1
|
Kattelus R, Starskaia I, Lindén M, Batkulwar K, Pietilä S, Moulder R, Marson A, Rasool O, Suomi T, Elo LL, Lahesmaa R, Buchacher T. Phenotypic profiling of human induced regulatory T cells at early differentiation: insights into distinct immunosuppressive potential. Cell Mol Life Sci 2024; 81:399. [PMID: 39264416 PMCID: PMC11393232 DOI: 10.1007/s00018-024-05429-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024]
Abstract
Regulatory T cells (Tregs) play a key role in suppressing systemic effector immune responses, thereby preventing autoimmune diseases but also potentially contributing to tumor progression. Thus, there is great interest in clinically manipulating Tregs, but the precise mechanisms governing in vitro-induced Treg (iTreg) differentiation are not yet fully understood. Here, we used multiparametric mass cytometry to phenotypically profile human iTregs during the early stages of in vitro differentiation at single-cell level. A panel of 25 metal-conjugated antibodies specific to markers associated with human Tregs was used to characterize these immunomodulatory cells. We found that iTregs highly express the transcription factor FOXP3, as well as characteristic Treg-associated surface markers (e.g. CD25, PD1, CD137, CCR4, CCR7, CXCR3, and CD103). Expression of co-inhibitory factors (e.g. TIM3, LAG3, and TIGIT) increased slightly at late stages of iTreg differentiation. Further, CD103 was upregulated on a subpopulation of iTregs with greater suppressive capacity than their CD103- counterparts. Using mass-spectrometry-based proteomics, we showed that sorted CD103+ iTregs express factors associated with immunosuppression. Overall, our study highlights that during early stages of differentiation, iTregs resemble memory-like Treg features with immunosuppressive activity, and provides opportunities for further investigation into the molecular mechanisms underlying Treg function.
Collapse
Affiliation(s)
- Roosa Kattelus
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Inna Starskaia
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Markus Lindén
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Kedar Batkulwar
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Sami Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Robert Moulder
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, 94158, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Omid Rasool
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Tanja Buchacher
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
| |
Collapse
|
2
|
Lee BH, Bang YJ, Lim SH, Kang SJ, Kim SH, Kim-Schulze S, Park CG, Kim HJ, Kim TG. High-dimensional profiling of regulatory T cells in psoriasis reveals an impaired skin-trafficking property. EBioMedicine 2024; 100:104985. [PMID: 38306895 PMCID: PMC10847473 DOI: 10.1016/j.ebiom.2024.104985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/21/2023] [Accepted: 01/13/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease with a Th17-skewed immune phenotype. Although it has been generally accepted that regulatory T cells (Tregs) in lesional psoriatic skin have functional impairment due to the local inflammatory microenvironment, the molecular properties of skin-homing psoriatic Tregs have not been well explored. METHODS We designed an extensive 39 marker mass cytometry (CyTOF) panel to deeply profile the immune landscape of skin-homing Tregs from 31 people with psoriasis stratified by psoriasis area severity index score as mild (n = 15) to moderate-severe (n = 16) and 32 healthy controls. We further validated the findings with an in-vitro chemokine-mediated Treg migration assay, immunofluorescent imaging of normal and psoriatic lesional skin and analysed public single-cell RNA-sequencing datasets to expand upon our findings into the local tissue microenvironments. FINDINGS We discovered an overall decrease in CLAhi Tregs and specifically, CLAhiCCR5+ Tregs in psoriasis. Functional markers CD39 and FoxP3 were elevated in psoriatic Tregs. However, CCR7 expression was significantly increased while CCR4 and CLA expression was reduced in psoriatic Tregs and CLAhi Tregs, which was associated with disease severity. Moreover, psoriatic Tregs revealed increased migratory capacity towards CCR7's ligands, CCL19/CCL21. Interrogation of public single-cell RNA sequencing data confirmed reduced expression of skin-trafficking markers in lesional-skin Tregs compared to non-lesioned skin, further substantiated by immunofluorescent staining. INTERPRETATION Psoriatic circulating Tregs showed an impaired skin-trafficking phenotype thus leading to insufficient suppression of ongoing inflammation in the lesional skin, expanding upon our current understanding of the impairment of Treg-mediated immunosuppression in psoriasis. FUNDING This research was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Science and Information and Communications Technology (2020R1C1C1014513, 2021R1A4A5032185, 2020R1F1A1073692); and the new faculty research seed money grant of Yonsei University College of Medicine for 2021 (2021-32-0033).
