1
|
Mas G, Man N, Nakata Y, Martinez-Caja C, Karl D, Beckedorff F, Tamiro F, Chen C, Duffort S, Itonaga H, Mookhtiar AK, Kunkalla K, Valencia AM, Collings CK, Kadoch C, Vega F, Kogan SC, Shiekhattar R, Morey L, Bilbao D, Nimer SD. The SWI/SNF chromatin-remodeling subunit DPF2 facilitates NRF2-dependent antiinflammatory and antioxidant gene expression. J Clin Invest 2023; 133:e158419. [PMID: 37200093 PMCID: PMC10313367 DOI: 10.1172/jci158419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/16/2023] [Indexed: 05/20/2023] Open
Abstract
During emergency hematopoiesis, hematopoietic stem cells (HSCs) rapidly proliferate to produce myeloid and lymphoid effector cells, a response that is critical against infection or tissue injury. If unresolved, this process leads to sustained inflammation, which can cause life-threatening diseases and cancer. Here, we identify a role of double PHD fingers 2 (DPF2) in modulating inflammation. DPF2 is a defining subunit of the hematopoiesis-specific BAF (SWI/SNF) chromatin-remodeling complex, and it is mutated in multiple cancers and neurological disorders. We uncovered that hematopoiesis-specific Dpf2-KO mice developed leukopenia, severe anemia, and lethal systemic inflammation characterized by histiocytic and fibrotic tissue infiltration resembling a clinical hyperinflammatory state. Dpf2 loss impaired the polarization of macrophages responsible for tissue repair, induced the unrestrained activation of Th cells, and generated an emergency-like state of HSC hyperproliferation and myeloid cell-biased differentiation. Mechanistically, Dpf2 deficiency resulted in the loss of the BAF catalytic subunit BRG1 from nuclear factor erythroid 2-like 2-controlled (NRF2-controlled) enhancers, impairing the antioxidant and antiinflammatory transcriptional response needed to modulate inflammation. Finally, pharmacological reactivation of NRF2 suppressed the inflammation-mediated phenotypes and lethality of Dpf2Δ/Δ mice. Our work establishes an essential role of the DPF2-BAF complex in licensing NRF2-dependent gene expression in HSCs and immune effector cells to prevent chronic inflammation.
Collapse
Affiliation(s)
- Gloria Mas
- Sylvester Comprehensive Cancer Center and
| | - Na Man
- Sylvester Comprehensive Cancer Center and
| | - Yuichiro Nakata
- Sylvester Comprehensive Cancer Center and
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | - Felipe Beckedorff
- Sylvester Comprehensive Cancer Center and
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Chuan Chen
- Sylvester Comprehensive Cancer Center and
| | | | | | | | | | - Alfredo M. Valencia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Chemical Biology Program, Harvard University, Cambridge, Massachusetts, USA
| | - Clayton K. Collings
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Cigall Kadoch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Francisco Vega
- Sylvester Comprehensive Cancer Center and
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Scott C. Kogan
- Helen Diller Family Comprehensive Cancer Center and
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Ramin Shiekhattar
- Sylvester Comprehensive Cancer Center and
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Lluis Morey
- Sylvester Comprehensive Cancer Center and
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center and
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Stephen D. Nimer
- Sylvester Comprehensive Cancer Center and
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
2
|
Liu Y, Zhang H, Zhang W, Xiang L, Yin Z, Xu H, Lu P, Ma Y, Xiong L, Zhang X, Liang X, Luo J, Liang X. circ_0004140 promotes LUAD tumor progression and immune resistance through circ_0004140/miR-1184/CCL22 axis. Cell Death Dis 2022; 8:181. [PMID: 35396377 PMCID: PMC8993797 DOI: 10.1038/s41420-022-00983-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022]
Abstract
Lung adenocarcinoma (LUAD) is a highly prevalent cancer with high mortality. Immune resistance and tumor metastasis are the pivotal factors for the promotion of LUAD. CircRNAs have been revealed a crucial pre-clinical diagnostic and therapeutic potentials in LUAD. Herein, we identify a novel circRNA (circ_0004140), derived from the oncogene YAP1, which is up-regulated in LUAD. The high expression of circ_0004140 is correlated with poor prognosis and CTL cells dysfunction in LUAD patients. Knockdown of circ_0004140 regulated LUAD cells proliferation, migration, and apoptosis. Mechanistically, circ_0004140 served as a sponge of miR-1184 targeting C-C motif chemokine ligand 22(CCL22). Overexpression of CCL22 reversed the inhibitory effect induced by si-circ_0004140 on cells proliferation and migration. Moreover, we also revealed that elevated circ_ooo4140 was related to cytotoxic lymphocyte exhaustion, and a combination therapy of C-021 (CCL22/CCR4 axis inhibitor) and anti-PD-1 attenuated LUAD promotion and immune resistance. In conclusion, circ_0004140 may drive resistance to anti-PD-1 immunotherapy, providing a novel potential therapeutic target for LUAD treatment.
Collapse
Affiliation(s)
- Yanyan Liu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, 430030, Wuhan, Hubei, P. R. China
| | - Haodong Zhang
- School of life science and technology, Huazhong Agricultural University, 430070, Wuhan, Hubei, P. R. China
| | - Wangli Zhang
- School of life science and technology, Huazhong Agricultural University, 430070, Wuhan, Hubei, P. R. China
| | - Lanxin Xiang
- School of life science and technology, Huazhong Agricultural University, 430070, Wuhan, Hubei, P. R. China
| | - Zhucheng Yin
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Hongli Xu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Ping Lu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Yifei Ma
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Lingyi Xiong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Xiangchen Zhang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Xin Liang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xinjun Liang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 116 Zhuodaoquan South Load, Hongshan District, 430079, Wuhan, Hubei, P. R. China.
| |
Collapse
|
3
|
Korenfeld D, Roussak K, Dinkel S, Vogel TP, Pollack H, Levy J, Leiding JW, Milner J, Cooper M, Klechevsky E. STAT3 Gain-of-Function Mutations Underlie Deficiency in Human Nonclassical CD16 + Monocytes and CD141 + Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:2423-2432. [PMID: 34654687 DOI: 10.4049/jimmunol.2000841] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
Genetic analysis of human inborn errors of immunity has defined the contribution of specific cell populations and molecular pathways in the host defense against infection. The STAT family of transcription factors orchestrate hematopoietic cell differentiation. Patients with de novo activating mutations of STAT3 present with multiorgan autoimmunity, lymphoproliferation, and recurrent infections. We conducted a detailed characterization of the blood monocyte and dendritic cell (DC) subsets in patients with gain-of-function (GOF) mutations across the gene. We found a selective deficiency in circulating nonclassical CD16+ and intermediate CD16+CD14+ monocytes and a significant increase in the percentage of classical CD14+ monocytes. This suggests a role for STAT3 in the transition of classical CD14+ monocytes into the CD16+ nonclassical subset. Developmentally, ex vivo-isolated STAT3GOF CD14+ monocytes fail to differentiate into CD1a+ monocyte-derived DCs. Moreover, patients with STAT3GOF mutations display reduced circulating CD34+ hematopoietic progenitors and frequency of myeloid DCs. Specifically, we observed a reduction in the CD141+ DC population, with no difference in the frequencies of CD1c+ and plasmacytoid DCs. CD34+ hematopoietic progenitor cells from patients were found to differentiate into CD1c+ DCs, but failed to differentiate into CD141+ DCs indicating an intrinsic role for STAT3 in this process. STAT3GOF-differentiated DCs produced lower amounts of CCL22 than healthy DCs, which could further explain some of the patient pathological phenotypes. Thus, our findings provide evidence that, in humans, STAT3 serves to regulate development and differentiation of nonclassical CD16+ monocytes and a subset of myeloid DCs.
