found that 11.9% of cortical capillaries were plugged by Day 6 after chimeric antigen receptor T-cell treatment, compared to 1.1% in controls treated with mock transduced T cells. The capillary plugs comprised CD45+ leucocytes, a subset of which were CD3+ T cells. Plugging of this severity is expected to compromise cerebral perfusion. Indeed, we found widely distributed patchy hypoxia by hypoxyprobe immunolabelling. Increased serum levels of soluble ICAM-1 and VCAM-1 support a putative mechanism of increased leucocyte-endothelial adhesion. These data reveal that brain capillary obstruction may cause sufficient microvascular compromise to explain the clinical phenotype of chimeric antigen receptor T-cell neurotoxicity. The translational impact of this finding is strengthened by the fact that our mouse model closely approximates the kinetics and histologic findings of the chimeric antigen receptor T-cell neurotoxicity syndrome seen in human patients. This new link between systemic immune activation and neurovascular unit injury may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Lila D. Faulhaber
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Anthea Q. Phuong
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Kendra Jae Hartsuyker
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yeheun Cho
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Katie K. Mand
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Stuart D. Harper
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Aaron K. Olson
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Gwenn A. Garden
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Andy Y. Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Juliane Gust
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Goddery EN, Fain CE, Lipovsky CG, Ayasoufi K, Yokanovich LT, Malo CS, Khadka RH, Tritz ZP, Jin F, Hansen MJ, Johnson AJ. Microglia and Perivascular Macrophages Act as Antigen Presenting Cells to Promote CD8 T Cell Infiltration of the Brain. Front Immunol 2021; 12:726421. [PMID: 34526998 PMCID: PMC8435747 DOI: 10.3389/fimmu.2021.726421] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
CD8 T cell infiltration of the central nervous system (CNS) is necessary for host protection but contributes to neuropathology. Antigen presenting cells (APCs) situated at CNS borders are thought to mediate T cell entry into the parenchyma during neuroinflammation. The identity of the CNS-resident APC that presents antigen via major histocompatibility complex (MHC) class I to CD8 T cells is unknown. Herein, we characterize MHC class I expression in the naïve and virally infected brain and identify microglia and macrophages (CNS-myeloid cells) as APCs that upregulate H-2Kb and H-2Db upon infection. Conditional ablation of H-2Kb and H-2Db from CNS-myeloid cells allowed us to determine that antigen presentation via H-2Db, but not H-2Kb, was required for CNS immune infiltration during Theiler's murine encephalomyelitis virus (TMEV) infection and drives brain atrophy as a consequence of infection. These results demonstrate that CNS-myeloid cells are key APCs mediating CD8 T cell brain infiltration.
Collapse
Affiliation(s)
- Emma N. Goddery
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Cori E. Fain
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Chloe G. Lipovsky
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | | | - Lila T. Yokanovich
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Courtney S. Malo
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Roman H. Khadka
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Zachariah P. Tritz
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
13
|
Zhang X, Zhang X, Qiu C, Shen H, Zhang H, He Z, Song Z, Zhou W. The imbalance of Th17/Treg via STAT3 activation modulates cognitive impairment in P. gingivalis LPS-induced periodontitis mice. J Leukoc Biol 2021; 110:511-524. [PMID: 34342041 DOI: 10.1002/jlb.3ma0521-742rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 01/05/2023] Open
Abstract
Periodontitis is one of the most common oral diseases worldwide, and it is associated with various systemic diseases, including cognitive diseases. STAT3 regulates the inflammatory cascade and influences adaptive immunity by modulating Th17/Treg cell differentiation. In this study, we aimed to explore the effect of adaptive immunity inside and outside the brain on the association between periodontitis and cognitive impairment and understand the role of the STAT3 signaling pathway. We established Porphyromonas gingivalis LPS-induced periodontitis mice models by injecting P. gingivalis LPS into the gingival sulcus of mice. Behavioral tests showed that learning and memory abilities were impaired. The flow cytometry data showed an imbalance in the Th17/Treg ratio in the blood and brain samples of the mice. The expression of Th17-related cytokines (IL-1β, IL-17A, IL-21, and IL-22) increased, whereas that of Treg-related cytokines (IL-2 and IL-10) decreased in both the blood and the brain. The level of LPS increased and the STAT3 signaling pathway was activated during this process. These effects were reversed by C188-9, a STAT3 inhibitor. In conclusion, P. gingivalis LPS-induced periodontitis may promote the occurrence and progression of cognitive impairment by modulating the Th17/Treg balance inside and outside the brain. The STAT3 signaling pathway may have immunoregulatory effects on the mouth-to-brain axis.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China.,Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xuan Zhang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Che Qiu
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hui Shen