Collapse
Affiliation(s)
- Brian Hyohyoung Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yoon Ji Bang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Sung Ha Lim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Seong-Jun Kang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Sung Hee Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chung-Gyu Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea; Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, South Korea; Transplantation Research Institute, Seoul National University Medical Research Center, Seoul, South Korea; Seoul National University Hospital, Seoul, South Korea.
| | - Hyun Je Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea; Cancer Research Institute, Seoul National University, Seoul, South Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea; Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, South Korea; Seoul National University Hospital, Seoul, South Korea; Genome Medicine Institute, Seoul National University Medical Research Center, Seoul, South Korea.
| | - Tae-Gyun Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
3
|
Lekan AA, Weiner LM. The Role of Chemokines in Orchestrating the Immune Response to Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:559. [PMID: 38339310 PMCID: PMC10854906 DOI: 10.3390/cancers16030559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Chemokines are small molecules that function as chemotactic factors which regulate the migration, infiltration, and accumulation of immune cells. Here, we comprehensively assess the structural and functional role of chemokines, examine the effects of chemokines that are present in the pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME), specifically those produced by cancer cells and stromal components, and evaluate their impact on immune cell trafficking, both in promoting and suppressing anti-tumor responses. We further explore the impact of chemokines on patient outcomes in PDAC and their role in the context of immunotherapy treatments, and review clinical trials that have targeted chemokine receptors and ligands in the treatment of PDAC. Lastly, we highlight potential strategies that can be utilized to harness chemokines in order to increase cytotoxic immune cell infiltration and the anti-tumor effects of immunotherapy.
Collapse
Affiliation(s)
| | - Louis M. Weiner
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC 20057, USA;
| |
Collapse
|
4
|
Jarosch S, Köhlen J, Ghimire S, Orberg ET, Hammel M, Gaag D, Evert M, Janssen KP, Hiergeist A, Gessner A, Weber D, Meedt E, Poeck H, D'Ippolito E, Holler E, Busch DH. Multimodal immune cell phenotyping in GI biopsies reveals microbiome-related T cell modulations in human GvHD. Cell Rep Med 2023; 4:101125. [PMID: 37467715 PMCID: PMC10394271 DOI: 10.1016/j.xcrm.2023.101125] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/13/2023] [Accepted: 06/22/2023] [Indexed: 07/21/2023]
Abstract
Acute graft-versus-host disease (aGvHD) is a significant complication after allogeneic hematopoietic stem cell transplantation (aHSCT), but major factors determining disease severity are not well defined yet. By combining multiplexed tissue imaging and single-cell RNA sequencing on gastrointestinal biopsies from aHSCT-treated individuals with fecal microbiome analysis, we link high microbiome diversity and the abundance of short-chain fatty acid-producing bacteria to the sustenance of suppressive regulatory T cells (Tregs). Furthermore, aGvHD severity strongly associates with the clonal expansion of mainly CD8 T cells, which we find distributed over anatomically distant regions of the gut, persistent over time, and inversely correlated with the presence of suppressive Tregs. Overall, our study highlights the pathophysiological importance of expanded CD8 T cell clones in the progression of aGvHD toward more severe clinical manifestations and strongly supports the further development of microbiome interventions as GvHD treatment via repopulation of the gut Treg niche to suppress inflammation.
Collapse
Affiliation(s)
- Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, 88397 Biberach an der Riß, Germany
| | - Jan Köhlen
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Sakhila Ghimire
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany
| | - Erik Thiele Orberg
- Department of Medicine III, Technical University of Munich (TUM), School of Medicine, Klinikum rechts der Isar TUM, 81675 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Doris Gaag
- Institute for Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Matthias Evert
- Institute for Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany
| | - Daniela Weber
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany
| | - Elisabeth Meedt
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany; Leibniz Institute for Immuntherapie (LIT), Regensburg, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Ernst Holler
- Department of Internal Medicine 3, University Medical Center, 93053 Regensburg, Germany.