Collapse
Affiliation(s)
- Daniel Korenfeld
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Kate Roussak
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Sabrina Dinkel
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Tiphanie P Vogel
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St. Louis, MO
| | - Henry Pollack
- Department of Pediatrics, New York University School of Medicine, New York, NY
| | - Joseph Levy
- Department of Pediatrics, New York University School of Medicine, New York, NY
| | - Jennifer W Leiding
- Department of Pediatrics, Division of Allergy and Immunology, University of South Florida, Tampa, FL; and
| | - Joshua Milner
- Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Megan Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St. Louis, MO
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO;
| |
Collapse
|
4
|
Knopick P, Terman D, Riha N, Alvine T, Larson R, Badiou C, Lina G, Ballantyne J, Bradley D. Endogenous HLA-DQ8αβ programs superantigens (SEG/SEI) to silence toxicity and unleash a tumoricidal network with long-term melanoma survival. J Immunother Cancer 2021; 8:jitc-2020-001493. [PMID: 33109631 PMCID: PMC7592263 DOI: 10.1136/jitc-2020-001493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
Background As the most powerful T cell agonists known, superantigens (SAgs) have enormous potential for cancer immunotherapy. Their development has languished due to high incidence (60%–80%) of seroreactive neutralizing antibodies in humans and tumor necrosis factor-α (TNFα)-mediated cardiopulmonary toxicity. Such toxicity has narrowed their therapeutic index while neutralizing antibodies have nullified their therapeutic effects. Methods Female HLA-DQ8 (DQA*0301/DQB*0302) tg mice expressing the human major histocompatibility complex II (MHCII) HLA-DQ8 allele on a high proportion of PBL, spleen and lymph node cells were used. In the established tumor model, staphylococcal enterotoxin G and staphylococcal enterotoxin I (SEG/ SEI) (50 µg each) were injected on days 6 and 9 following tumor inoculation. Lymphoid, myeloid cells and tumor cell digests from tumor tissue were assayed using flow cytometry or quantitated using a cytometric bead array. Tumor density, necrotic and viable areas were quantitated using the ImageJ software. Results In a discovery-driven effort to address these problems we introduce a heretofore unrecognized binary complex comprizing SEG/SEI SAgs linked to the endogenous human MHCII HLA-DQ8 allele in humanized mice. By contrast to staphylococcal enterotoxin A (SEA) and staphylococcal enterotoxin B (SEB) deployed previously in clinical trials, SEG and SEI does not exhibit neutralizing antibodies in humans or TNFα-mediated toxicity in humanized HLA-DQ8 mice. In the latter model wherein SAg behavior is known to be ‘human-like’, SEG/SEI induced a powerful tumoricidal response and long-term survival against established melanoma in 82% of mice. Other SAgs deployed in the same model displayed toxic shock. Initially, HLA-DQ8 mediated melanoma antigen priming, after which SEG/SEI unleashed a broad CD4+ and CD8+ antitumor network marked by expansion of melanoma reactive T cells and interferon-γ (IFNy) in the tumor microenvironment (TME). SEG/SEI further initiated chemotactic recruitment of tumor reactive T cells to the TME converting the tumor from ‘cold’ to a ‘hot’. Long-term survivors displayed remarkable resistance to parental tumor rechallenge along with the appearance of tumor specific memory and tumor reactive T memory cells. Conclusions Collectively, these findings show for the first time that the SEG/SEI-(HLA-DQ8) empowers priming, expansion and recruitment of a population of tumor reactive T cells culminating in tumor specific memory and long-term survival devoid of toxicity. These properties distinguish SEG/SEI from other SAgs used previously in human tumor immunotherapy. Consolidation of these principles within the SEG/SEI-(HLA-DQ8) complex constitutes a conceptually new therapeutic weapon with compelling translational potential.
Collapse
Affiliation(s)
- Peter Knopick
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - David Terman
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Nathan Riha
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Travis Alvine
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Riley Larson
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Cedric Badiou
- University of Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Gerard Lina
- University of Lyon 1 University Institute of Tecnology Lyon 1, Villeurbanne, Auvergne-Rhône-Alpes, France
| | | | - David Bradley
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| |
Collapse
|
5
|
Krentz NAJ, Shea LD, Huising MO, Shaw JAM. Restoring normal islet mass and function in type 1 diabetes through regenerative medicine and tissue engineering. Lancet Diabetes Endocrinol 2021; 9:708-724. [PMID: 34480875 PMCID: PMC10881068 DOI: 10.1016/s2213-8587(21)00170-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/17/2021] [Accepted: 06/08/2021] [Indexed: 02/09/2023]
Abstract
Type 1 diabetes is characterised by autoimmune-mediated destruction of pancreatic β-cell mass. With the advent of insulin therapy a century ago, type 1 diabetes changed from a progressive, fatal disease to one that requires lifelong complex self-management. Replacing the lost β-cell mass through transplantation has proven successful, but limited donor supply and need for lifelong immunosuppression restricts widespread use. In this Review, we highlight incremental advances over the past 20 years and remaining challenges in regenerative medicine approaches to restoring β-cell mass and function in type 1 diabetes. We begin by summarising the role of endocrine islets in glucose homoeostasis and how this is altered in disease. We then discuss the potential regenerative capacity of the remaining islet cells and the utility of stem cell-derived β-like cells to restore β-cell function. We conclude with tissue engineering approaches that might improve the engraftment, function, and survival of β-cell replacement therapies.