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Huanyu Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhiyan He
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhongchen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wei Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
14
|
Wongthida P, Schuelke MR, Driscoll CB, Kottke T, Thompson JM, Tonne J, Stone C, Huff AL, Wetmore C, Davies JA, Parker AL, Evgin L, Vile RG. Ad-CD40L mobilizes CD4 T cells for the treatment of brainstem tumors. Neuro Oncol 2020; 22:1757-1770. [PMID: 32459315 PMCID: PMC7746943 DOI: 10.1093/neuonc/noaa126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Diffuse midline glioma, formerly DIPG (diffuse intrinsic pontine glioma), is the deadliest pediatric brainstem tumor with median survival of less than one year. Here, we investigated (i) whether direct delivery of adenovirus-expressing cluster of differentiation (CD)40 ligand (Ad-CD40L) to brainstem tumors would induce immune-mediated tumor clearance and (ii) if so, whether therapy would be associated with a manageable toxicity due to immune-mediated inflammation in the brainstem. METHODS Syngeneic gliomas in the brainstems of immunocompetent mice were treated with Ad-CD40L and survival, toxicity, and immune profiles determined. A clinically translatable vector, whose replication would be tightly restricted to tumor cells, rAd-Δ24-CD40L, was tested in human patient-derived diffuse midline gliomas and immunocompetent models. RESULTS Expression of Ad-CD40L restricted to brainstem gliomas by pre-infection induced complete rejection, associated with immune cell infiltration, of which CD4+ T cells were critical for therapy. Direct intratumoral injection of Ad-CD40L into established brainstem tumors improved survival and induced some complete cures but with some acute toxicity. RNA-sequencing analysis showed that Ad-CD40L therapy induced neuroinflammatory immune responses associated with interleukin (IL)-6, IL-1β, and tumor necrosis factor α. Therefore, to generate a vector whose replication, and transgene expression, would be tightly restricted to tumor cells, we constructed rAd-Δ24-CD40L, the backbone of which has already entered clinical trials for diffuse midline gliomas. Direct intratumoral injection of rAd-Δ24-CD40L, with systemic blockade of IL-6 and IL-1β, generated significant numbers of cures with readily manageable toxicity. CONCLUSIONS Virus-mediated delivery of CD40L has the potential to be effective in treating diffuse midline gliomas without obligatory neuroinflammation-associated toxicity.
Collapse
Affiliation(s)
| | | | | | - Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jill M Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Cathy Stone
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Amanda L Huff
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Cynthia Wetmore
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - James A Davies
- Division of Cancer and Genetics, Cardiff University, Cardiff, UK
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University, Cardiff, UK
| | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard G Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Ayasoufi K, Pfaller CK, Evgin L, Khadka RH, Tritz ZP, Goddery EN, Fain CE, Yokanovich LT, Himes BT, Jin F, Zheng J, Schuelke MR, Hansen MJ, Tung W, Parney IF, Pease LR, Vile RG, Johnson AJ. Brain cancer induces systemic immunosuppression through release of non-steroid soluble mediators. Brain 2020; 143:3629-3652. [PMID: 33253355 PMCID: PMC7954397 DOI: 10.1093/brain/awaa343] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 01/09/2023] Open
Abstract
Immunosuppression of unknown aetiology is a hallmark feature of glioblastoma and is characterized by decreased CD4 T-cell counts and downregulation of major histocompatibility complex class II expression on peripheral blood monocytes in patients. This immunosuppression is a critical barrier to the successful development of immunotherapies for glioblastoma. We recapitulated the immunosuppression observed in glioblastoma patients in the C57BL/6 mouse and investigated the aetiology of low CD4 T-cell counts. We determined that thymic involution was a hallmark feature of immunosuppression in three distinct models of brain cancer, including mice harbouring GL261 glioma, B16 melanoma, and in a spontaneous model of diffuse intrinsic pontine glioma. In addition to thymic involution, we determined that tumour growth in the brain induced significant splenic involution, reductions in peripheral T cells, reduced MHC II expression on blood leucocytes, and a modest increase in bone marrow resident CD4 T cells. Using parabiosis we report that thymic involution, declines in peripheral T-cell counts, and reduced major histocompatibility complex class II expression levels were mediated through circulating blood-derived factors. Conversely, T-cell sequestration in the bone marrow was not governed through circulating factors. Serum isolated from glioma-bearing mice potently inhibited proliferation and functions of T cells both in vitro and in vivo. Interestingly, the factor responsible for immunosuppression in serum is non-steroidal and of high molecular weight. Through further analysis of neurological disease models, we determined that the immunosuppression was not unique to cancer itself, but rather occurs in response to brain injury. Non-cancerous acute neurological insults also induced significant thymic involution and rendered serum immunosuppressive. Both thymic involution and serum-derived immunosuppression were reversible upon clearance of brain insults. These findings demonstrate that brain cancers cause multifaceted immunosuppression and pinpoint circulating factors as a target of intervention to restore immunity.