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), 81675 Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany.
| |
Collapse
|
5
|
Stone TW, Clanchy FIL, Huang YS, Chiang NY, Darlington LG, Williams RO. An integrated cytokine and kynurenine network as the basis of neuroimmune communication. Front Neurosci 2022; 16:1002004. [PMID: 36507331 PMCID: PMC9729788 DOI: 10.3389/fnins.2022.1002004] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Two of the molecular families closely associated with mediating communication between the brain and immune system are cytokines and the kynurenine metabolites of tryptophan. Both groups regulate neuron and glial activity in the central nervous system (CNS) and leukocyte function in the immune system, although neither group alone completely explains neuroimmune function, disease occurrence or severity. This essay suggests that the two families perform complementary functions generating an integrated network. The kynurenine pathway determines overall neuronal excitability and plasticity by modulating glutamate receptors and GPR35 activity across the CNS, and regulates general features of immune cell status, surveillance and tolerance which often involves the Aryl Hydrocarbon Receptor (AHR). Equally, cytokines and chemokines define and regulate specific populations of neurons, glia or immune system leukocytes, generating more specific responses within restricted CNS regions or leukocyte populations. In addition, as there is a much larger variety of these compounds, their homing properties enable the superimposition of dynamic variations of cell activity upon local, spatially limited, cell populations. This would in principle allow the targeting of potential treatments to restricted regions of the CNS. The proposed synergistic interface of 'tonic' kynurenine pathway affecting baseline activity and the superimposed 'phasic' cytokine system would constitute an integrated network explaining some features of neuroimmune communication. The concept would broaden the scope for the development of new treatments for disorders involving both the CNS and immune systems, with safer and more effective agents targeted to specific CNS regions.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom,*Correspondence: Trevor W. Stone,
| | - Felix I. L. Clanchy
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Yi-Shu Huang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Nien-Yi Chiang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - L. Gail Darlington
- Department of Internal Medicine, Ashtead Hospital, Ashtead, United Kingdom
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Cheung J, Zahorowska B, Suranyi M, Wong JKW, Diep J, Spicer ST, Verma ND, Hodgkinson SJ, Hall BM. CD4 +CD25 + T regulatory cells in renal transplantation. Front Immunol 2022; 13:1017683. [PMID: 36426347 PMCID: PMC9681496 DOI: 10.3389/fimmu.2022.1017683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/13/2022] [Indexed: 09/14/2023] Open
Abstract
The immune response to an allograft activates lymphocytes with the capacity to cause rejection. Activation of CD4+CD25+Foxp3+T regulatory cells (Treg) can down-regulate allograft rejection and can induce immune tolerance to the allograft. Treg represent <10% of peripheral CD4+T cells and do not markedly increase in tolerant hosts. CD4+CD25+Foxp3+T cells include both resting and activated Treg that can be distinguished by several markers, many of which are also expressed by effector T cells. More detailed characterization of Treg to identify increased activated antigen-specific Treg may allow reduction of non-specific immunosuppression. Natural thymus derived resting Treg (tTreg) are CD4+CD25+Foxp3+T cells and only partially inhibit alloantigen presenting cell activation of effector cells. Cytokines produced by activated effector cells activate these tTreg to more potent alloantigen-activated Treg that may promote a state of operational tolerance. Activated Treg can be distinguished by several molecules they are induced to express, or whose expression they have suppressed. These include CD45RA/RO, cytokine receptors, chemokine receptors that alter pathways of migration and transcription factors, cytokines and suppression mediating molecules. As the total Treg population does not increase in operational tolerance, it is the activated Treg which may be the most informative to monitor. Here we review the methods used to monitor peripheral Treg, the effect of immunosuppressive regimens on Treg, and correlations with clinical outcomes such as graft survival and rejection. Experimental therapies involving ex vivo Treg expansion and administration in renal transplantation are not reviewed.