Collapse
Affiliation(s)
- Nicole A J Krentz
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Lonnie D Shea
- Departments of Biomedical Engineering, Chemical Engineering, and Surgery, College of Engineering and School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA; Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, USA
| | - James A M Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
6
|
Barlier C, Barriales D, Samosyuk A, Jung S, Ravichandran S, Medvedeva YA, Anguita J, Del Sol A. A Catalogus Immune Muris of the mouse immune responses to diverse pathogens. Cell Death Dis 2021; 12:798. [PMID: 34404761 PMCID: PMC8370971 DOI: 10.1038/s41419-021-04075-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/09/2022]
Abstract
Immunomodulation strategies are crucial for several biomedical applications. However, the immune system is highly heterogeneous and its functional responses to infections remains elusive. Indeed, the characterization of immune response particularities to different pathogens is needed to identify immunomodulatory candidates. To address this issue, we compiled a comprehensive map of functional immune cell states of mouse in response to 12 pathogens. To create this atlas, we developed a single-cell-based computational method that partitions heterogeneous cell types into functionally distinct states and simultaneously identifies modules of functionally relevant genes characterizing them. We identified 295 functional states using 114 datasets of six immune cell types, creating a Catalogus Immune Muris. As a result, we found common as well as pathogen-specific functional states and experimentally characterized the function of an unknown macrophage cell state that modulates the response to Salmonella Typhimurium infection. Thus, we expect our Catalogus Immune Muris to be an important resource for studies aiming at discovering new immunomodulatory candidates.
Collapse
Affiliation(s)
- Céline Barlier
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Diego Barriales
- Inflammation and Macrophage Plasticity laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, 48160, Spain
| | - Alexey Samosyuk
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russian Federation
| | - Sascha Jung
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, 48160, Spain
| | - Srikanth Ravichandran
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Yulia A Medvedeva
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russian Federation
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Science, Moscow, Russian Federation
- Department of Computational Biology, Vavilov Institute of General Genetics, Russian Academy of Science, Moscow, Russian Federation
| | - Juan Anguita
- Inflammation and Macrophage Plasticity laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, 48160, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, 48012, Spain
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg.
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, 48160, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, 48012, Spain.
| |
Collapse
|
7
|
Li Y, Sun F, Yue TT, Wang FX, Yang CL, Luo JH, Rong SJ, Xiong F, Zhang S, Wang CY. Revisiting the Antigen-Presenting Function of β Cells in T1D Pathogenesis. Front Immunol 2021; 12:690783. [PMID: 34335595 PMCID: PMC8318689 DOI: 10.3389/fimmu.2021.690783] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/30/2021] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by the unresolved autoimmune inflammation and islet β cell destruction. The islet resident antigen-presenting cells (APCs) including dendritic cells and macrophages uptake and process the β cell-derived antigens to prime the autoreactive diabetogenic T cells. Upon activation, those autoreactive T cells produce copious amount of IFN-γ, TNF-α and IL-1β to induce β cell stress and death. Autoimmune attack and β cell damage intertwine together to push forward this self-destructive program, leading to T1D onset. However, β cells are far beyond a passive participant during the course of T1D development. Herein in this review, we summarized how β cells are actively involved in the initiation of autoimmune responses in T1D setting. Specifically, β cells produce modified neoantigens under stressed condition, which is coupled with upregulated expression of MHC I/II and co-stimulatory molecules as well as other immune modules, that are essential properties normally exhibited by the professional APCs. At the cellular level, this subset of APC-like β cells dynamically interacts with plasmacytoid dendritic cells (pDCs) and manifests potency to activate autoreactive CD4 and CD8 T cells, by which β cells initiate early autoimmune responses predisposing to T1D development. Overall, the antigen-presenting function of β cells helps to explain the tissue specificity of T1D and highlights the active roles of structural cells played in the pathogenesis of various immune related disorders.
Collapse
Affiliation(s)
- Yang Li
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian-Tian Yue
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fa-Xi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Liang Yang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Hui Luo
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Jie Rong
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Jamal Jameel K, Gallert WJ, Yanik SD, Panek S, Kronsbein J, Jungck D, Koch A, Knobloch J. Biomarkers for Comorbidities Modulate the Activity of T-Cells in COPD. Int J Mol Sci 2021; 22:ijms22137187. [PMID: 34281240 PMCID: PMC8269158 DOI: 10.3390/ijms22137187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/17/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
In smoking-induced chronic obstructive pulmonary disease (COPD), various comorbidities are linked to systemic inflammation and infection-induced exacerbations. The underlying mechanisms are unclear but might provide therapeutic targets. T-cell activity is central in systemic inflammation and for infection-defense mechanisms and might be influenced by comorbidities. Hypothesis: Circulating biomarkers of comorbidities modulate the activity of T-cells of the T-helper type 1 (Th1) and/or T-cytotoxic type 1 (Tc1). T-cells in peripheral blood mononuclear cells (PBMCs) from non-smokers (NS), current smokers without COPD (S), and COPD subjects (total n = 34) were ex vivo activated towards Th1/Tc1 and were then stimulated with biomarkers for metabolic and/or cardiovascular comorbidities (Brain Natriuretic Peptide, BNP; chemokine (C-C motif) ligand 18, CCL18; C-X3-C motif chemokine ligand 1, CX3CL1; interleukin-18, IL-18) or for asthma- and/or cancer-related comorbidities (CCL22; epidermal growth factor, EGF; IL-17; periostin) each at 10 or 50 ng/mL. The Th1/Tc1 activation markers interferon-γ (IFNγ), tumor necrosis factor-α (TNFα), and granulocyte-macrophage colony-stimulating factor (GM-CSF) were analyzed in culture supernatants by Enzyme-Linked Immunosorbent Assay (ELISA). Ex-vivo activation induced IFNγ and TNFα without differences between the groups but GM-CSF more in S vs. NS. At 10 ng/mL, the different biomarkers increased or reduced the T-cell activation markers without a clear trend for one direction in the different categories of comorbidities or for the different T-cell activation markers. At 50 ng/mL, there was a clear shift towards suppressive effects, particularly for the asthma— and cancer-related biomarkers and in cells of S and COPD. Comorbidities might suppress T-cell immunity in COPD. This could explain the association of comorbidities with frequent exacerbations.
Collapse
Affiliation(s)
- Kaschin Jamal Jameel
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Willem-Jakob Gallert
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Sarah D. Yanik
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Susanne Panek
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
| | - David Jungck
- Department of Internal Medicine II, Pneumology, Allergology and Respiratory Medicine, Bethel Teaching Hospital, 12207 Berlin, Germany;
| | - Andrea Koch
- Pyhrn-Eisenwurzen-Klinikum Steyr, Klinik für Pneumologie, Lehrkrankenhaus der Uniklinik Linz, Sierninger Str. 170, 4400 Steyr, Austria;
- Ludwig-Maximilians-University of Munich (LMU) and DZL (German Center of Lung Science), 81377 Munich, Germany
| | - Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, 44789 Bochum, Germany; (K.J.J.); (W.-J.G.); (S.D.Y.); (S.P.); (J.K.)