Collapse
Affiliation(s)
| | - Christian K Pfaller
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Paul-Ehrlich-Institute, Division of Veterinary Medicine, Langen, Germany
| | - Laura Evgin
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
| | - Roman H Khadka
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Zachariah P Tritz
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Emma N Goddery
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Cori E Fain
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Lila T Yokanovich
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Benjamin T Himes
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN, USA
| | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Jiaying Zheng
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Matthew R Schuelke
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Immunology, Mayo Clinic Medical Scientist Training Program, Rochester, Minnesota, USA
| | | | - Wesley Tung
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Ian F Parney
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Neurologic Surgery, Rochester, MN, USA
| | - Larry R Pease
- Mayo Clinic Department of Immunology, Rochester, MN, USA
| | - Richard G Vile
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN, USA
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, USA
- Mayo Clinic Department of Neurology, Rochester, MN, USA
| |
Collapse
|
16
|
Tritz ZP, Orozco RC, Malo CS, Ayasoufi K, Fain CE, Khadka RH, Goddery EN, Yokanovich LT, Settell ML, Hansen MJ, Jin F, Pavelko KD, Pease LR, Johnson AJ. Conditional Silencing of H-2D b Class I Molecule Expression Modulates the Protective and Pathogenic Kinetics of Virus-Antigen-Specific CD8 T Cell Responses during Theiler's Virus Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:1228-1238. [PMID: 32737149 DOI: 10.4049/jimmunol.2000340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection of the CNS is cleared in C57BL/6 mice by a CD8 T cell response restricted by the MHC class I molecule H-2Db The identity and function of the APC(s) involved in the priming of this T cell response is (are) poorly defined. To address this gap in knowledge, we developed an H-2Db LoxP-transgenic mouse system using otherwise MHC class I-deficient C57BL/6 mice, thereby conditionally ablating MHC class I-restricted Ag presentation in targeted APC subpopulations. We observed that CD11c+ APCs are critical for early priming of CD8 T cells against the immunodominant TMEV peptide VP2121-130 Loss of H-2Db on CD11c+ APCs mitigates the CD8 T cell response, preventing early viral clearance and immunopathology associated with CD8 T cell activity in the CNS. In contrast, animals with H-2Db-deficient LysM+ APCs retained early priming of Db:VP2121-130 epitope-specific CD8 T cells, although a modest reduction in immune cell entry into the CNS was observed. This work establishes a model enabling the critical dissection of H-2Db-restricted Ag presentation to CD8 T cells, revealing cell-specific and temporal features involved in the generation of CD8 T cell responses. Employing this novel system, we establish CD11c+ cells as pivotal to the establishment of acute antiviral CD8 T cell responses against the TMEV immunodominant epitope VP2121-130, with functional implications both for T cell-mediated viral control and immunopathology.