Collapse
Affiliation(s)
- Jason Cheung
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
| | | | - Michael Suranyi
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | | | - Jason Diep
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Stephen T. Spicer
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Nirupama D. Verma
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Suzanne J. Hodgkinson
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Bruce M. Hall
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| |
Collapse
|
7
|
Fu J, Lehmann CHK, Wang X, Wahlbuhl M, Allabauer I, Wilde B, Amon L, Dolff S, Cesnjevar R, Kribben A, Woelfle J, Rascher W, Hoyer PF, Dudziak D, Witzke O, Hoerning A. CXCR4 blockade reduces the severity of murine heart allograft rejection by plasmacytoid dendritic cell-mediated immune regulation. Sci Rep 2021; 11:23815. [PMID: 34893663 PMCID: PMC8664946 DOI: 10.1038/s41598-021-03115-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Allograft-specific regulatory T cells (Treg cells) are crucial for long-term graft acceptance after transplantation. Although adoptive Treg cell transfer has been proposed, major challenges include graft-specificity and stability. Thus, there is an unmet need for the direct induction of graft-specific Treg cells. We hypothesized a synergism of the immunotolerogenic effects of rapamycin (mTOR inhibition) and plerixafor (CXCR4 antagonist) for Treg cell induction. Thus, we performed fully-mismatched heart transplantations and found combination treatment to result in prolonged allograft survival. Moreover, fibrosis and myocyte lesions were reduced. Although less CD3+ T cell infiltrated, higher Treg cell numbers were observed. Noteworthy, this was accompanied by a plerixafor-dependent plasmacytoid dendritic cells-(pDCs)-mobilization. Furthermore, in vivo pDC-depletion abrogated the plerixafor-mediated Treg cell number increase and reduced allograft survival. Our pharmacological approach allowed to increase Treg cell numbers due to pDC-mediated immune regulation. Therefore pDCs can be an attractive immunotherapeutic target in addition to plerixafor treatment.
Collapse
Affiliation(s)
- Jian Fu
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany.,The Emergency and Trauma Center, The First Affiliated Hospital of Hai Nan Medical University, Haikou, China
| | - Christian H K Lehmann
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Research Module II, Hartmannstr. 14, 91052, Erlangen, Germany. .,Medical Immunology Campus and German Centre for Immuntherapy (Deutsches Zentrum für Immuntherapie-DZI) Erlangen, FAU Erlangen-Nürnberg, 91054, Erlangen, Germany.
| | - Xinning Wang
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Mandy Wahlbuhl
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Ida Allabauer
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lukas Amon
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Research Module II, Hartmannstr. 14, 91052, Erlangen, Germany
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, Universitätsmedizin Essen, University Duisburg-Essen, Essen, Germany
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany.,Department of Cardiac Surgery, Universitäts-Kinderspital Zürich, Zurich, Switzerland
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Joachim Woelfle
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Wolfgang Rascher
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Peter F Hoyer
- Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children's Hospital Essen, University Duisburg-Essen, Duisburg, Germany
| | - Diana Dudziak
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Research Module II, Hartmannstr. 14, 91052, Erlangen, Germany.,Medical Immunology Campus and German Centre for Immuntherapy (Deutsches Zentrum für Immuntherapie-DZI) Erlangen, FAU Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, Universitätsmedizin Essen, University Duisburg-Essen, Essen, Germany
| | - André Hoerning
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Loschgestrasse 15, 91054, Erlangen, Germany. .,Department of Pediatrics II, Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, Children's Hospital Essen, University Duisburg-Essen, Duisburg, Germany.
| |
Collapse
|
8
|
Mammadli M, Harris R, Suo L, May A, Gentile T, Waickman AT, Bah A, August A, Nurmemmedov E, Karimi M. Interleukin-2-inducible T-cell kinase (Itk) signaling regulates potent noncanonical regulatory T cells. Clin Transl Med 2021; 11:e625. [PMID: 34919342 PMCID: PMC8679839 DOI: 10.1002/ctm2.625] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) play an important role in controlling autoimmunity and limiting tissue damage and inflammation. IL2-inducible T cell kinase (Itk) is part of the Tec family of tyrosine kinases and is a critical component of T cell receptor mediated signaling. Here, we showed that either genetic ablation of Itk signaling or inhibition of Itk signaling pathways resulted in increased frequency of "noncanonical" CD4+ CD25- FOXP3+ Tregs (ncTregs), as well as of "canonical" CD4+ CD25+ FOXP3+ Tregs (canTregs). Using in vivo models, we showed that ncTregs can avert the formation of acute graft-versus-host disease (GVHD), in part by reducing conventional T cell proliferation, proinflammatory cytokine production, and tissue damage. This reduction in GVHD occurred without disruption of graft-versus-leukaemia (GVL) effects. RNA sequencing revealed that a number of effector, cell adhesion, and migration molecules were upregulated in Itk-/- ncTregs. Furthermore, disrupting the SLP76: ITK interaction using a specific peptide inhibitor led to enhanced Treg development in both mouse and primary human cells. This peptide inhibitor also significantly reduced inflammatory cytokine production in primary GVHD patient samples and mouse T cells without causing cell death or apoptosis. We provide evidence that specifically targeting Itk signaling could be a therapeutic strategy to treat autoimmune disorders.