- Correspondence: ; Tel.: +49-234-302-3404; Fax: +49-234-302-6420
| |
Collapse
|
9
|
de Groot P, Nikolic T, Pellegrini S, Sordi V, Imangaliyev S, Rampanelli E, Hanssen N, Attaye I, Bakker G, Duinkerken G, Joosten A, Prodan A, Levin E, Levels H, Potter van Loon B, van Bon A, Brouwer C, van Dam S, Simsek S, van Raalte D, Stam F, Gerdes V, Hoogma R, Diekman M, Gerding M, Rustemeijer C, de Bakker B, Hoekstra J, Zwinderman A, Bergman J, Holleman F, Piemonti L, De Vos W, Roep B, Nieuwdorp M. Faecal microbiota transplantation halts progression of human new-onset type 1 diabetes in a randomised controlled trial. Gut 2021; 70:92-105. [PMID: 33106354 PMCID: PMC7788262 DOI: 10.1136/gutjnl-2020-322630] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Type 1 diabetes (T1D) is characterised by islet autoimmunity and beta cell destruction. A gut microbiota-immunological interplay is involved in the pathophysiology of T1D. We studied microbiota-mediated effects on disease progression in patients with type 1 diabetes using faecal microbiota transplantation (FMT). DESIGN Patients with recent-onset (<6 weeks) T1D (18-30 years of age) were randomised into two groups to receive three autologous or allogenic (healthy donor) FMTs over a period of 4 months. Our primary endpoint was preservation of stimulated C peptide release assessed by mixed-meal tests during 12 months. Secondary outcome parameters were changes in glycaemic control, fasting plasma metabolites, T cell autoimmunity, small intestinal gene expression profile and intestinal microbiota composition. RESULTS Stimulated C peptide levels were significantly preserved in the autologous FMT group (n=10 subjects) compared with healthy donor FMT group (n=10 subjects) at 12 months. Small intestinal Prevotella was inversely related to residual beta cell function (r=-0.55, p=0.02), whereas plasma metabolites 1-arachidonoyl-GPC and 1-myristoyl-2-arachidonoyl-GPC levels linearly correlated with residual beta cell preservation (rho=0.56, p=0.01 and rho=0.46, p=0.042, respectively). Finally, baseline CD4 +CXCR3+T cell counts, levels of small intestinal Desulfovibrio piger and CCL22 and CCL5 gene expression in duodenal biopsies predicted preserved beta cell function following FMT irrespective of donor characteristics. CONCLUSION FMT halts decline in endogenous insulin production in recently diagnosed patients with T1D in 12 months after disease onset. Several microbiota-derived plasma metabolites and bacterial strains were linked to preserved residual beta cell function. This study provides insight into the role of the intestinal gut microbiome in T1D. TRIAL REGISTRATION NUMBER NTR3697.
Collapse
Affiliation(s)
- Pieter de Groot
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Tanja Nikolic
- Department of Internal Medicine, LUMC, Leiden, Zuid-Holland, The Netherlands
| | - Silvia Pellegrini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Sordi
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Sultan Imangaliyev
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Elena Rampanelli
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Nordin Hanssen
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Ilias Attaye
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Guido Bakker
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Gaby Duinkerken
- Department of Internal Medicine, LUMC, Leiden, Zuid-Holland, The Netherlands
| | - Antoinette Joosten
- Department of Internal Medicine, LUMC, Leiden, Zuid-Holland, The Netherlands
| | - Andrei Prodan
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Evgeni Levin
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Han Levels
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | | | - Arianne van Bon
- Internal Medicine, Rijnstate, Arnhem, Gelderland, The Netherlands
| | - Catherina Brouwer
- Internal Medicine, OLVG, Location Oost, Amsterdam, Noord-Holland, The Netherlands
| | - Sytze van Dam
- Internal Medicine, OLVG, Location Oost, Amsterdam, Noord-Holland, The Netherlands
| | - Suat Simsek
- Internal Medicine, North West Hospital Group, Alkmaar, Noord-Holland, The Netherlands
| | - Daniel van Raalte
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Frank Stam
- Internal Medicine, North West Hospital Group, Alkmaar, Noord-Holland, The Netherlands
| | - Victor Gerdes
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Roel Hoogma
- Internal Medicine, Groene Hart Hospital, Gouda, Zuid-Holland, The Netherlands
| | - Martin Diekman
- Internal Medicine, Deventer Hospital, Deventer, Overijssel, The Netherlands
| | - Martin Gerding
- Internal Medicine, Deventer Hospital, Deventer, Overijssel, The Netherlands
| | - Cees Rustemeijer
- Internal Medicine, Hospital Amstelland, Amstelveen, North Holland, The Netherlands
| | - Bernadette de Bakker
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Joost Hoekstra
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Aeilko Zwinderman
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Jacques Bergman
- Department of Gastroenterology, Academic Medical Center, Amsterdam, The Netherlands
| | - Frits Holleman
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Bart Roep
- Department of Internal Medicine, LUMC, Leiden, Zuid-Holland, The Netherlands
- Department of Diabetes Immunology, Diabetes & Metabolism Research Institute at the Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| |
Collapse
|
10
|
Glucose-lowering activity of dark tea protein extract by modulating spleen–brain axis of diabetic mice. Br J Nutr 2020; 126:961-969. [DOI: 10.1017/s0007114520005012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AbstractThe present study aims to explore the glucose-lowering effects of the previously characterised dark tea (Camellia sinensis L.) protein extract (DTPE) from Heimaojian on the spleen–brain axis of diabetic mice. DTPE was orally administrated (50–100 mg/kg) to alloxan-induced mice for 21 d; a biochemical assay and transcriptome profiling (RNA sequencing (RNA-Seq)) were performed. The results showed that DTPE can improve glucose tolerance. Compared with the model group, at day 21, the fasting blood glucose values were significantly (P < 0·05) decreased by 44·9 % (13·8 v. 7·6 mmol/l) and 51·4 % (13·8 v. 6·7 mmol/l) for high dose of DTPE (100 mg/kg) and drug metformin (125 mg/kg) groups, respectively. Subsequently, transcriptome profiling (RNA-Seq) was performed on the spleen and brain of diabetic mice. Totally, fifty-two spleen-derived and forty-seven brain-derived differentially expressed genes related to the synthesis, transport and metabolism of glucose were identified. The regulatory network analysis indicated that DTPE may exert glucose-lowering effects through a thirty-seven-gene sub-network related to metabolism, Parkinson’s disease, oxidative phosphorylation and immunity. In summary, for the first time, the present data revealed that dark tea-derived DTPE could exert a potential anti-hyperglycaemic effect by modulating the spleen–brain axis.
