1
|
Nguyen NV, Lin JS, Parikh MJ, Cutri RM, Shibata SB. Targeted spiral ganglion neuron degeneration in parvalbumin-Cre neonatal mice. Mol Ther Methods Clin Dev 2025; 33:101440. [PMID: 40206512 PMCID: PMC11979521 DOI: 10.1016/j.omtm.2025.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
The spiral ganglion neurons (SGNs) are the primary afferent neurons in the cochlea; damage to the SGNs leads to irreversible hearing impairment. Mouse models that allow selective SGN degeneration while sparing other cell types in the cochlea are lacking. Here, we investigated a genetic ablation method of the SGN using a Cre-responsive adeno-associated virus (AAV) vector expressing diphtheria toxin subunit-A (DTA). We microinjected AAV2-retro-FLEX-DTA-mCherry driven by the EF1a or hSYN promoter in neonatal parvalbumin-Cre (PVCre) and wild-type strains via the posterior semicircular canal. Apoptotic markers were observed in the degenerating SGNs as early as 3 days. After 1 week, we assessed the SGN cell density, revealing an average degeneration of 60% for AAV-DTA driven by the EF1a promoter and 61% for that driven by the hSYN promoter. By 1 month, injected ears demonstrated a nearly complete loss of SGN, while hair cell morphology was intact. The auditory brain stem response result showed significantly elevated threshold shifts at 1 month, while the distortion-product otoacoustic emissions function remained intact. Furthermore, we show that our method did not effectively ablate SGN in adult PVCre mice. We generated a neonatal mouse model with primary SGN degeneration in PVCre mice, mimicking auditory neuropathy phenotype using an AAV Cre-dependent expression of DTA.
Collapse
Affiliation(s)
- Nhi V. Nguyen
- Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, CA 90033, USA
| | - Joshua S. Lin
- Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, CA 90033, USA
| | - Miti J. Parikh
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Raffaello M. Cutri
- Department of Otolaryngology-Head and Neck Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Seiji B. Shibata
- Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
2
|
Cui J, Xu H, Yu J, Ran S, Zhang X, Li Y, Chen Z, Niu Y, Wang S, Ye W, Chen W, Wu J, Xia J. Targeted depletion of PD-1-expressing cells induces immune tolerance through peripheral clonal deletion. Sci Immunol 2024; 9:eadh0085. [PMID: 38669317 DOI: 10.1126/sciimmunol.adh0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
Thymic negative selection of the T cell receptor (TCR) repertoire is essential for establishing self-tolerance and acquired allograft tolerance following organ transplantation. However, it is unclear whether and how peripheral clonal deletion of alloreactive T cells induces transplantation tolerance. Here, we establish that programmed cell death protein 1 (PD-1) is a hallmark of alloreactive T cells and is associated with clonal expansion after alloantigen encounter. Moreover, we found that diphtheria toxin receptor (DTR)-mediated ablation of PD-1+ cells reshaped the TCR repertoire through peripheral clonal deletion of alloreactive T cells and promoted tolerance in mouse transplantation models. In addition, by using PD-1-specific depleting antibodies, we found that antibody-mediated depletion of PD-1+ cells prevented heart transplant rejection and the development of experimental autoimmune encephalomyelitis (EAE) in humanized PD-1 mice. Thus, these data suggest that PD-1 is an attractive target for peripheral clonal deletion and induction of immune tolerance.
Collapse
Affiliation(s)
- Jikai Cui
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Heng Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Wenhao Chen
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute and Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Translational Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Translational Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Pinos I, Coronel J, Albakri A, Blanco A, McQueen P, Molina D, Sim J, Fisher EA, Amengual J. β-Carotene accelerates the resolution of atherosclerosis in mice. eLife 2024; 12:RP87430. [PMID: 38319073 PMCID: PMC10945528 DOI: 10.7554/elife.87430] [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] [Indexed: 02/07/2024] Open
Abstract
β-Carotene oxygenase 1 (BCO1) catalyzes the cleavage of β-carotene to form vitamin A. Besides its role in vision, vitamin A regulates the expression of genes involved in lipid metabolism and immune cell differentiation. BCO1 activity is associated with the reduction of plasma cholesterol in humans and mice, while dietary β-carotene reduces hepatic lipid secretion and delays atherosclerosis progression in various experimental models. Here we show that β-carotene also accelerates atherosclerosis resolution in two independent murine models, independently of changes in body weight gain or plasma lipid profile. Experiments in Bco1-/- mice implicate vitamin A production in the effects of β-carotene on atherosclerosis resolution. To explore the direct implication of dietary β-carotene on regulatory T cells (Tregs) differentiation, we utilized anti-CD25 monoclonal antibody infusions. Our data show that β-carotene favors Treg expansion in the plaque, and that the partial inhibition of Tregs mitigates the effect of β-carotene on atherosclerosis resolution. Our data highlight the potential of β-carotene and BCO1 activity in the resolution of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Johana Coronel
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Asma'a Albakri
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Amparo Blanco
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Patrick McQueen
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Donald Molina
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - JaeYoung Sim
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University Grossman School of Medicine, NYU Langone Medical CenterNew YorkUnited States
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| |
Collapse
|
4
|
Silva JG, Pais Ferreira D, Dumez A, Wyss T, Veber R, Danilo M, Pinschewer DD, Charmoy M, Held W. Emergence and fate of stem cell-like Tcf7+ CD8 + T cells during a primary immune response to viral infection. Sci Immunol 2023; 8:eadh3113. [PMID: 37976346 DOI: 10.1126/sciimmunol.adh3113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
In response to infection, naïve CD8+ T (TN) cells yield a large pool of short-lived terminal effector (TTE) cells that eliminate infected host cells. In parallel, a minor population of stem cell-like central memory (TCM) cells forms, which has the capacity to maintain immunity after pathogen clearance. It has remained uncertain whether stem-like TCM cells arise by dedifferentiation from a subset of cytolytic TTE cells or whether priming generates stem-like cells capable of seeding the TCM compartment and, if so, when cytolytic TTE cells branch off. Here, we show that CD8+ T cells with stem-like properties, which are identified by the expression of TCF1 (encoded by Tcf7), are present across the primary response to infection. Priming programs TN cells to undergo multiple cell divisions, over the course of which TCF1 expression is maintained. These TCF1+ cells further expand relatively independently of systemic inflammation, antigen dose, or affinity, and they quantitatively yield TCF1+ TCM cells after pathogen clearance. Inflammatory signals suppress TCF1 expression in early divided TCF1+ cells. TCF1 down-regulation is associated with the irreversible loss of self-renewal capacity and the silencing of stem/memory genes, which precedes the stable acquisition of a TTE state. TCF1 expression restrains cell cycling, explaining in part the limited expansion of TCF1+ relative to TCF1- cells during the primary response. Thus, our data are consistent with terminal differentiation of effector cells being a step-wise process that is initiated by inflammation in primed stem-like cells, which would otherwise become central memory cells by default.
Collapse
Affiliation(s)
- Joana Gomes Silva
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | | | - Alexandre Dumez
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Tania Wyss
- Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Romain Veber
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Maxime Danilo
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Daniel D Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Mélanie Charmoy
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Werner Held
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Santiago KN, Kozlik T, Liedhegner ES, Slick RA, Lawlor MW, Nardelli DT. Effects of Regulatory T Cell Depletion in BALB/c Mice Infected with Low Doses of Borrelia burgdorferi. Pathogens 2023; 12:189. [PMID: 36839461 PMCID: PMC9965304 DOI: 10.3390/pathogens12020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
We previously demonstrated that a depletion of regulatory T (Treg) cells in Lyme arthritis-resistant C57BL/6 mice leads to pathological changes in the tibiotarsal joints following infection with Borrelia burgdorferi. Here, we assessed the effects of Treg cells on the response to B. burgdorferi infection in BALB/c mice, which exhibit infection-dose-dependent disease and a different sequence of immune events than C57BL/6 mice. The depletion of Treg cells prior to infection with 1 × 102, but not 5 × 103, organisms led to increased swelling of the tibiotarsal joints. However, Treg cell depletion did not significantly affect the development of histopathology at these low doses of infection. BALB/c mice depleted of Treg cells before infection with 1 × 103 spirochetes harbored a higher borrelial load in the hearts and exhibited higher levels of serum interleukin-10 five weeks later. These results indicate that Treg cells regulate certain aspects of the response to B. burgdorferi in a mouse strain that may display a range of disease severities. As the presentation of Lyme disease may vary among humans, it is necessary to consider multiple animal models to obtain a complete picture of the various means by which Treg cells affect the host response to B. burgdorferi.