Collapse
Affiliation(s)
- Mahinbanu Mammadli
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Rebecca Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Liye Suo
- Department of Pathology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Adriana May
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Teresa Gentile
- Department of Hematology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Adam T Waickman
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Alaji Bah
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Elmar Nurmemmedov
- Department of Translational Neurosciences Saint John's Cancer Institute, Santa Monica, California, USA
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
9
|
Alderfer L, Hall E, Hanjaya-Putra D. Harnessing biomaterials for lymphatic system modulation. Acta Biomater 2021; 133:34-45. [PMID: 34118451 PMCID: PMC9113193 DOI: 10.1016/j.actbio.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. To bridge this knowledge gap, biomaterials can be used to investigate the lymphatic system and to provide novel understanding into complex disease processes, including cancer metastasis and inflammation. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. This review article focuses on recent advances in (i) biomaterials used for lymphatic vessel formation, (ii) models for studying lymphatic-immune cells interactions, (iii) pharmaceuticals and their interactions with the lymphatic system, (iv) and strategies for drug delivery via the lymphatic system. Finally, several challenges regarding adopting biomaterials for immunomodulation and future perspectives are discussed. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. This review article focuses on recent progresses in biomaterials to investigate the lymphatic system and to provide novel understanding into complex disease states. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. Finally, a number of challenges in adopting biomaterials for immunomodulation and future perspectives are discussed.
Collapse
|
10
|
Selck C, Dominguez-Villar M. Antigen-Specific Regulatory T Cell Therapy in Autoimmune Diseases and Transplantation. Front Immunol 2021; 12:661875. [PMID: 34054826 PMCID: PMC8160309 DOI: 10.3389/fimmu.2021.661875] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/27/2021] [Indexed: 12/30/2022] Open
Abstract
Regulatory T (Treg) cells are a heterogenous population of immunosuppressive T cells whose therapeutic potential for the treatment of autoimmune diseases and graft rejection is currently being explored. While clinical trial results thus far support the safety and efficacy of adoptive therapies using polyclonal Treg cells, some studies suggest that antigen-specific Treg cells are more potent in regulating and improving immune tolerance in a disease-specific manner. Hence, several approaches to generate and/or expand antigen-specific Treg cells in vitro or in vivo are currently under investigation. However, antigen-specific Treg cell therapies face additional challenges that require further consideration, including the identification of disease-relevant antigens as well as the in vivo stability and migratory behavior of Treg cells following transfer. In this review, we discuss these approaches and the potential limitations and describe prospective strategies to enhance the efficacy of antigen-specific Treg cell treatments in autoimmunity and transplantation.
Collapse
Affiliation(s)
- Claudia Selck
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | | |
Collapse
|
11
|
Recruitment and Expansion of Tregs Cells in the Tumor Environment-How to Target Them? Cancers (Basel) 2021; 13:cancers13081850. [PMID: 33924428 PMCID: PMC8069615 DOI: 10.3390/cancers13081850] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The immune response against cancer is generated by effector T cells, among them cytotoxic CD8+ T cells that destroy cancer cells and helper CD4+ T cells that mediate and support the immune response. This antitumor function of T cells is tightly regulated by a particular subset of CD4+ T cells, named regulatory T cells (Tregs), through different mechanisms. Even if the complete inhibition of Tregs would be extremely harmful due to their tolerogenic role in impeding autoimmune diseases in the periphery, the targeted blockade of their accumulation at tumor sites or their targeted depletion represent a major therapeutic challenge. This review focuses on the mechanisms favoring Treg recruitment, expansion and stabilization in the tumor microenvironment and the therapeutic strategies developed to block these mechanisms. Abstract Regulatory T cells (Tregs) are present in a large majority of solid tumors and are mainly associated with a poor prognosis, as their major function is to inhibit the antitumor immune response contributing to immunosuppression. In this review, we will investigate the mechanisms involved in the recruitment, amplification and stability of Tregs in the tumor microenvironment (TME). We will also review the strategies currently developed to inhibit Tregs’ deleterious impact in the TME by either inhibiting their recruitment, blocking their expansion, favoring their plastic transformation into other CD4+ T-cell subsets, blocking their suppressive function or depleting them specifically in the TME to avoid severe deleterious effects associated with Treg neutralization/depletion in the periphery and normal tissues.