Collapse
|
11
|
Sakai R, Ito M, Yoshimoto K, Chikuma S, Kurasawa T, Kondo T, Suzuki K, Takeuchi T, Amano K, Yoshimura A. Tocilizumab monotherapy uncovered the role of the CCL22/17-CCR4 + Treg axis during remission of crescentic glomerulonephritis. Clin Transl Immunology 2020; 9:e1203. [PMID: 33163184 PMCID: PMC7596393 DOI: 10.1002/cti2.1203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/08/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Objectives Tocilizumab (TCZ) is a humanised anti‐interleukin (IL)‐6 receptor (IL‐6R) monoclonal antibody that is a promising agent to treat various autoimmune diseases. However, the mechanism of TCZ efficacy is unclear. This study aims to elucidate the relationship between Tregs and IL‐6R blockade in autoimmunity‐mediated renal disease based on a TCZ‐treated cohort of patients with anti‐neutrophil cytoplasmic antibody (ANCA)‐associated vasculitis (AAV) and in an experimental model of crescentic glomerulonephritis (cGN). Methods We examined multiple serum levels of cytokines and chemokines and peripheral blood mononuclear cells in patients with AAV who received TCZ monotherapy and achieved drug‐free remission. Moreover, we investigated the mechanistic role of IL‐6R blockade in accelerated cGN model to analyse the local sites of inflammation. Results Serum chemokines CCL22 and CCL17, in addition to the CCR4+Foxp3+ Treg population, increased in patients who demonstrated drug‐free remission after the cessation of TCZ. In the cGN model, IL‐6R blockade ameliorated the disease, elevated CCL22/17 in CD206+CD11b+CD11c+ kidney M2‐like type macrophages, and increased the migration of Tregs into the kidney and regional lymph nodes. The local administration of CCL22 in the kidney facilitated Treg accumulation and reduced glomerular crescent formation. Conclusions This study revealed a new mechanism whereby effector Tregs migrate into the inflammatory kidney via the CCL22/17–CCR4 axis that is facilitated by M2‐like type macrophages that are induced by IL‐6R blockade.
Collapse
Affiliation(s)
- Ryota Sakai
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan.,Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Minako Ito
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan
| | - Keiko Yoshimoto
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan
| | - Takahiko Kurasawa
- Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Tsuneo Kondo
- Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Katsuya Suzuki
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Koichi Amano
- Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan
| |
Collapse
|
12
|
Elevated serum chemokine CCL22 levels in first-episode psychosis: associations with symptoms, peripheral immune state and in vivo brain glial cell function. Transl Psychiatry 2020; 10:94. [PMID: 32179746 PMCID: PMC7075957 DOI: 10.1038/s41398-020-0776-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023] Open
Abstract
Several lines of research support immune system dysregulation in psychotic disorders. However, it remains unclear whether the immunological marker alterations are stable and how they associate with brain glial cell function. This longitudinal study aimed at investigating whether peripheral immune functions are altered in the early phases of psychotic disorders, whether the changes are associated with core symptoms, remission, brain glial cell function, and whether they persist in a one-year follow-up. Two independent cohorts comprising in total of 129 first-episode psychosis (FEP) patients and 130 controls were assessed at baseline and at the one-year follow-up. Serum cyto-/chemokines were measured using a 38-plex Luminex assay. The FEP patients showed a marked increase in chemokine CCL22 levels both at baseline (p < 0.0001; Cohen's d = 0.70) and at the 12-month follow-up (p = 0.0007) compared to controls. The group difference remained significant (p = 0.0019) after accounting for relevant covariates including BMI, smoking, and antipsychotic medication. Elevated serum CCL22 levels were significantly associated with hallucinations (ρ = 0.20) and disorganization (ρ = 0.23), and with worse verbal performance (ρ = -0.23). Brain glial cell activity was indexed with positron emission tomography and the translocator protein radiotracer [11C]PBR28 in subgroups of 15 healthy controls and 14 FEP patients with serum CCL22/CCL17 measurements. The distribution volume (VT) of [11C]PBR28 was lower in patients compared to controls (p = 0.026; Cohen's d = 0.94) without regionally specific effects, and was inversely associated with serum CCL22 and CCL17 levels (p = 0.036). Our results do not support the over-active microglia hypothesis of psychosis, but indicate altered CCR4 immune signaling in early psychosis with behavioral correlates possibly mediated through cross-talk between chemokine networks and dysfunctional or a decreased number of glial cells.
Collapse
|
13
|
Fisher JD, Zhang W, Balmert SC, Aral AM, Acharya AP, Kulahci Y, Li J, Turnquist HR, Thomson AW, Solari MG, Gorantla VS, Little SR. In situ recruitment of regulatory T cells promotes donor-specific tolerance in vascularized composite allotransplantation. SCIENCE ADVANCES 2020; 6:eaax8429. [PMID: 32201714 PMCID: PMC7069700 DOI: 10.1126/sciadv.aax8429] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 12/17/2019] [Indexed: 05/04/2023]
Abstract
Vascularized composite allotransplantation (VCA) encompasses face and limb transplantation, but as with organ transplantation, it requires lifelong regimens of immunosuppressive drugs to prevent rejection. To achieve donor-specific immune tolerance and reduce the need for systemic immunosuppression, we developed a synthetic drug delivery system that mimics a strategy our bodies naturally use to recruit regulatory T cells (Treg) to suppress inflammation. Specifically, a microparticle-based system engineered to release the Treg-recruiting chemokine CCL22 was used in a rodent hindlimb VCA model. These "Recruitment-MP" prolonged hindlimb allograft survival indefinitely (>200 days) and promoted donor-specific tolerance. Recruitment-MP treatment enriched Treg populations in allograft skin and draining lymph nodes and enhanced Treg function without affecting the proliferative capacity of conventional T cells. With implications for clinical translation, synthetic human CCL22 induced preferential migration of human Treg in vitro. Collectively, these results suggest that Recruitment-MP promote donor-specific immune tolerance via local enrichment of suppressive Treg.
Collapse
Affiliation(s)
- James D. Fisher
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wensheng Zhang
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen C. Balmert
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ali M. Aral
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abhinav P. Acharya
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yalcin Kulahci
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jingjing Li
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heth R. Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Angus W. Thomson
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mario G. Solari
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vijay S. Gorantla
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Corresponding author. (S.R.L.); (V.S.G.)
| | - Steven R. Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Corresponding author. (S.R.L.); (V.S.G.)
| |
Collapse
|
14
|
He J, Chen G, Liu M, Xu Z, Chen H, Yang L, Lv Y. Scaffold strategies for modulating immune microenvironment during bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 108:110411. [PMID: 31923946 DOI: 10.1016/j.msec.2019.110411] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022]
Abstract
Implanted bone scaffolds often fail to successfully integrate with the host tissue because they do not elicit a favorable immune reaction. Properties of bone scaffold not only provide mechanical and chemical signals to support cell adhesion, migration, proliferation and differentiation, but also play a pivotal role in determining the extent of immune response during bone regeneration. Appropriate design parameters of bone scaffold are of great significance in the process of developing a new generation of bone implants. Herein, this article addresses the recent advances in the field of bone scaffolds for immune response, particularly focusing on the physical and chemical properties of bone scaffold in manipulating the host response. Furthermore, incorporation of bioactive molecules and cells with immunoregulatory function in bone scaffolds are also presented. Finally, continuing challenges and future directions of scaffold-based strategies for modulating immune microenvironment are discussed.