Collapse
Affiliation(s)
- Kaitlyn N. Santiago
- Department of Biomedical Sciences, University of Wisconsin–Milwaukee, Milwaukee, WI 53211, USA
| | - Tanya Kozlik
- Department of Biomedical Sciences, University of Wisconsin–Milwaukee, Milwaukee, WI 53211, USA
| | - Elizabeth S. Liedhegner
- Department of Biomedical Sciences, University of Wisconsin–Milwaukee, Milwaukee, WI 53211, USA
| | - Rebecca A. Slick
- Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael W. Lawlor
- Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Dean T. Nardelli
- Department of Biomedical Sciences, University of Wisconsin–Milwaukee, Milwaukee, WI 53211, USA
| |
Collapse
|
6
|
Yang H, Park SY, Baek H, Lee C, Chung G, Liu X, Lee JH, Kim B, Kwon M, Choi H, Kim HJ, Kim JY, Kim Y, Lee YS, Lee G, Kim SK, Kim JS, Chang YT, Jung WS, Kim KH, Bae H. Adoptive therapy with amyloid-β specific regulatory T cells alleviates Alzheimer's disease. Am J Cancer Res 2022; 12:7668-7680. [PMID: 36451854 PMCID: PMC9706584 DOI: 10.7150/thno.75965] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022] Open
Abstract
Rationale: Neuroinflammation is a primary feature of Alzheimer's disease (AD), for which an increasing number of drugs have been specifically developed. The present study aimed to define the therapeutic impact of a specific subpopulation of T cells that can suppress excessive inflammation in various immune and inflammatory disorders, namely, CD4+CD25+Foxp3+ regulatory T cells (Tregs). Methods: To generate Aβ antigen-specific Tregs (Aβ+ Tregs), Aβ 1-42 peptide was applied in vivo and subsequent in vitro splenocyte culture. After isolating Tregs by magnetic bead based purification method, Aβ+ Tregs were adoptively transferred into 3xTg-AD mice via tail vein injection. Therapeutic efficacy was confirmed with behavior test, Western blot, quantitative real-time PCR (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry staining (IHC). In vitro suppression assay was performed to evaluate the suppressive activity of Aβ+ Tregs using flow cytometry. Thy1.1+ Treg trafficking and distribution was analyzed to explore the infused Tregs migration into specific organs in an antigen-driven manner in AD mice. We further assessed cerebral glucose metabolism using 18F-FDG-PET, an imaging approach for AD biological definition. Subsequently, we evaluated the migration of Aβ+ Tregs toward Aβ activated microglia using live cell imaging, chemotaxis, antibody blocking and migration assay. Results: We showed that Aβ-stimulated Tregs inhibited microglial proinflammatory activity and modulated the microglial phenotype via bystander suppression. Single adoptive transfer of Aβ+ Tregs was enough to induce amelioration of cognitive impairments, Aβ accumulation, hyper-phosphorylation of tau, and neuroinflammation during AD pathology. Moreover, Aβ-specific Tregs effectively inhibited inflammation in primary microglia induced by Aβ exposure. It may indicate bystander suppression in which Aβ-specific Tregs promote immune tolerance by secreting cytokines to modulate immune responses during neurodegeneration. Conclusions: The administration of Aβ antigen-specific regulatory T cells may represent a new cellular therapeutic strategy for AD that acts by modulating the inflammatory status in AD.
Collapse
Affiliation(s)
- HyeJin Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Seon-Young Park
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Hyunjung Baek
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Chanju Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea,Cancer Immunology Branch, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Xiao Liu
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Ji Hwan Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Byungkyu Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Minjin Kwon
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Hyojung Choi
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Hyung Joon Kim
- Institute of Life Science & Biotechnology, VT Bio. Co., Ltd. 3 rd FL, 16 Samseong-ro 76-gil, Gangnam-gu, Seoul 06185, Korea
| | - Jae Yoon Kim
- Institute of Life Science & Biotechnology, VT Bio. Co., Ltd. 3 rd FL, 16 Samseong-ro 76-gil, Gangnam-gu, Seoul 06185, Korea
| | - Younsub Kim
- Department of Anatomy and Acupoint, College of Korean Medicine, Gachon University, Seongnam 13120, Korea
| | - Ye-Seul Lee
- Department of Anatomy and Acupoint, College of Korean Medicine, Gachon University, Seongnam 13120, Korea
| | - Gaheon Lee
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Jin Su Kim
- Division of RI Application, Korea Institute Radiological and Medical Sciences, 75 Nowon-ro, Nowon-Gu, Seoul 01812, Korea
| | - Young-Tae Chang
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Korea,Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang 37673, Korea
| | - Woo Sang Jung
- Stroke center, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea
| | - Kyung Hwa Kim
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea,✉ Corresponding authors: Kyung Hwa Kim: Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea; Tel.: +82-51‑200‑7534; Fax: +82-51-200-7905; . Hyunsu Bae: Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea; Tel.: +82-2-961-9316; Fax: +82-2-961-0333; .; © The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea,✉ Corresponding authors: Kyung Hwa Kim: Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea; Tel.: +82-51‑200‑7534; Fax: +82-51-200-7905; . Hyunsu Bae: Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea; Tel.: +82-2-961-9316; Fax: +82-2-961-0333; .; © The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions
| |
Collapse
|
7
|
Kiapour N, Wu B, Wang Y, Seyedsadr M, Kapoor S, Zhang X, Elzoheiry M, Kasimoglu E, Wan Y, Markovic-Plese S. Therapeutic Effect of Anti-CD52 Monoclonal Antibody in Multiple Sclerosis and Its Animal Models Is Mediated via T Regulatory Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:49-56. [PMID: 35750335 PMCID: PMC9458467 DOI: 10.4049/jimmunol.2100176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
The objective of this study is to determine the mechanism of action of anti-CD52 mAb treatment in patients with relapsing-remitting multiple sclerosis (RRMS). Experimental autoimmune encephalomyelitis (EAE), an animal model of the disease, was used to address the role of T regulatory cells (Tregs) in the anti-CD52 mAb-induced suppression of the disease. In vitro studies on PBMCs from RRMS patients and matched healthy controls determined the effect of IL-7 on the expansion of CD4+CD25+CD127- Tregs and induction of their suppressive phenotype. This study using EAE animal models of MS has shown that mouse anti-CD52 mAb suppression of clinical disease was augmented by coadministration of IL-7 and partially reversed by anti-IL-7 mAb. In vitro human studies showed that IL-7 induced expansion of CD4+CD25+CD127- Tregs and increased their FOXP3, GITIR, CD46, CTLA-4, granzyme B, and perforin expression. Anti-CD52 mAb treatment of mice with relapsing-remitting EAE induced expansion of Foxp3+CD4+ Tregs and the suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in peripheral immune organs and CNS infiltrates. The effect was detected immediately after the treatment and maintained over long-term follow-up. Foxp3+CD4+ Treg-mediated suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in the spinal cord infiltrates was reversed after inducible Foxp3 depletion. Our results demonstrated that the therapeutic effect of U.S. Food and Drug Administration-approved anti-CD52 mAb is dependent on the presence of Tregs.
Collapse
Affiliation(s)
- Nazanin Kiapour
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Bing Wu
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Frontier Science Center for Immunology and Metabolism of Medical Research Institute, Wuhan University, Wuhan, China
| | - Yan Wang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | | | - Sahil Kapoor
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xin Zhang
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Duke Molecular Physiology Institute, Department of Orthopedic Surgery, Duke University, Durham, NC
| | - Manal Elzoheiry
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | - Ezgi Kasimoglu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | - Yisong Wan
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Silva Markovic-Plese
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC;
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| |
Collapse
|
8
|
Yu F, Zhou X, Jin X, Zhao S, Zhao G, Jiang S, Geng S, Wang B. Rational construction of controllable autoimmune diabetes model depicting clinical features. PLoS One 2022; 17:e0260100. [PMID: 35061693 PMCID: PMC8782301 DOI: 10.1371/journal.pone.0260100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022] Open
Abstract
Through animal models, particularly non-obesity diabetes model (NOD), pathological understandings of human autoimmune diabetes have been gained. However, features of those mouse models and the human disease are not sufficiently analogous; it is therefore not unexpected that interventions based on the mouse data fail at an alarming rate in clinical settings. An improvised model that maximally resembles the real pathological course is highly desirable. Here we devised a 'double-hit' strategy, pancreas was first hit by chemical damage (streptozotocin, STZ) to unleash auto-antigens, then hit second time by transient immune-inflammation (regulatory T cell depletion). Comparing to NOD model, this strategy not only induced classical diabetic symptoms, but also depicted the crucial pathogenic features absent in conventional models, such as CD8+ T cell dominant infiltrates, strong ketoacidosis and epitope-specific T cell responses. In addition, this model allowed synchronized control of disease onset, permitting more refined temporal analysis of disease progression. We believe that this model would yield research outcomes with clinically relevant prediction power unattainable previously.
Collapse
Affiliation(s)
- Fan Yu
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xian Zhou
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiang Jin
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shushu Zhao
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Gan Zhao
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Sheng Jiang
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shuang Geng
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, Alberta, Canada
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of MOH and MOE, School of Basic Medical Sciences, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Children’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Bone marrow NG2 +/Nestin + mesenchymal stem cells drive DTC dormancy via TGFβ2. NATURE CANCER 2022; 2:327-339. [PMID: 34993493 DOI: 10.1038/s43018-021-00179-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In the bone marrow (BM) microenvironment, where breast cancer (BC) disseminated tumour cells (DTCs) can remain dormant for decades, NG2+/Nestin+ mesenchymal stem cells (MSCs) promote hematopoietic stem cell quiescence. Here, we reveal that periarteriolar BM-resident NG2+/Nestin+ MSCs can also instruct BC DTCs to enter dormancy. NG2+/Nestin+ MSCs produce TGFβ2 and BMP7 and activate a quiescence pathway dependent on TGFBRIII and BMPRII, which via p38-kinase result in p27 induction. Genetic depletion of MSCs or conditional knock-out of TGFβ2 in MSCs using an NG2-CreER driver led to bone metastatic outgrowth of otherwise dormant p27+/Ki67- DTCs. Also ER+ BC patients without systemic recurrence displayed higher frequency of TGFβ2 and BMP7 detection in the BM. Our results provide a direct proof that HSC dormancy niches control BC DTC dormancy and suggest that aging or extrinsic factors that affect the NG2+/Nestin+ MSC niche homeostasis may result in a break from dormancy and BC bone relapse.
Collapse
|
10
|
Peng W, Wang L, Zhang H, Zhang Z, Chen X. Effects of Recombinant IL-35-BCG on Treg/Th17 Cell Imbalance and Inflammatory Response in Asthmatic Newborn Mice Induced by RSV. Inflammation 2021; 44:2476-2485. [PMID: 34453228 DOI: 10.1007/s10753-021-01517-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 07/07/2021] [Indexed: 11/30/2022]
Abstract
Treg/Th17 cell imbalance and inflammatory response may occur in neonatal asthma. IL-35 and BCG have inhibitory effects on inflammatory responses in diseases. However, studies on neonatal asthma after combination of the two have not been reported so far. A respiratory syncytial virus (RSV)-induced neonatal asthma model was first developed in newborn mice. Pathological sections of lung tissue of asthmatic mice were observed by HE staining. Masson staining was used to observe the lung tissue and to compare the deposition of collagen fibers under bronchial epithelium in model mice. The expression of cytokines in serum was detected by ELISA. Giemsa staining analyzed each cell in bronchoalveolar lavage fluid (BALF). Flow cytometry was used to detect the differentiation and development of Treg and Th17 subgroups in BALF. The expression levels of inflammation-related factors were detected by RT-qPCR. Western blot was used to detect the expression of JNK pathway-related proteins. Recombinant IL-35-BCG improved the pathological response of asthmatic mice; inhibited the expression of IgE in serum, neutrophils, macrophages, and eosinophils in BALF; and increased the expression of lymphocytes. In addition, recombinant IL-35-BCG significantly inhibited Th17 differentiation, promoted Treg cell differentiation, and inhibited the expression of inflammatory factors in lung tissue homogenates, thereby reducing allergic airway inflammation. This process might be achieved by inhibiting the JNK signaling pathway. Recombinant IL-35-BCG can regulate Treg/Th17 cell imbalance and inflammatory response in asthmatic newborn mice induced by RSV through JNK signaling pathway, suggesting a new path to neonatal asthma treatment.