Collapse
Affiliation(s)
- Zachariah P Tritz
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Robin C Orozco
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Courtney S Malo
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | | | - Cori E Fain
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Roman H Khadka
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Emma N Goddery
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Lila T Yokanovich
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Megan L Settell
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905
| | | | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN 55905
| | | | - Larry R Pease
- Mayo Clinic Department of Immunology, Rochester, MN 55905.,Mayo Clinic Department of Biochemistry, Rochester, MN 55905
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN 55905; .,Mayo Clinic Department of Molecular Medicine, Rochester, MN 55905; and.,Mayo Clinic Department of Neurology, Rochester, MN 55905
| |
Collapse
|
17
|
Irekeola AA, E. A. R. ENS, Mat Lazim N, Mohamud R, Yean CY, Shueb RH. Technical Considerations in Ex Vivo Human Regulatory T Cell Migration and Suppression Assays. Cells 2020; 9:cells9020487. [PMID: 32093265 PMCID: PMC7072784 DOI: 10.3390/cells9020487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Regulatory T cells (Tregs) are renowned for maintaining homeostasis and self-tolerance through their ability to suppress immune responses. For over two decades, Tregs have been the subject of intensive research. The immunosuppressive and migratory potentials of Tregs have been exploited, especially in the areas of cancer, autoimmunity and vaccine development, and many assay protocols have since been developed. However, variations in assay conditions in different studies, as well as covert experimental factors, pose a great challenge to the reproducibility of results. Here, we focus on human Tregs derived from clinical samples and highlighted caveats that should be heeded when conducting Tregs suppression and migration assays. We particularly delineated how factors such as sample processing, choice of reagents and equipment, optimization and other experimental conditions could introduce bias into the assay, and we subsequently proffer recommendations to enhance reliability and reproducibility of results. It is hoped that prioritizing these factors will reduce the tendencies of generating false and misleading results, and thus, help improve our understanding and interpretation of Tregs functional studies.
Collapse
Affiliation(s)
- Ahmad Adebayo Irekeola
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Kelantan, Malaysia; (A.A.I.); (E.N.S.E.A.R.); (C.Y.Y.)
- Microbiology Unit, Department of Biological Sciences, College of Natural and Applied Sciences, Summit University Offa, Offa PMB 4412, Kwara State, Nigeria
| | - Engku Nur Syafirah E. A. R.
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Kelantan, Malaysia; (A.A.I.); (E.N.S.E.A.R.); (C.Y.Y.)
| | - Norhafiza Mat Lazim
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Chan Yean Yean
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Kelantan, Malaysia; (A.A.I.); (E.N.S.E.A.R.); (C.Y.Y.)
| | - Rafidah Hanim Shueb
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Kelantan, Malaysia; (A.A.I.); (E.N.S.E.A.R.); (C.Y.Y.)
- Correspondence:
| |
Collapse
|
18
|
Schuelke MR, Wongthida P, Thompson J, Kottke T, Driscoll CB, Huff AL, Shim KG, Coffey M, Pulido J, Evgin L, Vile RG. Diverse immunotherapies can effectively treat syngeneic brainstem tumors in the absence of overt toxicity. J Immunother Cancer 2019; 7:188. [PMID: 31315671 PMCID: PMC6637625 DOI: 10.1186/s40425-019-0673-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Immunotherapy has shown remarkable clinical promise in the treatment of various types of cancers. However, clinical benefits derive from a highly inflammatory mechanism of action. This presents unique challenges for use in pediatric brainstem tumors including diffuse intrinsic pontine glioma (DIPG), since treatment-related inflammation could cause catastrophic toxicity. Therefore, the goal of this study was to investigate whether inflammatory, immune-based therapies are likely to be too dangerous to pursue for the treatment of pediatric brainstem tumors. METHODS To complement previous immunotherapy studies using patient-derived xenografts in immunodeficient mice, we developed fully immunocompetent models of immunotherapy using transplantable, syngeneic tumors. These four models - HSVtk/GCV suicide gene immunotherapy, oncolytic viroimmunotherapy, adoptive T cell transfer, and CAR T cell therapy - have been optimized to treat tumors outside of the CNS and induce a broad spectrum of inflammatory profiles, maximizing the chances of observing brainstem toxicity. RESULTS All four models achieved anti-tumor efficacy in the absence of toxicity, with the exception of recombinant vaccinia virus expressing GMCSF, which demonstrated inflammatory toxicity. Histology, imaging, and flow cytometry confirmed the presence of brainstem inflammation in all models. Where used, the addition of immune checkpoint blockade did not introduce toxicity. CONCLUSIONS It remains imperative to regard the brainstem with caution for immunotherapeutic intervention. Nonetheless, we show that further careful development of immunotherapies for pediatric brainstem tumors is warranted to harness the potential potency of anti-tumor immune responses, despite their possible toxicity within this anatomically sensitive location.