Collapse
Affiliation(s)
- Jianhua He
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Guobao Chen
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Mengying Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Zhiling Xu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Hua Chen
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, PR China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China; Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|
15
|
Engineering human stellate cells for beta cell replacement therapy promotes in vivo recruitment of regulatory T cells. Mater Today Bio 2019; 2:100006. [PMID: 32159143 PMCID: PMC7061575 DOI: 10.1016/j.mtbio.2019.100006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/25/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by destruction of pancreatic β cells. One of the promising therapeutic approaches in T1D is the transplantation of islets; however, it has serious limitations. To address these limitations, immunotherapeutic strategies have focused on restoring immunologic tolerance, preventing transplanted cell destruction by patients’ own immune system. Macrophage-derived chemokines such as chemokine-ligand-22 (CCL22) can be utilized for regulatory T cell (Treg) recruitment and graft tolerance. Stellate cells (SCs) have various immunomodulatory functions: recruitment of Tregs and induction of T-cell apoptosis. Here, we designed a unique immune-privileged microenvironment around implantable islets through overexpression of CCL22 proteins by SCs. We prepared pseudoislets with insulin-secreting mouse insulinoma-6 (MIN6) cells and human SCs as a model to mimic naive islet morphology. Our results demonstrated that transduced SCs can secrete CCL22 and recruit Tregs toward the implantation site in vivo. This study is promising to provide a fundamental understanding of SC-islet interaction and ligand synthesis and transport from SCs at the graft site for ensuring local immune tolerance. Our results also establish a new paradigm for creating tolerable grafts for other chronic diseases such as diabetes, anemia, and central nervous system (CNS) diseases, and advance the science of graft tolerance.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW The application of regulatory T cell (Treg) therapy in organ transplantation is actively being pursued using unmodified, typically polyclonal cells. As the results of these ongoing clinical trials emerge, it is time to plan the next wave of clinical trials of Tregs. Here we will review a key strategy to improve Treg effectiveness and reduce side effects, namely increasing Treg specificity - both in terms of antigen recognition and localization to the allograft. RECENT FINDINGS Study of chemokine signatures accompanying acute rejection has revealed several chemokines that could be targeted to increase Treg homing. For example, Tregs possessing a Th1-like phenotype and expressing CXCR3 are better able to migrate towards local inflammation. Allografts themselves can be modified to increase Treg-attracting chemokines and Tregs themselves can produce chemokines, facilitating local proximity to their targets of suppression. Finally, tailoring Treg antigen specificity by T-cell or chimeric antigen receptor engineering is another approach to increase the specificity of suppression and optimize localization. SUMMARY Treg localization to the graft is important, but the important role of lymph node and germinal center homing cannot be overlooked. There is an opportunity to learn from advances made in cancer immunotherapy to optimize Treg therapy for transplantation.
Collapse
|
17
|
Obach M, Hosseini-Tabatabaei A, Montane J, Wind K, Soukhatcheva G, Dai D, Priatel JJ, Orban PC, Verchere CB. Prevention of autoimmune diabetes and islet allograft rejection by beta cell expression of XIAP: Insight into possible mechanisms of local immunomodulation. Mol Cell Endocrinol 2018; 477:48-56. [PMID: 29883690 DOI: 10.1016/j.mce.2018.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/22/2018] [Accepted: 05/29/2018] [Indexed: 12/01/2022]
Abstract
Overexpression of the X-linked inhibitor of apoptosis (XIAP) prevents islet allograft rejection. We constructed an adeno-associated virus expressing XIAP driven by the rat insulin promoter (dsAAV8-RIP-XIAP) for long-term beta-cell gene expression in vivo. Pancreatic delivery of dsAAV8-RIP-XIAP prevented autoimmune diabetes in 70% of non-obese diabetic (NOD) mice, associated with decreased insulitis. Islets from Balb/c mice transduced with dsAAV8-RIP-XIAP were protected following transplantation into streptozotocin (STZ)-diabetic Bl/6 recipients, associated with decreased graft infiltration. Interestingly, dsAAV8-RIP-XIAP transduction induced expression of lactate dehydrogenase (LDHA) and monocarboxylate transporter 1 (MCT1), two genes normally suppressed in beta cells and involved in production and release of lactate, a metabolite known to suppress local immune responses. Transduction of Balb/c islets with AAV8-RIP-LDHA-MCT1 tended to prolong allograft survival following transplant into STZ-diabetic Bl/6 recipients. These findings suggest that XIAP has therapeutic potential in autoimmune diabetes and raise the possibility that local lactate production may play a role in XIAP-mediated immunomodulation.
Collapse
Affiliation(s)
- Mercè Obach
- Departments of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Azadeh Hosseini-Tabatabaei
- Departments of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Surgery, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Joel Montane
- Departments of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Katarina Wind
- Departments of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Galina Soukhatcheva
- Surgery, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Derek Dai
- Surgery, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - John J Priatel
- Departments of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul C Orban
- Surgery, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - C Bruce Verchere
- Departments of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada; Surgery, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
18
|
Ushio A, Arakaki R, Otsuka K, Yamada A, Tsunematsu T, Kudo Y, Aota K, Azuma M, Ishimaru N. CCL22-Producing Resident Macrophages Enhance T Cell Response in Sjögren's Syndrome. Front Immunol 2018; 9:2594. [PMID: 30467506 PMCID: PMC6236111 DOI: 10.3389/fimmu.2018.02594] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/22/2018] [Indexed: 12/28/2022] Open
Abstract
Macrophages (MΦs) are critical regulators of immune response and serve as a link between innate and acquired immunity. The precise mechanism of involvement of tissue-resident MΦs in the pathogenesis of autoimmune diseases is not clear. Here, using a murine model for Sjögren's syndrome (SS), we investigated the role of tissue-resident MΦs in the onset and development of autoimmunity. Two unique populations of CD11bhigh and CD11blow resident MΦs were observed in the target tissue of the SS model. Comprehensive gene expression analysis of chemokines revealed effective production of CCL22 by the CD11bhigh MΦs. CCL22 upregulated the migratory activity of CD4+ T cells by increasing CCR4, a receptor of CCL22, on T cells in the SS model. In addition, CCL22 enhanced IFN-γ production of T cells of the SS model, thereby suggesting that CCL22 may impair the local immune tolerance in the target organ of the SS model. Moreover, administration of anti-CCL22 antibody suppressed autoimmune lesions in the SS model. Finally, histopathological analysis revealed numerous CCL22-producing MΦs in the minor salivary gland tissue specimens of the SS patients. CCL22-producing tissue-resident MΦs may control autoimmune lesions by enhancing T cell response in the SS model. These results suggest that specific chemokines and their receptors may serve as novel therapeutic or diagnostic targets for SS.