Collapse
Affiliation(s)
- Wansheng Peng
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, China
| | - Lian Wang
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, China
| | - Hui Zhang
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, China
| | - Zhen Zhang
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, China
| | - Xin Chen
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, China.
| |
Collapse
|
11
|
Estrada Brull A, Rost F, Oderbolz J, Kirchner FR, Leibundgut-Landmann S, Oxenius A, Joller N. CD85k Contributes to Regulatory T Cell Function in Chronic Viral Infections. Int J Mol Sci 2020; 22:E31. [PMID: 33375121 PMCID: PMC7792974 DOI: 10.3390/ijms22010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) prevent excessive immune responses and limit immune pathology upon infections. To fulfill this role in different immune environments elicited by different types of pathogens, Tregs undergo functional specialization into distinct subsets. During acute type 1 immune responses, type 1 Tregs are induced and recruited to the site of ongoing Th1 responses to efficiently control Th1 responses. However, whether a similar specialization process also takes place following chronic infections is still unknown. In this study, we investigated Treg specialization in persistent viral infections using lymphocytic choriomeningitis virus (LCMV) and murine cytomegalovirus (MCMV) infection as models for chronic and latent infections, respectively. We identify CD85k as a Th1-specific co-inhibitory receptor with sustained expression in persistent viral infections and show that recombinant CD85k inhibits LCMV-specific effector T cells. Furthermore, expression of the CD85k ligand ALCAM is induced on LCMV-specific and exhausted T cells during chronic LCMV infection. Finally, we demonstrate that type 1 Tregs arising during chronic LCMV infection suppress Th1 effector cells in an ALCAM-dependent manner. These results extend the current knowledge of Treg specialization from acute to persistent viral infections and reveal an important functional role of CD85k in Treg-mediated suppression of type 1 immunity.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cell Adhesion Molecules, Neuronal/immunology
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line
- Cells, Cultured
- Herpesviridae Infections/immunology
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/virology
- Lymphocytic Choriomeningitis/immunology
- Lymphocytic Choriomeningitis/metabolism
- Lymphocytic Choriomeningitis/virology
- Lymphocytic choriomeningitis virus/immunology
- Lymphocytic choriomeningitis virus/physiology
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice, Inbred C57BL
- Muromegalovirus/immunology
- Muromegalovirus/physiology
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/virology
- Th1 Cells/immunology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Anna Estrada Brull
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| | - Felix Rost
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| | - Josua Oderbolz
- ETH Zurich, Institute of Microbiology, 8093 Zurich, Switzerland; (J.O.); (A.O.)
| | - Florian R. Kirchner
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
- Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Salomé Leibundgut-Landmann
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
- Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Annette Oxenius
- ETH Zurich, Institute of Microbiology, 8093 Zurich, Switzerland; (J.O.); (A.O.)
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| |
Collapse
|
12
|
Camacho V, Matkins VR, Patel SB, Lever JM, Yang Z, Ying L, Landuyt AE, Dean EC, George JF, Yang H, Ferrell PB, Maynard CL, Weaver CT, Turnquist HR, Welner RS. Bone marrow Tregs mediate stromal cell function and support hematopoiesis via IL-10. JCI Insight 2020; 5:135681. [PMID: 33208555 PMCID: PMC7710301 DOI: 10.1172/jci.insight.135681] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 10/07/2020] [Indexed: 12/31/2022] Open
Abstract
The nonimmune roles of Tregs have been described in various tissues, including the BM. In this study, we comprehensively phenotyped marrow Tregs, elucidating their key features and tissue-specific functions. We show that marrow Tregs are migratory and home back to the marrow. For trafficking, marrow Tregs use S1P gradients, and disruption of this axis allows for specific targeting of the marrow Treg pool. Following Treg depletion, the function and phenotype of both mesenchymal stromal cells (MSCs) and hematopoietic stem cells (HSCs) was impaired. Transplantation also revealed that a Treg-depleted niche has a reduced capacity to support hematopoiesis. Finally, we found that marrow Tregs are high producers of IL-10 and that Treg-secreted IL-10 has direct effects on MSC function. This is the first report to our knowledge revealing that Treg-secreted IL-10 is necessary for stromal cell maintenance, and our work outlines an alternative mechanism by which this cytokine regulates hematopoiesis.
Collapse
Affiliation(s)
| | | | | | - Jeremie M. Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, and
| | - Zhengqin Yang
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li Ying
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Ashley E. Landuyt
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Emma C. Dean
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James F. George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Henry Yang
- Cancer Science Institute of Singapore & Department of Biochemistry, National University of Singapore, Singapore
| | - Paul Brent Ferrell
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey T. Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Heth R. Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
13
|
Regulatory T Cells Contribute to Resistance against Lyme Arthritis. Infect Immun 2020; 88:IAI.00160-20. [PMID: 32778610 DOI: 10.1128/iai.00160-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/30/2020] [Indexed: 12/17/2022] Open
Abstract
The symptoms of Lyme disease are caused by inflammation induced by species of the Borrelia burgdorferi sensu lato complex. The various presentations of Lyme disease in the population suggest that differences exist in the intensity and regulation of the host response to the spirochete. Previous work has described correlations between the presence of regulatory T cells and recovery from Lyme arthritis. However, the effects of Foxp3-expressing CD4+ T cells existing prior to, and during, B. burgdorferi infection have not been well characterized. Here, we used C57BL/6 "depletion of regulatory T cell" mice to assess the effects these cells have on the arthritis-resistant phenotype characteristic of this mouse strain. We showed that depletion of regulatory T cells prior to infection with B. burgdorferi resulted in sustained swelling, as well as histopathological changes, of the tibiotarsal joints that were not observed in infected control mice. Additionally, in vitro stimulation of splenocytes from these regulatory T cell-depleted mice resulted in increases in gamma interferon and interleukin-17 production and decreases in interleukin-10 production that were not evident among splenocytes of infected mice in which Treg cells were not depleted. Depletion of regulatory T cells at various times after infection also induced rapid joint swelling. Collectively, these findings provide evidence that regulatory T cells existing at the time of, and possibly after, B. burgdorferi infection may play an important role in limiting the development of arthritis.
Collapse
|
14
|
Purde V, Kudryashova E, Heisler DB, Shakya R, Kudryashov DS. Intein-mediated cytoplasmic reconstitution of a split toxin enables selective cell ablation in mixed populations and tumor xenografts. Proc Natl Acad Sci U S A 2020; 117:22090-22100. [PMID: 32839344 PMCID: PMC7486740 DOI: 10.1073/pnas.2006603117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The application of proteinaceous toxins for cell ablation is limited by their high on- and off-target toxicity, severe side effects, and a narrow therapeutic window. The selectivity of targeting can be improved by intein-based toxin reconstitution from two dysfunctional fragments provided their cytoplasmic delivery via independent, selective pathways. While the reconstitution of proteins from genetically encoded elements has been explored, exploiting cell-surface receptors for boosting selectivity has not been attained. We designed a robust splitting algorithm and achieved reliable cytoplasmic reconstitution of functional diphtheria toxin from engineered intein-flanked fragments upon receptor-mediated delivery of one of them to the cells expressing the counterpart. Retargeting the delivery machinery toward different receptors overexpressed in cancer cells enables selective ablation of specific subpopulations in mixed cell cultures. In a mouse model, the transmembrane delivery of a split-toxin construct potently inhibits the growth of xenograft tumors expressing the split counterpart. Receptor-mediated delivery of engineered split proteins provides a platform for precise therapeutic and experimental ablation of tumors or desired cell populations while also greatly expanding the applicability of the intein-based protein transsplicing.
Collapse
Affiliation(s)
- Vedud Purde
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210;
| | - David B Heisler
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210
| | - Reena Shakya
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Dmitri S Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210;
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
15
|
Regulatory T cells in ischemic cardiovascular injury and repair. J Mol Cell Cardiol 2020; 147:1-11. [PMID: 32777294 DOI: 10.1016/j.yjmcc.2020.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 01/03/2023]
Abstract
Ischemic injury triggers a heightened inflammatory response that is essential for tissue repair, but excessive and chronic inflammatory responses contribute to the pathogenesis of ischemic cardiovascular disease. Regulatory T cells (Tregs), a major regulator of self-tolerance and immune suppression, control innate and adaptive immune responses, modulate specific immune cell subsets, prevent excessive inflammation, and participate in tissue repair after ischemia. Herein, we summarize the multiple potential mechanisms by which Tregs exert suppressor functions including modulation of cytokine production, alteration of cell-cell interactions, and disruption of metabolic pathways. Furthermore, we review the role of Tregs implicated in ischemic injury and repair including myocardial, limb, and cerebral ischemia. We conclude with a perspective on the therapeutic opportunities and future challenges of Treg biology in understanding the pathogenesis of ischemic cardiovascular disease states.