Collapse
Affiliation(s)
- Matthew R Schuelke
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Amanda L Huff
- Virology and Gene Therapy Track, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin G Shim
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matt Coffey
- Oncolytics Biotech, Inc., Calgary, AB, T2N 1X7, Canada
| | - Jose Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Laura Evgin
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard G Vile
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Leeds Cancer Research UK Clinical Centre, Faculty of Medicine and Health, St James' University Hospital, University of Leeds, West Yorkshire, UK.
| |
Collapse
|
19
|
Malo CS, Khadka RH, Ayasoufi K, Jin F, AbouChehade JE, Hansen MJ, Iezzi R, Pavelko KD, Johnson AJ. Immunomodulation Mediated by Anti-angiogenic Therapy Improves CD8 T Cell Immunity Against Experimental Glioma. Front Oncol 2018; 8:320. [PMID: 30211113 PMCID: PMC6124655 DOI: 10.3389/fonc.2018.00320] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/26/2018] [Indexed: 01/13/2023] Open
Abstract
Glioblastoma (GBM) is a lethal cancer of the central nervous system with a median survival rate of 15 months with treatment. Thus, there is a critical need to develop novel therapies for GBM. Immunotherapy is emerging as a promising therapeutic strategy. However, current therapies for GBM, in particular anti-angiogenic therapies that block vascular endothelial growth factor (VEGF), may have undefined consequences on the efficacy of immunotherapy. While this treatment is primarily prescribed to reduce tumor vascularization, multiple immune cell types also express VEGF receptors, including the most potent antigen-presenting cell, the dendritic cell (DC). Therefore, we assessed the role of anti-VEGF therapy in modifying DC function. We found that VEGF blockade results in a more mature DC phenotype in the brain, as demonstrated by an increase in the expression of the co-stimulatory molecules B7-1, B7-2, and MHC II. Furthermore, we observed reduced levels of the exhaustion markers PD-1 and Tim-3 on brain-infiltrating CD8 T cells, indicating improved functionality. Thus, anti-angiogenic therapy has the potential to be used in conjunction with and enhance immunotherapy for GBM.
Collapse
Affiliation(s)
- Courtney S Malo
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Roman H Khadka
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | | | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Michael J Hansen
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Raymond Iezzi
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, United States
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Department of Neurology, Mayo Clinic, Rochester, MN, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
20
|
Malo CS, Huggins MA, Goddery EN, Tolcher HMA, Renner DN, Jin F, Hansen MJ, Pease LR, Pavelko KD, Johnson AJ. Non-equivalent antigen presenting capabilities of dendritic cells and macrophages in generating brain-infiltrating CD8 + T cell responses. Nat Commun 2018; 9:633. [PMID: 29434238 PMCID: PMC5809416 DOI: 10.1038/s41467-018-03037-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/15/2018] [Indexed: 01/05/2023] Open
Abstract
The contribution of antigen-presenting cell (APC) types in generating CD8+ T cell responses in the central nervous system (CNS) is not fully defined, limiting the development of vaccines and understanding of immune-mediated neuropathology. Here, we generate a transgenic mouse that enables cell-specific deletion of the H-2Kb MHC class I molecule. By deleting H-2Kb on dendritic cells and macrophages, we compare the effect of each APC in three distinct models of neuroinflammation: picornavirus infection, experimental cerebral malaria, and a syngeneic glioma. Dendritic cells and macrophages both activate CD8+ T cell responses in response to these CNS immunological challenges. However, the extent to which each of these APCs contributes to CD8+ T cell priming varies. These findings reveal distinct functions for dendritic cells and macrophages in generating CD8+ T cell responses to neurological disease.