Collapse
Affiliation(s)
- Aya Ushio
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kunihiro Otsuka
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Akiko Yamada
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takaaki Tsunematsu
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keiko Aota
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masayuki Azuma
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
19
|
Li W, Amet T, Xing Y, Yang D, Liangpunsakul S, Puri P, Kamath P, Sanyal A, Shah V, Katz B, Radaeva S, Crabb D, Chalasani N, Yu Q. Alcohol abstinence ameliorates the dysregulated immune profiles in patients with alcoholic hepatitis: A prospective observational study. Hepatology 2017; 66:575-590. [PMID: 28466561 PMCID: PMC5548491 DOI: 10.1002/hep.29242] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/27/2017] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
Abstract
Alcoholic hepatitis (AH) develops in only a small proportion of heavy drinkers. To better understand the mechanisms underlying this disparity, we conducted a study to define the relationship between AH development and dysregulated immune responses that might be ameliorated by alcohol abstinence. Sixty-eight AH patients, 65 heavy drinking controls without liver disease (HDC), and 20 healthy controls were enrolled and followed up to 12 months. At baseline, HDC and healthy controls had no significant differences in their plasma levels of 38 inflammatory cytokines/chemokines measured using multiplex immunoassays. However, compared to HDC, AH patients had higher baseline levels of 11 cytokines/chemokines (tumor necrosis factor alpha, interleukin 6 [IL-6], IL-8, interferon gamma-induced protein 10, IL-4, IL-9, IL-10, fibroblast growth factor 2, IL-7, IL-15, and transforming growth factor alpha) but lower levels of the anti-inflammatory macrophage-derived chemokine. AH patients also had more activated yet dysfunctional immune cells as monocytes, T cells, and B cells expressed higher levels of cluster of differentiation 38 (CD38) and CD69 but low levels of human leukocyte antigen DR, CD80, and CD86 at baseline. In addition, CD4 T cells produced less interferon-gamma in response to T-cell stimulation. Up-regulated IL-6, IL-8, CD38, and CD69 and down-regulated macrophage-derived chemokine, human leukocyte antigen DR, CD86, and CD80 correlated positively and negatively, respectively, with disease severity. Longitudinal analysis indicated that levels of IL-6, IL-8, CD38, and CD69 were reduced, whereas levels of macrophage-derived chemokine, human leukocyte antigen DR, CD80, and CD86 were increased in abstinent AH patients. All of the cellular immune abnormalities were reversed by day 360 in abstinent AH patients; however, plasma levels of tumor necrosis factor alpha, IL-8, IL-10, fibroblast growth factor 2, and IL-7 remained higher. CONCLUSION AH patients were in a highly immune-dysregulated state, whereas HDC showed little evidence of immune activation; alcohol abstinence reversed most, but not all, of the immunological abnormalities. (Hepatology 2017;66:575-590).
Collapse
Affiliation(s)
- Wei Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Tohti Amet
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Yanyan Xing
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Dennis Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202-5175
- Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Puneet Puri
- Division of Gastroenterology and Hepatology, Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Patrick Kamath
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Arun Sanyal
- Division of Gastroenterology and Hepatology, Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Vijay Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Barry Katz
- Department of Biostatistics, Indiana University School of Medicine and Richard M. Fairbanks School of Public Health, Indianapolis, IN 46202
| | - Svetlana Radaeva
- National Institute of Alcoholism and Alcohol Abuse, National Institutes of Health, Rockville, MD
| | - David Crabb
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202-5175
- Internal Medicine, Eskenazi Health, Indianapolis, IN 46202
| | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202-5175
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
20
|
Goihman-Yahr M. The see-saw of immune dysregulation association of diverse organ autoimmune conditions in a woman. Evolution over twenty-seven years. Int J Dermatol 2017; 56:327-329. [PMID: 28078771 DOI: 10.1111/ijd.13536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/03/2016] [Accepted: 11/14/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Mauricio Goihman-Yahr
- Chairs of Dermatology and Immunology, Instituto Nacional de Dermatología (now Instituto de Biomedicina,"Jacinto Convit"), Vargas School of Medicine, Central University of Venezuela, Caracas, Venezuela
| |
Collapse
|
21
|
Sharma R, Di Dalmazi G, Caturegli P. Exacerbation of Autoimmune Thyroiditis by CTLA-4 Blockade: A Role for IFNγ-Induced Indoleamine 2, 3-Dioxygenase. Thyroid 2016; 26:1117-24. [PMID: 27296629 PMCID: PMC4976247 DOI: 10.1089/thy.2016.0092] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cytotoxic T-lymphocyte associated protein 4 (CTLA-4) is a negative regulator of immune responses that suppresses the activity of effector T cells and contributes to the maintenance of self tolerance. When blocked therapeutically, CTLA-4 leads to an overall activation of T cells that has been exploited for cancer control, a control associated however with a variety of immune-related side effects such as autoimmune thyroiditis. To investigate the mechanism(s) underlying this form of thyroiditis, we used the NOD-H2(h4) mouse, a model that develops thyroiditis at very high incidence after addition of iodine to the drinking water. METHODS NOD-H2(h4) mice were started on drinking water supplemented with 0.05% sodium iodide when 8 weeks old and then injected with a hamster monoclonal antibody against mouse CTLA-4, polyclonal hamster immunoglobulins, or phosphate buffered saline when 11 weeks old. One month later (15 weeks of age), mice were sacrificed to assess thyroiditis, general immune responses in blood and spleen, and expression of indoleamine 2, 3-dioxygenase (IDO) in the thyroid and in isolated antigen-presenting cells after stimulation with interferon gamma. The study also analyzed IDO expression in four autopsy cases of metastatic melanoma who had received treatment with a CTLA-4 blocking antibody, and six surgical pathology Hashimoto thyroiditis controls. RESULTS CTLA-4 blockade worsened autoimmune thyroiditis, as assessed by a greater incidence, a more aggressive mononuclear cell infiltration in thyroids, and higher thyroglobulin antibody levels when compared to the control groups. CTLA-4 blockade also expanded the proportion of splenic CD4+ effector T cells, as well as the production of interleukin (IL)-2, interferon gamma, IL-10, and IL-13 cytokines. Interestingly, CTLA-4 blockade induced a strong expression of IDO in mouse and human thyroid glands, an expression that could represent a counter-regulatory mechanism to protect against the inflammatory environment. CONCLUSIONS This study shows that CTLA-4 blockade exacerbates the iodine-accelerated form of thyroiditis typical of the NOD-H2(h4) mouse. The study could also have implications for cancer patients who develop thyroiditis as an immune-related adverse event after CTLA-4 blockade.