Collapse
|
16
|
Li R, Zhang J, Pan S, Yuan Y, Qi H, Shu H, Hu Y, Ren L, Jiang Y, Yuan S. HMGB1 aggravates lipopolysaccharide-induced acute lung injury through suppressing the activity and function of Tregs. Cell Immunol 2020; 356:104192. [PMID: 32853967 DOI: 10.1016/j.cellimm.2020.104192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND CD4+CD25+FoxP3+ T helper cells (Tregs), a subgroup of CD4+ T helper cells, are critical effectors that protect against acute lung injury (ALI) by contact-dependent suppression or releasing anti-inflammatory cytokines including interleukin-10 (IL-10), and transforming growth factor (TGF-β). HMGB1 (High mobility group box 1 protein) was identified as a nuclear non-histone DNA-binding chromosomal protein, which participates in the regulation of lung inflammatory response and pathological processes in ALI. Previous studies have suggested that Tregs overexpresses the HMGB1-recognizing receptor. However, the interaction of HMGB1 with Tregs in ALI is still unclear. OBJECTIVE To investigate whether HMGB1 aggravates ALI by suppressing immunosuppressive function of Tregs. METHODS Anti-HMGB1 antibody and recombinant mouse HMGB1 (rHMGB1) were administered in lipopolysaccharide (LPS)-induced ALI mice and polarized LPS-primed Tregs in vitro. The Tregs pre-stimulated with or without rHMGB1 were adoptively transferred to ALI mice and depleted by Diphtheria toxin (DT). For coculture experiment, isolated Tregs were first pre-stimulated with or without rHMGB1 or anti-HMGB1 antibody, then they were cocultured with bone marrow-derived macrophages (BMMs) under LPS stimulation. RESULTS Tregs protected against acute lung pathological injury. HMGB1 modulated the suppressive function of Tregs as follows: reduction in the number of the cells and the activity of Tregs, the secretion of anti-inflammatory cytokines (IL-10, TGF-β) from Tregs, the production of IL-2 from CD4+ T cells and CD11c+ DCs, and the M2 polarization of macrophages, as well as inducing proinflammatory response of macrophages. CONCLUSIONS HMGB1 could aggravate LPS induced-ALI through suppressing the activity and function of Tregs.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Shangwen Pan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yin Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Hong Qi
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Huaqing Shu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yingying Hu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Lehao Ren
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yongxiang Jiang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Shiying Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
17
|
Liu F, Dai S, Feng D, Peng X, Qin Z, Kearns AC, Huang W, Chen Y, Ergün S, Wang H, Rappaport J, Bryda EC, Chandrasekhar A, Aktas B, Hu H, Chang SL, Gao B, Qin X. Versatile cell ablation tools and their applications to study loss of cell functions. Cell Mol Life Sci 2019; 76:4725-4743. [PMID: 31359086 PMCID: PMC6858955 DOI: 10.1007/s00018-019-03243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Zhongnan Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Yong Chen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, 261053, Weifang, People's Republic of China
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximillan University, 97070, Wurzburg, Germany
| | - Hong Wang
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, 4011 Discovery Drive, Columbia, MO, 65201, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences, 340D Life Sciences Center, University of Missouri, 1201 Rollins St, Columbia, MO, USA
| | - Bertal Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
18
|
Baek H, Yang H, Lee JH, Kang NH, Lee J, Bae H, Hwang DS. Prophylactic Effects of Bee Venom Phospholipase A2 in Lipopolysaccharide-Induced Pregnancy Loss. Toxins (Basel) 2019; 11:toxins11070404. [PMID: 31336883 PMCID: PMC6669565 DOI: 10.3390/toxins11070404] [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] [Received: 06/21/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 11/23/2022] Open
Abstract
Spontaneous abortion represents a common form of embryonic loss caused by early pregnancy failure. In the present study, we investigated the prophylactic effects of bee venom phospholipase A2 (bvPLA2), a regulatory T cell (Treg) inducer, on a lipopolysaccharide (LPS)-induced abortion mouse model. Fetal loss, including viable implants, the fetal resorption rate, and the fetal weight, were measured after LPS and bvPLA2 treatment. The levels of serum and tissue inflammatory cytokines were determined. To investigate the involvement of the Treg population in bvPLA2-mediated protection against fetal loss, the effect of Treg depletion was evaluated following bvPLA2 and LPS treatment. The results clearly revealed that bvPLA2 can prevent fetal loss accompanied by growth restriction in the remaining viable fetus. When the LPS-induced abortion mice were treated with bvPLA2, Treg cells were significantly increased compared with those in the non-pregnant, PBS, and LPS groups. After LPS injection, the levels of proinflammatory cytokines were markedly increased compared with those in the PBS mouse group, while bvPLA2 treatment showed significantly decreased TNF-α and IFN-γ expression compared with that in the LPS group. The protective effects of bvPLA2 treatment were not detected in Treg-depleted abortion-prone mice. These findings suggest that bvPLA2 has protective effects in the LPS-induced abortion mouse model by regulating Treg populations.
Collapse
Affiliation(s)
- Hyunjung Baek
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
| | - HyeJin Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
| | - Jong Hoon Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
| | - Na-Hoon Kang
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea
| | - Jinwook Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea.
| | - Deok-Sang Hwang
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02453, Korea.
| |
Collapse
|
19
|
Tan W, Zhang C, Liu J, Miao Q. Regulatory T-cells promote pulmonary repair by modulating T helper cell immune responses in lipopolysaccharide-induced acute respiratory distress syndrome. Immunology 2019; 157:151-162. [PMID: 30919408 DOI: 10.1111/imm.13060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/07/2019] [Accepted: 03/21/2019] [Indexed: 12/29/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) induces a strong local infiltration of regulatory T-cells (Tregs) in the lungs. However, at present, there remains a lack of adequate evidence showing the direct effect of Tregs on pulmonary repair and the related mechanisms of ARDS. Therefore, in this project, we studied the impact of Tregs on lipopolysaccharide (LPS)-induced ARDS and pulmonary inflammation. Surprisingly, we found that depletion of Tregs by injection of PC61 anti-CD25 antibody not only interfered with the inflammation resolution, such as inhibited total cell infiltration into the alveolar space, downregulated neutrophils, upregulated macrophages, but also impaired pulmonary epithelium and endothelial cell proliferation. Consistent with the attenuation of pulmonary repair, we found that the Th1 and Th17 immune responses were also impaired in Treg-depleted mice, suggesting that the presence of Tregs is vital for tissue repair, as Tregs modulate and promote the Th immune response in LPS-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Wen Tan
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chaoji Zhang
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianzhou Liu
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Miao
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Naran K, Nundalall T, Chetty S, Barth S. Principles of Immunotherapy: Implications for Treatment Strategies in Cancer and Infectious Diseases. Front Microbiol 2018; 9:3158. [PMID: 30622524 PMCID: PMC6308495 DOI: 10.3389/fmicb.2018.03158] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022] Open
Abstract
The advances in cancer biology and pathogenesis during the past two decades, have resulted in immunotherapeutic strategies that have revolutionized the treatment of malignancies, from relatively non-selective toxic agents to specific, mechanism-based therapies. Despite extensive global efforts, infectious diseases remain a leading cause of morbidity and mortality worldwide, necessitating novel, innovative therapeutics that address the current challenges of increasing antimicrobial resistance. Similar to cancer pathogenesis, infectious pathogens successfully fashion a hospitable environment within the host and modulate host metabolic functions to support their nutritional requirements, while suppressing host defenses by altering regulatory mechanisms. These parallels, and the advances made in targeted therapy in cancer, may inform the rational development of therapeutic interventions for infectious diseases. Although "immunotherapy" is habitually associated with the treatment of cancer, this review accentuates the evolving role of key targeted immune interventions that are approved, as well as those in development, for various cancers and infectious diseases. The general features of adoptive therapies, those that enhance T cell effector function, and ligand-based therapies, that neutralize or eliminate diseased cells, are discussed in the context of specific diseases that, to date, lack appropriate remedial treatment; cancer, HIV, TB, and drug-resistant bacterial and fungal infections. The remarkable diversity and versatility that distinguishes immunotherapy is emphasized, consequently establishing this approach within the armory of curative therapeutics, applicable across the disease spectrum.
Collapse
Affiliation(s)
- Krupa Naran
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Trishana Nundalall
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Shivan Chetty
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
21
|
Foxp3 + T Regulatory Cells as a Potential Target for Immunotherapy against Primary Infection with Echinococcus multilocularis Eggs. Infect Immun 2018; 86:IAI.00542-18. [PMID: 30037796 PMCID: PMC6204723 DOI: 10.1128/iai.00542-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/13/2018] [Indexed: 02/08/2023] Open
Abstract
Alveolar echinococcosis (AE) is a lethal disease caused by infection with the metacestode stage of the helminth Echinococcus multilocularis, which develops into a tumorlike mass in susceptible intermediate hosts. The growth potential of this parasite stage is directly linked to the nature of the surrounding periparasitic immune-mediated processes. In a first step (experiment 1), mice were orally infected with E. multilocularis eggs, to be used for assessing the hepatic expression profiles of 15 selected cytokine and chemokine genes related to acquired immunity from 21 to 120 days postinfection. The early stage of infection in immunocompetent animals was marked by a mixed Th1/Th2 immune response, as characterized by the concomitant presence of gamma interferon (IFN-γ) and interleukin-4 (IL-4) and their related chemokines. At the late stage of AE, the profile extended to a combined tolerogenic mode including Foxp3, IL-10, and transforming growth factor beta (TGF-β) as key components. In a second step (experiment 2), the effect of T regulatory cell (Treg) deficiency on metacestode growth was assessed in E. multilocularis-infected DEREG (depletion of regulatory T cells) mice upon induction of Treg deficiency with diphtheria toxin (DT). The parasite lesions were significantly smaller in the livers of treated mice than in corresponding control groups. Foxp3+ Tregs appear to be one of the key players in immune-regulatory processes favoring metacestode survival by affecting antigen presentation and suppressing Th1-type immune responses. For these reasons, we suggest that affecting Foxp3+ Tregs could offer an attractive target in the development of an immunotherapy against AE.