Collapse
|
12
|
Fan F, Yoo HJ, Stock S, Wang L, Liu Y, Schubert ML, Wang S, Neuber B, Hückelhoven-Krauss A, Gern U, Schmitt A, Müller-Tidow C, Dreger P, Schmitt M, Sellner L. Ibrutinib for improved chimeric antigen receptor T-cell production for chronic lymphocytic leukemia patients. Int J Cancer 2020; 148:419-428. [PMID: 32683672 DOI: 10.1002/ijc.33212] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/05/2020] [Accepted: 07/03/2020] [Indexed: 12/28/2022]
Abstract
Chimeric antigen receptor T (CART) cells targeting CD19 have shown promising results in the treatment of chronic lymphocytic leukemia (CLL). However, efficacy seems to be inferior compared to diffuse large B-cell lymphoma or acute lymphoblastic leukemia. Impaired T-cell fitness of CLL patients may be involved in treatment failure. Less-differentiated naïve-like T cells play an important role in CART expansion and long-term persistence in vivo. These cells are sparse in CLL patients. Therefore, optimization of CART cell production protocols enriching less differentiated T cell subsets may overcome treatment resistance. The B-cell receptor inhibitor ibrutinib targeting Bruton's tyrosine kinase (BTK) is approved for the treatment of CLL. Besides BTK, ibrutinib additionally inhibits interleukin-2-inducible T-cell kinase (ITK) which is involved in T-cell differentiation. To evaluate the effect of ibrutinib on CART cell production, peripheral blood mononuclear cells from nine healthy donors and eight CLL patients were used to generate CART cells. T-cell expansion and phenotype, expression of homing and exhaustion makers as well as functionality of CART cells were evaluated. CART cell generation in the presence of ibrutinib resulted in increased cell viability and expansion of CLL patient-derived CART cells. Furthermore, ibrutinib enriched CART cells with less-differentiated naïve-like phenotype and decreased expression of exhaustion markers including PD-1, TIM-3 and LAG-3. In addition, ibrutinib increased the cytokine release capacity of CLL patient-derived CART cells. In summary, BTK/ITK inhibition with ibrutinib during CART cell culture can improve yield and function of CLL patient-derived CART cell products.
Collapse
Affiliation(s)
- Fuli Fan
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hyeon Joo Yoo
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Sophia Stock
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Lei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Yibin Liu
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Sanmei Wang
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Ulrike Gern
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Heidelberg, Germany.,Oncology Business Unit - Medical Affairs, Takeda Pharma Vertrieb GmbH & Co. KG, Berlin, Germany
| |
Collapse
|
13
|
Li CX, Yang XX, Wang HW, Li XC, Ng KTP, Lo CM, Man K. FTY720 Suppresses Liver Tumor Growth and Metastasis by Reducing Circulating Regulating T Cells and Enhancing the Anti-Tumor Effect of Rapamycin. Onco Targets Ther 2020; 13:4743-4754. [PMID: 32547103 PMCID: PMC7262652 DOI: 10.2147/ott.s234394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/25/2020] [Indexed: 01/13/2023] Open
Abstract
Background In this study, we aimed to study the effect of FTY720 treatment in reducing circulating Tregs level and then suppressing liver tumor metastasis after hepatectomy and I/R injury in animal models. Furthermore, we also investigated the synergistic anti-tumor effect of FTY720 combined with rapamycin on hepatocellular carcinoma. Methods The effect of FTY720 on suppressing Tregs mobilization and tumor metastasis after hepatectomy was investigated in an orthotopic liver tumor rat model with hepatectomy and hepatic ischemia/reperfusion (I/R) injury. The synergistic anti-tumor effect of FTY720 combined with rapamycin was further explored both in in vitro functional study and in orthotopic liver tumor mouse model. Results In rat model, hepatic I/R promoted tumor metastasis and increased circulating Tregs after hepatectomy. The treatment of FTY720 reduced liver tumor metastasis and the number of circulating Tregs. Furthermore, FTY720 enhanced the anti-tumor capacity of rapamycin by inhibiting tumor cell proliferation and migration in vitro and reducing tumor growth in vivo through suppressing hepatic stellate cell activation and tumor angiogenesis. Conclusion FTY720 suppressed liver tumor growth and metastasis by reducing the population of circulating Tregs and enhancing the anti-tumor effect of rapamycin. It was suggested that FTY720 single or combined with rapamycin might provide novel insight for suppressing tumor growth and metastasis for HCC patients.