Collapse
Affiliation(s)
- Rajni Sharma
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Giulia Di Dalmazi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Medicine, G. d'Annunzio University of Chieti, Cheti, Italy
| | - Patrizio Caturegli
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
22
|
Cameron AR, Morrison VL, Levin D, Mohan M, Forteath C, Beall C, McNeilly AD, Balfour DJK, Savinko T, Wong AKF, Viollet B, Sakamoto K, Fagerholm SC, Foretz M, Lang CC, Rena G. Anti-Inflammatory Effects of Metformin Irrespective of Diabetes Status. Circ Res 2016; 119:652-65. [PMID: 27418629 PMCID: PMC4990459 DOI: 10.1161/circresaha.116.308445] [Citation(s) in RCA: 481] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: The diabetes mellitus drug metformin is under investigation in cardiovascular disease, but the molecular mechanisms underlying possible benefits are poorly understood. Objective: Here, we have studied anti-inflammatory effects of the drug and their relationship to antihyperglycemic properties. Methods and Results: In primary hepatocytes from healthy animals, metformin and the IKKβ (inhibitor of kappa B kinase) inhibitor BI605906 both inhibited tumor necrosis factor-α–dependent IκB degradation and expression of proinflammatory mediators interleukin-6, interleukin-1β, and CXCL1/2 (C-X-C motif ligand 1/2). Metformin suppressed IKKα/β activation, an effect that could be separated from some metabolic actions, in that BI605906 did not mimic effects of metformin on lipogenic gene expression, glucose production, and AMP-activated protein kinase activation. Equally AMP-activated protein kinase was not required either for mitochondrial suppression of IκB degradation. Consistent with discrete anti-inflammatory actions, in macrophages, metformin specifically blunted secretion of proinflammatory cytokines, without inhibiting M1/M2 differentiation or activation. In a large treatment naive diabetes mellitus population cohort, we observed differences in the systemic inflammation marker, neutrophil to lymphocyte ratio, after incident treatment with either metformin or sulfonylurea monotherapy. Compared with sulfonylurea exposure, metformin reduced the mean log-transformed neutrophil to lymphocyte ratio after 8 to 16 months by 0.09 U (95% confidence interval, 0.02–0.17; P=0.013) and increased the likelihood that neutrophil to lymphocyte ratio would be lower than baseline after 8 to 16 months (odds ratio, 1.83; 95% confidence interval, 1.22–2.75; P=0.00364). Following up these findings in a double-blind placebo controlled trial in nondiabetic heart failure (trial registration: NCT00473876), metformin suppressed plasma cytokines including the aging-associated cytokine CCL11 (C-C motif chemokine ligand 11). Conclusion: We conclude that anti-inflammatory properties of metformin are exerted irrespective of diabetes mellitus status. This may accelerate investigation of drug utility in nondiabetic cardiovascular disease groups. Clinical Trial Registration: Name of the trial registry: TAYSIDE trial (Metformin in Insulin Resistant Left Ventricular [LV] Dysfunction). URL: https://www.clinicaltrials.gov. Unique identifier: NCT00473876.
Collapse
Affiliation(s)
- Amy R Cameron
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Vicky L Morrison
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Daniel Levin
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Mohapradeep Mohan
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Calum Forteath
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Craig Beall
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Alison D McNeilly
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - David J K Balfour
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Terhi Savinko
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Aaron K F Wong
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Benoit Viollet
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Kei Sakamoto
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Susanna C Fagerholm
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Marc Foretz
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.)
| | - Chim C Lang
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.).
| | - Graham Rena
- From the Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School (A.R.C., D.L., M.M., C.F., C.B., A.D.M., A.K.F.W., C.C.L., G.R.) and Division of Neuroscience, Ninewells Hospital and Medical School (D.J.K.B.), MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences (K.S.), University of Dundee, Scotland, United Kingdom; Institute of Biotechnology, University of Helsinki, Finland (V.L.M., T.S., S.C.F.); INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, France (B.V., M.F.); and Institute of Infection, Immunity, and Inflammation, University of Glasgow, United Kingdom (V.L.M.).
| |
Collapse
|
23
|
Ferretti C, La Cava A. Adaptive immune regulation in autoimmune diabetes. Autoimmun Rev 2016; 15:236-41. [DOI: 10.1016/j.autrev.2015.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 11/19/2015] [Indexed: 12/16/2022]
|
24
|
Abstract
The undesired destruction of healthy cells, either endogenous or transplanted, by the immune system results in the loss of tissue function or limits strategies to restore tissue function. Current therapies typically involve nonspecific immunosuppression that may prevent the appropriate response to an antigen, thereby decreasing humoral immunity and increasing the risks of patient susceptibility to opportunistic infections, viral reactivation, and neoplasia. The induction of antigen-specific immunological tolerance to block undesired immune responses to self- or allogeneic antigens, while maintaining the integrity of the remaining immune system, has the potential to transform the current treatment of autoimmune disease and serve as a key enabling technology for therapies based on cell transplantation.
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Division of Nephrology and Hypertension.,Comprehensive Cancer Center, and
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; ,
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109;
| |
Collapse
|
25
|
Kumai T, Kobayashi H, Harabuchi Y. Novel targets for natural killer/T-cell lymphoma immunotherapy. Immunotherapy 2015; 8:45-55. [PMID: 26642249 DOI: 10.2217/imt.15.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Extranodal natural killer/T-cell lymphoma, nasal type (NKTL) is a rare but highly aggressive Epstein-Barr virus-related malignancy, which mainly occurs in nasopharyngeal and nasal/paranasal areas. In addition to its high prevalence in Asian, Central American and South American populations, its incidence rate has been gradually increasing in Western countries. The current mainstay of treatment is a combination of multiple chemotherapies and irradiation. Although chemoradiotherapy can cure NKTL, it often causes severe and fatal adverse events. Because a growing body of evidence suggests that immunotherapy is effective against hematological malignancies, this treatment could provide an alternative to chemoradiotherapy for treatment of NKTL. In this review, we focus on how recent findings could be used to develop efficient immunotherapies against NKTL.
Collapse
Affiliation(s)
- Takumi Kumai
- Department of Otolaryngology, Head & Neck Surgery, Asahikawa Medical University, Midorigaoka-East 2-1-1-1, Asahikawa 078-8510, Japan
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Midorigaoka-East 2-1-1-1, Asahikawa 078-8510, Japan
| | - Yasuaki Harabuchi
- Department of Otolaryngology, Head & Neck Surgery, Asahikawa Medical University, Midorigaoka-East 2-1-1-1, Asahikawa 078-8510, Japan.,Department of Pathology, Asahikawa Medical University, Midorigaoka-East 2-1-1-1, Asahikawa 078-8510, Japan
| |
Collapse
|
26
|
Dumont CM, Park J, Shea LD. Controlled release strategies for modulating immune responses to promote tissue regeneration. J Control Release 2015; 219:155-166. [PMID: 26264833 DOI: 10.1016/j.jconrel.2015.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 01/06/2023]
Abstract
Advances in the field of tissue engineering have enhanced the potential of regenerative medicine, yet the efficacy of these strategies remains incomplete, and is limited by the innate and adaptive immune responses. The immune response associated with injury or disease combined with that mounted to biomaterials, transplanted cells, proteins, and gene therapies vectors can contribute to the inability to fully restore tissue function. Blocking immune responses such as with anti-inflammatory or immunosuppressive agents are either ineffective, as the immune response contributes significantly to regeneration, or have significant side effects. This review describes targeted strategies to modulate the immune response in order to limit tissue damage following injury, promote an anti-inflammatory environment that leads to regeneration, and induce antigen (Ag)-specific tolerance that can target degenerative diseases that destroy tissues and promote engraftment of transplanted cells. Focusing on targeted immuno-modulation, we describe local delivery techniques to sites of inflammation as well as systemic approaches that preferentially target subsets of immune populations.
Collapse
Affiliation(s)
- Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|