Collapse
|
22
|
Rogers MC, Lamens KD, Shafagati N, Johnson M, Oury TD, Joyce S, Williams JV. CD4 + Regulatory T Cells Exert Differential Functions during Early and Late Stages of the Immune Response to Respiratory Viruses. THE JOURNAL OF IMMUNOLOGY 2018; 201:1253-1266. [PMID: 29997123 DOI: 10.4049/jimmunol.1800096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/19/2018] [Indexed: 01/30/2023]
Abstract
Acute respiratory virus infection (ARI) induces CD8+ T cells with diminished cytokine production and functional impairment. The role of cellular mediators of immune impairment, specifically CD4+ regulatory T cells (Tregs), is incompletely understood in ARI. Tregs are known suppressors of effector T cell function, but whether they are detrimental or beneficial in ARI remains controversial. We show in this paper that Treg depletion leads to increased CD8+ T cell function and lower virus titer in mice infected with human metapneumovirus. We further demonstrate that Tregs play a temporal role in the immune response to human metapneumovirus and influenza: Treg depletion before infection pathologically reduces virus-specific CD8+ T cell numbers and delays virus clearance, whereas depletion 2 d postinoculation enhances CD8+ T cell functionality without reducing virus-specific CD8+ T cell numbers. Mechanistically, Treg depletion during immune priming led to impaired dendritic cell and CD8+ T cell migration. Further, early Treg depletion was associated with immune skewing toward a type 2 phenotype characterized by increased type 2 innate lymphoid cells and TH2 CD4+ T cells, which was not observed when Treg depletion was delayed until after inoculation. These results indicate that the presence of Tregs at inoculation is critical for efficient priming of the CD8+ T cell response to ARI, whereas later in infection, Tregs are dispensable for virus clearance.
Collapse
Affiliation(s)
- Meredith C Rogers
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37212
| | - Kristina D Lamens
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Nazly Shafagati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224
| | - Monika Johnson
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and
| | - Sebastian Joyce
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37212.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212
| | - John V Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224;
| |
Collapse
|
23
|
Wang J, Müller S, Lin R, Siffert M, Vuitton DA, Wen H, Gottstein B. Depletion of FoxP3 + Tregs improves control of larval Echinococcus multilocularis infection by promoting co-stimulation and Th1/17 immunity. IMMUNITY INFLAMMATION AND DISEASE 2017. [PMID: 28621034 PMCID: PMC5691311 DOI: 10.1002/iid3.181] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction The growth potential of the tumor‐like Echinococcus multilocularis metacestode (causing alveolar echinococcosis, AE) is directly linked to the nature/function of the periparasitic host immune‐mediated processes. Previous studies had shown that regulatory T cells (Tregs) become gradually up‐regulated in the course of both chronic human and murine AE. Thus we now tackled the role of FoxP3+ Tregs and FoxP3+‐Treg‐regulated immune response in contributing to the control of this helminthic infection. Methods The infection outcome in E. multilocularis‐infected DEREG mice was measured upon determining parasite load (wet weight of parasitic metacestode tissue). Flow cytometry and qRT‐PCR were used to assess Treg, Th17‐, Th1‐, Th2‐type immune responses and antigen presenting cell activation. Results We showed that E. multilocularis‐infected DEREG‐mice treated with DT (as compared to infected control DEREG‐mice without DT application) exhibited a significantly lower parasite load, associated with a persisting capacity of co‐stimulation, and an increased Th1/Th17‐polarization. Conclusions FoxP3+ Tregs appear as one of the key players in immune regulatory processes favoring (i) metacestode survival by inhibiting the maturation potential of co‐stimulatory activity and (ii) T cell exhaustion (suppressing Th1/Th17‐type immune responses). We showed as well that prospectively, targeting FoxP3+ Tregs could be an option to develop an immunotherapy against AE.
Collapse
Affiliation(s)
- Junhua Wang
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, Institute of Parasitology, University of Bern, Bern, Switzerland.,State Key Lab Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Stephan Müller
- FACSLab, c/o Institute of Pathology, University of Bern, Bern, Switzerland
| | - Renyong Lin
- State Key Lab Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Myriam Siffert
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, Central Animal Facilities, University of Bern, Bern, Switzerland
| | - Dominique A Vuitton
- WHO-Collaborating Centre on Prevention and Treatment of Human Echinococcosis and French National Reference Centre on Alveolar Echinococcosis, University of Franche-Comté and University Hospital, Besançon, France
| | - Hao Wen
- State Key Lab Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bruno Gottstein
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, Institute of Parasitology, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Konishi H, Ohgami N, Matsushita A, Kondo Y, Aoyama Y, Kobayashi M, Nagai T, Ugawa S, Yamada K, Kato M, Kiyama H. Exposure to diphtheria toxin during the juvenile period impairs both inner and outer hair cells in C57BL/6 mice. Neuroscience 2017; 351:15-23. [PMID: 28344071 DOI: 10.1016/j.neuroscience.2017.03.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Abstract
Diphtheria toxin (DT) administration into transgenic mice that express the DT receptor (DTR) under control of specific promoters is often used for cell ablation studies in vivo. Because DTR is not expressed in mice, DT injection has been assumed to be nontoxic to cells in vivo. In this study, we demonstrated that DT application during the juvenile stage leads to hearing loss in wild-type mice. Auditory brainstem response measurement showed severe hearing loss in C57BL/6 mice administered DT during the juvenile period, and the hearing loss persisted into adulthood. However, ototoxicity did not occur when DT was applied on postnatal day 28 or later. Histological studies demonstrated that hearing loss was accompanied by significant degeneration of inner and outer hair cells (HCs), as well as spiral ganglion neurons. Scanning electron microscopy showed quick degeneration of inner HCs within 3days and gradual degeneration of outer HCs within 1week. These results demonstrated that DT has ototoxic action on C57BL/6 mice during the juvenile period, but not thereafter, and the hearing loss was due to degeneration of inner and outer HCs by unknown DT-related mechanisms.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Nobutaka Ohgami
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Nutritional Health Science Research Center, Chubu University, Kasugai 487-8501, Japan.
| | - Aika Matsushita
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Yuki Kondo
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Yuki Aoyama
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Masaaki Kobayashi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Shinya Ugawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
25
|
Dombrowski Y, O'Hagan T, Dittmer M, Penalva R, Mayoral SR, Bankhead P, Fleville S, Eleftheriadis G, Zhao C, Naughton M, Hassan R, Moffat J, Falconer J, Boyd A, Hamilton P, Allen IV, Kissenpfennig A, Moynagh PN, Evergren E, Perbal B, Williams AC, Ingram RJ, Chan JR, Franklin RJM, Fitzgerald DC. Regulatory T cells promote myelin regeneration in the central nervous system. Nat Neurosci 2017; 20:674-680. [PMID: 28288125 PMCID: PMC5409501 DOI: 10.1038/nn.4528] [Citation(s) in RCA: 348] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/03/2017] [Indexed: 02/08/2023]
Abstract
Regeneration of CNS myelin involves differentiation of oligodendrocytes from oligodendrocyte progenitor cells. In multiple sclerosis, remyelination can fail despite abundant oligodendrocyte progenitor cells, suggesting impairment of oligodendrocyte differentiation. T cells infiltrate the CNS in multiple sclerosis, yet little is known about T cell functions in remyelination. We report that regulatory T cells (Treg) promote oligodendrocyte differentiation and (re)myelination. Treg-deficient mice exhibited substantially impaired remyelination and oligodendrocyte differentiation, which was rescued by adoptive transfer of Treg. In brain slice cultures, Treg accelerated developmental myelination and remyelination, even in the absence of overt inflammation. Treg directly promoted oligodendrocyte progenitor cell differentiation and myelination in vitro. We identified CCN3 as a Treg-derived mediator of oligodendrocyte differentiation and myelination in vitro. These findings reveal a new regenerative function of Treg in the CNS, distinct from immunomodulation. Although the cells were originally named 'Treg' to reflect immunoregulatory roles, this also captures emerging, regenerative Treg functions.
Collapse
Affiliation(s)
- Yvonne Dombrowski
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Thomas O'Hagan
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Marie Dittmer
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Rosana Penalva
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Sonia R Mayoral
- Department of Neurology and Program in Neurosciences, University of California, San Francisco, California, USA
| | - Peter Bankhead
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Samara Fleville
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - George Eleftheriadis
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Clifford Allbutt Building, Cambridge Biomedical Campus, University of Cambridge, UK
| | - Michelle Naughton
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Rachel Hassan
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Jill Moffat
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - John Falconer
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Amanda Boyd
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Peter Hamilton
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Ingrid V Allen
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Adrien Kissenpfennig
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Paul N Moynagh
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK.,Institute of Immunology, Department of Biology, National University of Ireland Maynooth, Ireland
| | - Emma Evergren
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Bernard Perbal
- Université Côte d'Azur, CNRS, GREDEG, Nice, France.,International CCN Society, Paris, France
| | - Anna C Williams
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Rebecca J Ingram
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Jonah R Chan
- Department of Neurology and Program in Neurosciences, University of California, San Francisco, California, USA
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Clifford Allbutt Building, Cambridge Biomedical Campus, University of Cambridge, UK
| | - Denise C Fitzgerald
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| |
Collapse
|
26
|
Fisher SA, Aston WJ, Chee J, Khong A, Cleaver AL, Solin JN, Ma S, Lesterhuis WJ, Dick I, Holt RA, Creaney J, Boon L, Robinson B, Lake RA. Transient Treg depletion enhances therapeutic anti-cancer vaccination. Immun Inflamm Dis 2017; 5:16-28. [PMID: 28250921 PMCID: PMC5322183 DOI: 10.1002/iid3.136] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Regulatory T cells (Treg) play an important role in suppressing anti- immunity and their depletion has been linked to improved outcomes. To better understand the role of Treg in limiting the efficacy of anti-cancer immunity, we used a Diphtheria toxin (DTX) transgenic mouse model to specifically target and deplete Treg. METHODS Tumor bearing BALB/c FoxP3.dtr transgenic mice were subjected to different treatment protocols, with or without Treg depletion and tumor growth and survival monitored. RESULTS DTX specifically depleted Treg in a transient, dose-dependent manner. Treg depletion correlated with delayed tumor growth, increased effector T cell (Teff) activation, and enhanced survival in a range of solid tumors. Tumor regression was dependent on Teffs as depletion of both CD4 and CD8 T cells completely abrogated any survival benefit. Severe morbidity following Treg depletion was only observed, when consecutive doses of DTX were given during peak CD8 T cell activation, demonstrating that Treg can be depleted on multiple occasions, but only when CD8 T cell activation has returned to base line levels. Finally, we show that even minimal Treg depletion is sufficient to significantly improve the efficacy of tumor-peptide vaccination. CONCLUSIONS BALB/c.FoxP3.dtr mice are an ideal model to investigate the full therapeutic potential of Treg depletion to boost anti-tumor immunity. DTX-mediated Treg depletion is transient, dose-dependent, and leads to strong anti-tumor immunity and complete tumor regression at high doses, while enhancing the efficacy of tumor-specific vaccination at low doses. Together this data highlight the importance of Treg manipulation as a useful strategy for enhancing current and future cancer immunotherapies.