Collapse
Affiliation(s)
- Courtney S Malo
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Matthew A Huggins
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Emma N Goddery
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Heather M A Tolcher
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Danielle N Renner
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Neurobiology of Disease Graduate Program, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Fang Jin
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Michael J Hansen
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
- Department of Neurology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
21
|
Pavelko KD, Bell MP, Harrington SM, Dong H. B7-H1 Influences the Accumulation of Virus-Specific Tissue Resident Memory T Cells in the Central Nervous System. Front Immunol 2017; 8:1532. [PMID: 29170671 PMCID: PMC5684101 DOI: 10.3389/fimmu.2017.01532] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/27/2017] [Indexed: 01/01/2023] Open
Abstract
Therapies that target the PD-1/B7-H1 axis have revolutionized cancer treatment, yet precise knowledge of how this pathway provides benefit continues to evolve. Here, we report a novel role for the immune checkpoint ligand B7-H1 in the accumulation of tissue-resident memory CD8+ T-cells (TRM). After intracranial infection, Theiler's murine encephalomyelitis virus (TMEV) generates TRM that are maintained in the central nervous system (CNS) tissues of B7-H1WT animals. Although no differences in acute T-cell responses between B7-H1WT and B7-H1KO are observed, at long-term periods post-infection the maintenance of CD8+ TRM is diminished in B7-H1KO animals. This is accompanied by redistribution of the resident CD8+ population from primarily CD103+ TRM to a diminished population of TRM and a preponderance of non-specified PD-1+ CD103- CD8+ T-cells. T-cell transfer studies demonstrate that host B7-H1 is necessary for maintaining TRM and limiting accumulation of PD-1+ CD103- CD8+ T-cells. The lack of host B7-H1 results in compromised control of a heterologous virus re-challenge demonstrating a functional defect in TRM mediated virus control. This study reveals a new role for B7-H1 in TRM and pro-inflammatory PD-1+ CD103- CD8+ T-cell accumulation in the CNS and gives insight for using B7-H1/PD-1 blockade in modulating long-term T-cell protection.
Collapse
Affiliation(s)
- Kevin D. Pavelko
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Michael P. Bell
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Susan M. Harrington
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Haidong Dong
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Urology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
22
|
Huseby Kelcher AM, Atanga PA, Gamez JD, Cumba Garcia LM, Teclaw SJ, Pavelko KD, Macura SI, Johnson AJ. Brain atrophy in picornavirus-infected FVB mice is dependent on the H-2D b class I molecule. FASEB J 2017; 31:2267-2275. [PMID: 28188174 DOI: 10.1096/fj.201601055r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 01/09/2023]
Abstract
Brain atrophy is a common feature of numerous neurologic diseases in which the role of neuroinflammation remains ill-defined. In this study, we evaluated the contribution of major histocompatibility complex class I molecules to brain atrophy in Theiler's murine encephalomyelitis virus (TMEV)-infected transgenic FVB mice that express the Db class I molecule. FVB/Db and wild-type FVB mice were evaluated for changes in neuroinflammation, virus clearance, neuropathology, and development of brain atrophy via T2-weighted MRI and subsequent 3-dimensional volumetric analysis. Significant brain atrophy and hippocampal neuronal loss were observed in TMEV-infected FVB/Db mice, but not in wild-type FVB mice. Brain atrophy was observed at 1 mo postinfection and persisted through the 4-mo observation period. Of importance, virus-infected FVB/Db mice elicited a strong CD8 T-cell response toward the immunodominant Db-restricted TMEV-derived peptide, VP2121-130, and cleared TMEV from the CNS. In addition, immunofluorescence revealed CD8 T cells near virus-infected neurons; therefore, we hypothesize that class I restricted CD8 T-cell responses promote development of brain atrophy. This model provides an opportunity to analyze the contribution of immune cells to brain atrophy in a system where persistent virus infection and demyelination are not factors in long-term neuropathology.-Huseby Kelcher, A. M., Atanga, P. A., Gamez, J. D., Cumba Garcia, L. M., Teclaw, S. J., Pavelko, K. D., Macura, S. I., Johnson. A. J. Brain atrophy in picornavirus-infected FVB mice is dependent on the H-2Db class I molecule.
Collapse
Affiliation(s)
- April M Huseby Kelcher
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Pascal A Atanga
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey D Gamez
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Luz M Cumba Garcia
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Slobodan I Macura
- Nuclear Magnetic Resonance Core Facility, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA; .,Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|