Collapse
Affiliation(s)
- Scott A. Fisher
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Wayne J. Aston
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Jonathan Chee
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Andrea Khong
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Amanda L. Cleaver
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Jessica N. Solin
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Shaokang Ma
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - W. Joost Lesterhuis
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Ian Dick
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Robert A. Holt
- British Columbia Cancer AgencyVancouverBritish ColumbiaCanada
| | - Jenette Creaney
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | | | - Bruce Robinson
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Richard A. Lake
- School of Medicine and PharmacologyUniversity of Western Australia, QEII Medical CentreNedlandsWestern AustraliaAustralia
- National Research Centre for Asbestos Related DiseasesQEII Medical CentreNedlandsWestern AustraliaAustralia
| |
Collapse
|
27
|
Glatman Zaretsky A, Konradt C, Dépis F, Wing JB, Goenka R, Atria DG, Silver JS, Cho S, Wolf AI, Quinn WJ, Engiles JB, Brown DC, Beiting D, Erikson J, Allman D, Cancro MP, Sakaguchi S, Lu LF, Benoist CO, Hunter CA. T Regulatory Cells Support Plasma Cell Populations in the Bone Marrow. Cell Rep 2017; 18:1906-1916. [PMID: 28228257 PMCID: PMC5361408 DOI: 10.1016/j.celrep.2017.01.067] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 11/20/2016] [Accepted: 01/25/2017] [Indexed: 01/07/2023] Open
Abstract
Long-lived plasma cells (PCs) in the bone marrow (BM) are a critical source of antibodies after infection or vaccination, but questions remain about the factors that control PCs. We found that systemic infection alters the BM, greatly reducing PCs and regulatory T (Treg) cells, a population that contributes to immune privilege in the BM. The use of intravital imaging revealed that BM Treg cells display a distinct behavior characterized by sustained co-localization with PCs and CD11c-YFP+ cells. Gene expression profiling indicated that BM Treg cells express high levels of Treg effector molecules, and CTLA-4 deletion in these cells resulted in elevated PCs. Furthermore, preservation of Treg cells during systemic infection prevents PC loss, while Treg cell depletion in uninfected mice reduced PC populations. These studies suggest a role for Treg cells in PC biology and provide a potential target for the modulation of PCs during vaccine-induced humoral responses or autoimmunity.
Collapse
Affiliation(s)
| | - Christoph Konradt
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fabien Dépis
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - James B Wing
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Radhika Goenka
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniela Gomez Atria
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan S Silver
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sunglim Cho
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Amaya I Wolf
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - William J Quinn
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julie B Engiles
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dorothy C Brown
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Beiting
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jan Erikson
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - David Allman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Cancro
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Li-Fan Lu
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christophe O Benoist
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher A Hunter
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Rombouts M, Cools N, Grootaert MOJ, de Bakker F, Van Brussel I, Wouters A, De Meyer GRY, De Winter BY, Schrijvers DM. Long-Term Depletion of Conventional Dendritic Cells Cannot Be Maintained in an Atherosclerotic Zbtb46-DTR Mouse Model. PLoS One 2017; 12:e0169608. [PMID: 28060909 PMCID: PMC5218565 DOI: 10.1371/journal.pone.0169608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/18/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND AND AIMS Increased evidence suggests a pro-atherogenic role for conventional dendritic cells (cDC). However, due to the lack of an exclusive marker for cDC, their exact contribution to atherosclerosis remains elusive. Recently, a unique transcription factor was described for cDC, namely Zbtb46, enabling us to selectively target this cell type in mice. METHODS Low-density lipoprotein receptor-deficient (Ldlr-/-) mice were transplanted with bone marrow from Zbtb46-diphtheria toxin receptor (DTR) transgenic mice following total body irradiation. Zbtb46-DTR→Ldlr-/- chimeras were fed a Western-type diet for 18 weeks while cDC were depleted by administering diphtheria toxin (DT). RESULTS Although we confirmed efficient direct induction of cDC death in vitro and in vivo upon DT treatment of Zbtb46-DTR mice, advanced atherosclerotic plaque size and composition was not altered. Surprisingly, however, analysis of Zbtb46-DTR→Ldlr-/- chimeras showed that depletion of cDC was not sustained following 18 weeks of DT treatment. In contrast, high levels of anti-DT antibodies were detected. CONCLUSIONS Because of the observed generation of anti-DT antibodies and consequently the partial depletion of cDC, no clear decision can be taken on the role of cDC in atherosclerosis. Our results underline the unsuitability of Zbtb46-DTR→Ldlr-/- mice for studying the involvement of cDC in maintaining the disease process of atherosclerosis, as well as of other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Miche Rombouts
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Mandy O. J. Grootaert
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Flore de Bakker
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Ilse Van Brussel
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - An Wouters
- Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y. De Winter
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Dorien M. Schrijvers
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
29
|
Repeated Long-Term DT Application in the DEREG Mouse Induces a Neutralizing Anti-DT Antibody Response. J Immunol Res 2016; 2016:1450398. [PMID: 28074191 PMCID: PMC5198145 DOI: 10.1155/2016/1450398] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/26/2016] [Accepted: 11/08/2016] [Indexed: 11/29/2022] Open
Abstract
Regulatory T (Tregs) cells play an important role in mediating tolerance to self-antigens but can also mediate detrimental tolerance to tumours and pathogens in a Foxp3-dependent manner. Genetic tools exploiting the foxp3 locus including bacterial artificial chromosome- (BAC-) transgenic DEpletion of REGulatory T cells (DEREG) mice have provided essential information on Treg biology and the potential therapeutic modulation of tolerance. In DEREG mice, Foxp3+ Tregs selectively express enhanced green fluorescent protein (eGFP) and diphtheria toxin (DT) receptor, allowing for the specific depletion of Tregs through DT administration. We here provide a detailed overview about an important consideration that long-term administration of DT induces a humoral immune response with an appropriate production of anti-DT antibodies that can inactivate DT and thus abrogate its effect in the DEREG mouse. Additionally, we showed that anti-DT mouse serum partially neutralized DT-induced Foxp3 inhibition.
Collapse
|
30
|
Christiaansen AF, Syed MA, Ten Eyck PP, Hartwig SM, Durairaj L, Kamath SS, Varga SM. Altered Treg and cytokine responses in RSV-infected infants. Pediatr Res 2016; 80:702-709. [PMID: 27486703 PMCID: PMC6215710 DOI: 10.1038/pr.2016.130] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in children under 1 y of age in the USA. The host immune response is believed to contribute to RSV-induced disease. We hypothesize that severe RSV infection in infants is mediated by insufficient regulation of the host immune response of regulatory T cells (Tregs) resulting in immunopathology. METHODS Blood and nasal aspirates from 23 RSV-infected and 17 control infants under 1 y of age were collected. Treg frequencies were determined by flow cytometry from peripheral blood mononuclear cells. Analysis of 24 cytokines was measured by multiplex assay on nasal aspirates. RESULTS We demonstrate that the frequency of activated Tregs is significantly reduced in the peripheral blood of RSV-infected infants compared with age-matched controls. Surprisingly, T helper (Th)17 related cytokines including interleukin (IL)-1β, IL-17A, and IL-23 were associated with a reduction in clinical symptoms of respiratory distress. In addition, the amount of IL-33 protein in nasal washes, a cytokine important in maintaining Treg homeostasis in mucosal tissues, was decreased in RSV-infected children. CONCLUSION These results suggest that decreased Treg numbers and an inability to properly control the host inflammatory response results in severe RSV infection.
Collapse
Affiliation(s)
| | | | - Patrick P. Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA
| | | | - Lakshmi Durairaj
- Department of Internal Medicine, University of Iowa, Iowa City, IA
| | | | - Steven M. Varga
- Department of Microbiology, University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
- Department of Pathology, University of Iowa, Iowa City, IA
| |
Collapse
|
31
|
Goudin N, Chappert P, Mégret J, Gross DA, Rocha B, Azogui O. Depletion of Regulatory T Cells Induces High Numbers of Dendritic Cells and Unmasks a Subset of Anti-Tumour CD8+CD11c+ PD-1lo Effector T Cells. PLoS One 2016; 11:e0157822. [PMID: 27341421 PMCID: PMC4920347 DOI: 10.1371/journal.pone.0157822] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/06/2016] [Indexed: 12/20/2022] Open
Abstract
Natural regulatory T (Treg) cells interfere with multiple functions, which are crucial for the development of strong anti-tumour responses. In a model of 4T1 mammary carcinoma, depletion of CD25+Tregs results in tumour regression in Balb/c mice, but the mechanisms underlying this process are not fully understood. Here, we show that partial Treg depletion leads to the generation of a particular effector CD8 T cell subset expressing CD11c and low level of PD-1 in tumour draining lymph nodes. These cells have the capacity to migrate into the tumour, to kill DCs, and to locally regulate the anti-tumour response. These events are concordant with a substantial increase in CD11b+ resident dendritic cells (DCs) subsets in draining lymph nodes followed by CD8+ DCs. These results indicate that Treg depletion leads to tumour regression by unmasking an increase of DC subsets as a part of a program that optimizes the microenvironment by orchestrating the activation, amplification, and migration of high numbers of fully differentiated CD8+CD11c+PD1lo effector T cells to the tumour sites. They also indicate that a critical pattern of DC subsets correlates with the evolution of the anti-tumour response and provide a template for Treg depletion and DC-based therapy.
Collapse
Affiliation(s)
- Nicolas Goudin
- Plateau technique de Cytometrie et d’Imagerie Cellulaire, Structure Fédérative de Recherche Necker, INSERM US 24-CNRS, UMS 3633, Paris, France
| | - Pascal Chappert
- Institut Necker Enfants Malades, INSERM U1151, CNRS, UMR8253, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jérome Mégret
- Plateau technique de Cytometrie et d’Imagerie Cellulaire, Structure Fédérative de Recherche Necker, INSERM US 24-CNRS, UMS 3633, Paris, France
| | - David-Alexandre Gross
- Institut Necker Enfants Malades, INSERM U1151, CNRS, UMR8253, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benedita Rocha
- Institut Necker Enfants Malades, INSERM U1151, CNRS, UMR8253, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Orly Azogui
- Institut Necker Enfants Malades, INSERM U1151, CNRS, UMR8253, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
32
|
Feng D, Dai S, Liu F, Ohtake Y, Zhou Z, Wang H, Zhang Y, Kearns A, Peng X, Zhu F, Hayat U, Li M, He Y, Xu M, Zhao C, Cheng M, Zhang L, Wang H, Yang X, Ju C, Bryda EC, Gordon J, Khalili K, Hu W, Li S, Qin X, Gao B. Cre-inducible human CD59 mediates rapid cell ablation after intermedilysin administration. J Clin Invest 2016; 126:2321-33. [PMID: 27159394 DOI: 10.1172/jci84921] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/03/2016] [Indexed: 11/17/2022] Open
Abstract
Cell ablation is a powerful tool for studying cell lineage and/or function; however, current cell-ablation models have limitations. Intermedilysin (ILY), a cytolytic pore-forming toxin that is secreted by Streptococcus intermedius, lyses human cells exclusively by binding to the human complement regulator CD59 (hCD59), but does not react with CD59 from nonprimates. Here, we took advantage of this feature of ILY and developed a model of conditional and targeted cell ablation by generating floxed STOP-CD59 knockin mice (ihCD59), in which expression of human CD59 only occurs after Cre-mediated recombination. The administration of ILY to ihCD59+ mice crossed with various Cre-driver lines resulted in the rapid and specific ablation of immune, epithelial, or neural cells without off-target effects. ILY had a large pharmacological window, which allowed us to perform dose-dependent studies. Finally, the ILY/ihCD59-mediated cell-ablation method was tested in several disease models to study immune cell functionalities, hepatocyte and/or biliary epithelial damage and regeneration, and neural cell damage. Together, the results of this study demonstrate the utility of the ihCD59 mouse model for studying the effects of cell ablation in specific organ systems in a variety of developmental and disease states.
Collapse
|
33
|
Amiezer M, Phan TG. Disentangling Tfr cells from Treg cells and Tfh cells: How to untie the Gordian knot. Eur J Immunol 2016; 46:1101-4. [PMID: 27109022 DOI: 10.1002/eji.201646389] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 03/18/2016] [Accepted: 03/29/2016] [Indexed: 01/16/2023]
Abstract
T follicular regulatory (Tfr) cells are a subpopulation of Treg cells that have adopted the T follicular helper cell program to localize to the B-cell follicle. Because of the difficulties in generating mouse models in which Tfr cells are selectively affected, determining where and how Tfr cells regulate the germinal center response remains to be resolved. In this issue of the European Journal of Immunology, Dent and colleagues [Eur. J. Immunol. 2016. 46: 1152-1161] describe a simple, elegant mouse model to conditionally delete Tfr cells without impacting on the Treg- and Tfh-cell populations. Their initial studies suggest that Tfr cells have a more complex role than previously thought, particularly with respect to the regulation of immunoglobulin isotype switching to IgA.
Collapse
Affiliation(s)
- Mayan Amiezer
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Darlinghurst, Australia
| | - Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Australia, Darlinghurst, Australia
| |
Collapse
|
34
|
Low-level regulatory T-cell activity is essential for functional type-2 effector immunity to expel gastrointestinal helminths. Mucosal Immunol 2016; 9:428-43. [PMID: 26286232 PMCID: PMC4677460 DOI: 10.1038/mi.2015.73] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 06/26/2015] [Indexed: 02/04/2023]
Abstract
Helminth infection is frequently associated with the expansion of regulatory T cells (Tregs) and suppression of immune responses to bystander antigens. We show that infection of mice with the chronic gastrointestinal helminth Heligmosomoides polygyrus drives rapid polyclonal expansion of Foxp3(+)Helios(+)CD4(+) thymic (t)Tregs in the lamina propria and mesenteric lymph nodes while Foxp3(+)Helios(-)CD4(+) peripheral (p)Treg expand more slowly. Notably, in partially resistant BALB/c mice parasite survival positively correlates with Foxp3(+)Helios(+)CD4(+) tTreg numbers. Boosting of Foxp3(+)Helios(+)CD4(+) tTreg populations by administration of recombinant interleukin-2 (rIL-2):anti-IL-2 (IL-2C) complex increased worm persistence by diminishing type-2 responsiveness in vivo, including suppression of alternatively activated macrophage and granulomatous responses at the sites of infection. IL-2C also increased innate lymphoid cell (ILC) numbers, indicating that Treg functions dominate over ILC effects in this setting. Surprisingly, complete removal of Tregs in transgenic Foxp3-DTR mice also resulted in increased worm burdens, with "immunological chaos" evident in high levels of the pro-inflammatory cytokines IL-6 and interferon-γ. In contrast, worm clearance could be induced by anti-CD25 antibody-mediated partial depletion of early Treg, alongside increased T helper type 2 responses and without incurring pathology. These findings highlight the overarching importance of the early Treg response to infection and the non-linear association between inflammation and the prevailing Treg frequency.
Collapse
|
35
|
Richert-Spuhler LE, Lund JM. The Immune Fulcrum: Regulatory T Cells Tip the Balance Between Pro- and Anti-inflammatory Outcomes upon Infection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:217-43. [PMID: 26615099 DOI: 10.1016/bs.pmbts.2015.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) are indispensable for immune homeostasis and the prevention of autoimmunity. In the context of infectious diseases, Tregs are multidimensional. Here, we describe how they may potentiate effector responses by assisting in recruitment of T cells into the infection site to resolve infection, facilitate accelerated antigen-specific memory responses, limit pathology, and contribute to disease resolution and healing, to the great benefit of the host. We also explore the villainous functions of Tregs during infection by reviewing several diseases in which the depletion or reduction in Treg frequency allows for better generation of effector memory, and results in acute resolution of infection, as opposed to chronicity or severe long-term outcomes. We describe findings generated using mouse models of infection as well as experiments performed using human cells and tissues. We propose that Tregs represent an immunologic fulcrum, promoting both pathogen clearance and damage control by preventing excessive destruction of infected tissues though unchecked immune responses.
Collapse
Affiliation(s)
- Laura E Richert-Spuhler
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jennifer M Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Department of Global Health, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
36
|
Mandl M, Drechsler M, Jansen Y, Neideck C, Noels H, Faussner A, Soehnlein O, Weber C, Döring Y. Evaluation of the BDCA2-DTR Transgenic Mouse Model in Chronic and Acute Inflammation. PLoS One 2015; 10:e0134176. [PMID: 26252890 PMCID: PMC4529211 DOI: 10.1371/journal.pone.0134176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND AIMS Plasmacytoid dendritic cells (pDCs) are a small subset of dendritic cells and the main producers of type I interferons. Besides their contribution to tolerance, they are known to be involved in autoimmune diseases and have recently been implicated in atherosclerosis. However, their precise involvement, particularly in advanced lesion development, remains elusive. Hence, we investigated the role of pDCs in atherogenesis vs atheroprogression by specifically depleting this cell population using the BDCA2-DTR mouse model bred to Apolipoprotein E (Apoe-/-) deficient mice. METHODS AND RESULTS Our results revealed that continuous diphtheria toxin-induced pDC depletion in Apoe-/- BDCA2-DTR mice receiving a high-fat diet (HFD) for 4 weeks did not alter lesion size or composition. Instead, these mice displayed increased B cell numbers and altered levels of inflammatory cytokines. Analysis of depletion efficiency showed that complete pDC depletion could only be sustained for one week and reoccurring pDCs sorted after 4 weeks did not express DTR anymore. Consequently, we analyzed lesion development in a model of partial carotid ligation, inducing established lesions after 5 weeks of HFD feeding, and only depleted pDCs during the last week of 5 weeks HFD feeding. Despite short-term, but efficient pDC depletion, we observed no differences in atherosclerotic lesion development, but changes in inflammatory cytokine titers. To assure the functionality of the BDCA2-DTR model in acute settings, we additionally examined the effect of pDC depletion in an indirect acute lung injury (iALI) model. This time, efficient pDC depletion resulted in a significantly reduced macrophage and neutrophil accumulation in the lung 12 hours after LPS challenge, underlining a pro-inflammatory role of pDCs in the innate immune response in iALI. CONCLUSION Taken together, the BDCA2-DTR mouse model only allows efficient pDC depletion for one week, which subsequently restricts its usability to more acute but not chronic inflammatory disease models.
Collapse
Affiliation(s)
- Manuela Mandl
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maik Drechsler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; Academic Medical Center, Department of Pathology, Amsterdam University, Amsterdam, the Netherlands; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Yvonne Jansen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Carlos Neideck
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Aachen, Aachen, Germany
| | - Alexander Faussner
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; Academic Medical Center, Department of Pathology, Amsterdam University, Amsterdam, the Netherlands; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
37
|
McSorley HJ, Blair NF, Robertson E, Maizels RM. Suppression of OVA-alum induced allergy by Heligmosomoides polygyrus products is MyD88-, TRIF-, regulatory T- and B cell-independent, but is associated with reduced innate lymphoid cell activation. Exp Parasitol 2015; 158:8-17. [PMID: 25728231 DOI: 10.1016/j.exppara.2015.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/09/2015] [Accepted: 02/22/2015] [Indexed: 02/06/2023]
Abstract
The murine intestinal nematode Heligmosomoides polygyrus exerts multiple immunomodulatory effects in the host, including the suppression of allergic inflammation in mice sensitized to allergen presented with alum adjuvant. Similar suppression is attained by co-administration of H. polygyrus excretory/secretory products (HES) with the sensitizing dose of ovalbumin (OVA) in alum. We investigated the mechanism of suppression by HES in this model, and found it was maintained in MyD88xTRIF-deficient mice, implying no role for helminth- or host-derived TLR ligands, or IL-1 family cytokines that signal in a MyD88- or TRIF-dependent manner. We also found suppression was unchanged in µMT mice, which lack B2 B cells, and that suppression was not abrogated when regulatory T cells were depleted in Foxp3.LuciDTR-4 mice. However, reduced IL-5 production was seen in the first 12 h after injection of OVA-alum when HES was co-administered, associated with reduced activation of IL-5(+) and IL-13(+) group 2 innate lymphoid cells. Thus, the suppressive effects of HES on alum-mediated OVA sensitization are reflected in the very earliest innate response to allergen exposure in vivo.
Collapse
Affiliation(s)
- Henry J McSorley
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK; Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Natalie F Blair
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| | - Elaine Robertson
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| | - Rick M Maizels
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| |
Collapse
|
38
|
Schulze B, Piehler D, Eschke M, von Buttlar H, Köhler G, Sparwasser T, Alber G. CD4(+) FoxP3(+) regulatory T cells suppress fatal T helper 2 cell immunity during pulmonary fungal infection. Eur J Immunol 2014; 44:3596-604. [PMID: 25187063 DOI: 10.1002/eji.201444963] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/22/2014] [Accepted: 09/01/2014] [Indexed: 11/11/2022]
Abstract
The opportunistic fungal pathogen Cryptococcus neoformans causes lung inflammation and fatal meningitis in immunocompromised patients. Regulatory T (Treg) cells play an important role in controlling immunity and homeostasis. However, their functional role during fungal infection is largely unknown. In this study, we investigated the role of Treg cells during experimental murine pulmonary C. neoformans infection. We show that the number of CD4(+) FoxP3(+) Treg cells in the lung increases significantly within the first 4 weeks after intranasal infection of BALB/c wild-type mice. To define the function of Treg cells we used DEREG mice allowing selective depletion of CD4(+) FoxP3(+) Treg cells by application of diphtheria toxin. In Treg cell-depleted mice, stronger pulmonary allergic inflammation with enhanced mucus production and pronounced eosinophilia, increased IgE production, and elevated fungal lung burden were found. This was accompanied by higher frequencies of GATA-3(+) T helper (Th) 2 cells with elevated capacity to produce interleukin (IL)-4, IL-5, and IL-13. In contrast, only a mild increase in the Th1-associated immune response unrelated to the fungal infection was observed. In conclusion, the data demonstrate that during fungal infection pulmonary Treg cells are induced and preferentially suppress Th2 cells thereby mediating enhanced fungal control.
Collapse
Affiliation(s)
- Bianca Schulze
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Mayer CT, Lahl K, Milanez-Almeida P, Watts D, Dittmer U, Fyhrquist N, Huehn J, Kopf M, Kretschmer K, Rouse B, Sparwasser T. Advantages of Foxp3(+) regulatory T cell depletion using DEREG mice. IMMUNITY INFLAMMATION AND DISEASE 2014; 2:162-5. [PMID: 25505550 PMCID: PMC4257761 DOI: 10.1002/iid3.33] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/12/2014] [Indexed: 12/17/2022]
Abstract
Several mechanisms enable immunological self-tolerance. Regulatory T cells (Tregs) are a specialized T cell subset that prevents autoimmunity and excessive immune responses, but can also mediate detrimental tolerance to tumors and pathogens in a Foxp3-dependent manner. Genetic tools exploiting the foxp3 locus including bacterial artificial chromosome (BAC)-transgenic DEREG mice have provided essential information on Treg biology and the potential therapeutic modulation of tolerance. In DEREG mice, Foxp3(+) Tregs selectively express eGFP and diphtheria toxin (DT) receptor, allowing for the specific depletion of Tregs through DT administration. We here provide a detailed overview about important considerations such as DT toxicity, which affects any mouse strain treated with DT, and Treg rebound after depletion. Additionally, we point out the specific advantages of BAC-transgenic DEREG mice including their suitability to study organ-specific autoimmunity such as type I diabetes. Moreover, we discuss recent insights into the role of Tregs in viral infections. In summary, DEREG mice are an important tool to study Treg-mediated tolerance and its therapeutic circumvention.
Collapse
Affiliation(s)
- Christian T Mayer
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Katharina Lahl
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine Lane Building, Mailcode 5324, Stanford, CA, 94305, USA ; The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
| | - Pedro Milanez-Almeida
- Experimental Immunology, Helmholtz Centre for Infection Research Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Deepika Watts
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden Fetscherstr. 105, 01307, Dresden, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen Virchowstr.179, 45122, Essen, Germany
| | - Nanna Fyhrquist
- Unit of Systems Toxicology, Finnish Institute of Occupational Health Topeliuksenkatu 41b, 00250, Helsinki, Finland
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Manfred Kopf
- Institute for Molecular Health Sciences, Swiss Federal Institute of Technology Zuerich Otto-Stern-Weg 7, 8093, Zuerich, Switzerland
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden Fetscherstr. 105, 01307, Dresden, Germany ; Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD) Fetscherstr. 74, 01307, Dresden, Germany
| | - Barry Rouse
- Department of Pathobiology, College of Veterinary Medicine, University of Tennessee Knoxville, TN, 37996, USA
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| |
Collapse
|
40
|
Mayer CT, Ghorbani P, Kühl AA, Stüve P, Hegemann M, Berod L, Gershwin ME, Sparwasser T. Few Foxp3⁺ regulatory T cells are sufficient to protect adult mice from lethal autoimmunity. Eur J Immunol 2014; 44:2990-3002. [PMID: 25042334 DOI: 10.1002/eji.201344315] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 06/03/2014] [Accepted: 07/11/2014] [Indexed: 12/12/2022]
Abstract
Foxp3 specifies the Treg cell lineage and is indispensable for immune tolerance. Accordingly, rare Foxp3 mutations cause lethal autoimmunity. The mechanisms precipitating more prevalent human autoimmune diseases are poorly understood, but involve a combination of genetic and environmental factors. Many autoimmune diseases associate with a partial Treg-cell dysfunction, yet mouse models reflecting such complex pathophysiological processes are rare. Around 95% of Foxp3(+) Treg cells can be specifically depleted in bacterial artifical chromosome (BAC)-transgenic Depletion of REGulatory T cells (DEREG) mice through diphtheria toxin (DT) treatment. However, Treg-cell depletion fails to cause autoimmunity in adult DEREG mice for unclear reasons. By crossing Foxp3(GFP) knock-in mice to DEREG mice, we introduced additional genetic susceptibility that does not affect untreated mice. Strikingly, DT treatment of DEREG × Foxp3(GFP) mice rapidly causes autoimmunity characterized by blepharitis, tissue damage, and autoantibody production. This inflammatory disease is associated with augmented T-cell activation, increased Th2 cytokine production and myeloproliferation, and is caused by defective Treg-cell homeostasis, preventing few DT-insensitive Treg cells from repopulating the niche after Treg-cell depletion. Our study provides important insights into self-tolerance. We further highlight DEREG × Foxp3(GFP) mice as a model to investigate the role of environmental factors in precipitating autoimmunity. This may help to better understand and treat human autoimmunity.
Collapse
Affiliation(s)
- Christian T Mayer
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ehrlich A, Castilho TM, Goldsmith-Pestana K, Chae WJ, Bothwell ALM, Sparwasser T, McMahon-Pratt D. The immunotherapeutic role of regulatory T cells in Leishmania (Viannia) panamensis infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:2961-70. [PMID: 25098291 DOI: 10.4049/jimmunol.1400728] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Leishmania (Viannia) parasites are etiological agents of cutaneous leishmaniasis in the New World. Infection is characterized by a mixed Th1/Th2 inflammatory response, which contributes to disease pathology. However, the role of regulatory T cells (Tregs) in Leishmania (Viannia) disease pathogenesis is unclear. Using the mouse model of chronic L. (V.) panamensis infection, we examined the hypothesis that Treg functionality contributes to control of pathogenesis. Upon infection, Tregs (CD4(+)Foxp3(+)) presented with a dysregulated phenotype, in that they produced IFN-γ, expressed Tbet, and had a reduced ability to suppress T cell proliferation in vitro. Targeted ablation of Tregs resulted in enlarged lesions, increased parasite load, and enhanced production of IL-17 and IFN-γ, with no change in IL-10 and IL-13 levels. This indicated that an increased inflammatory response was commensurate with disease exacerbation and that the remaining impaired Tregs were important in regulation of disease pathology. Conversely, adoptive transfer of Tregs from naive mice halted disease progression, lowered parasite burden, and reduced cytokine production (IL-10, IL-13, IL-17, IFN-γ). Because Tregs appeared to be important for controlling infection, we hypothesized that their expansion could be used as an immunotherapeutic treatment approach. As a proof of principle, chronically infected mice were treated with rIL-2/anti-IL-2 Ab complex to expand Tregs. Treatment transitorily increased the numbers and percentage of Tregs (draining lymph node, spleen), which resulted in reduced cytokine responses, ameliorated lesions, and reduced parasite load (10(5)-fold). Thus, immunotherapy targeting Tregs could provide an alternate treatment strategy for leishmaniasis caused by Leishmania (Viannia) parasites.
Collapse
Affiliation(s)
- Allison Ehrlich
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520
| | - Tiago Moreno Castilho
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520
| | - Karen Goldsmith-Pestana
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520
| | - Wook-Jin Chae
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520; and
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520; and
| | - Tim Sparwasser
- Institute of Infection Immunology, Centre for Experimental and Clinical Infection Research, TWINCORE, 30625 Hanover, Germany
| | - Diane McMahon-Pratt
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520;
| |
Collapse
|