Collapse
Affiliation(s)
- Chang Xian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu Province, People's Republic of China
| | - Xin Xiang Yang
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Hong Wei Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu Province, People's Republic of China
| | - Xiang Cheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu Province, People's Republic of China
| | - Kevin Tak-Pan Ng
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Kwan Man
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
14
|
Radhakrishnan RK, Thandi RS, Tripathi D, Paidipally P, McAllister MK, Mulik S, Samten B, Vankayalapati R. BCG vaccination reduces the mortality of Mycobacterium tuberculosis-infected type 2 diabetes mellitus mice. JCI Insight 2020; 5:133788. [PMID: 32161191 DOI: 10.1172/jci.insight.133788] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a significant risk factor for the development of active tuberculosis. In this study, we used a mouse model of type 2 diabetes mellitus (T2DM) to determine the effect of prior Bacillus Calmette-Guérin (BCG) vaccination on immune responses to Mycobacterium tuberculosis (Mtb) infection. We found that, at 6-7 months after Mtb infection, 90% of the Mtb-infected T2DM mice died, whereas only 50% of BCG-vaccinated T2DM-Mtb-infected mice died. Moreover, 40% of the PBS-treated uninfected T2DM mice and 30% of the uninfected BCG-vaccinated T2DM mice died, whereas all uninfected and infected nondiabetic mice survived. BCG vaccination was less effective in reducing the lung bacterial burden of Mtb-infected T2DM mice compared with Mtb-infected nondiabetic mice. BCG vaccination significantly reduced lung inflammation in Mtb-infected T2DM mice compared with that of unvaccinated T2DM mice infected with Mtb. Furthermore, reduced mortality of BCG-vaccinated Mtb-infected T2DM mice is associated with expansion of IL-13-producing CXCR3+ Tregs in the lungs of Mtb-infected T2DM mice. Recombinant IL-13 and Tregs from BCG-vaccinated Mtb-infected T2DM mice converted proinflammatory M1 macrophages to antiinflammatory M2 macrophages. Our findings suggest a potentially novel role for BCG in preventing excess inflammation and mortality in T2DM mice infected with Mtb.
Collapse
|
15
|
Emamnejad R, Sahraian M, Shakiba Y, Salehi Z, Masoomi A, Imani D, Najafi F, Laribi B, Shirzad H, Izad M. Circulating mesenchymal stem cells, stromal derived factor (SDF)-1 and IP-10 levels increased in clinically active multiple sclerosis patients but not in clinically stable patients treated with beta interferon. Mult Scler Relat Disord 2019; 35:233-238. [DOI: 10.1016/j.msard.2019.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 06/27/2019] [Accepted: 08/11/2019] [Indexed: 12/19/2022]
|
16
|
Deyhle MR, Hafen PS, Parmley J, Preece CN, Robison M, Sorensen JR, Jackson B, Eggett DL, Hancock CR, Hyldahl RD. CXCL10 increases in human skeletal muscle following damage but is not necessary for muscle regeneration. Physiol Rep 2019; 6:e13689. [PMID: 29696819 PMCID: PMC5917067 DOI: 10.14814/phy2.13689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 03/29/2018] [Indexed: 01/11/2023] Open
Abstract
CXCL10 is a chemokine for activated and memory T cells with many important immunological functions. We recently found that CXCL10 is upregulated in human muscle following contraction‐induced damage. No information is available on the role of CXCL10 in the context of muscle damage or repair. In this study, we confirm that CXCL10 is elevated in human muscle at 2 and 3 days following damage and perform cell culture and animal studies to examine the role of CXCL10 in muscle repair. CXCL10 did not impact proliferation rates of human primary myoblasts but it did promote myogenic differentiation in vitro, suggesting a possible direct impact on muscle regeneration. To test if CXCL10 was dispensable for effective muscle regeneration in vivo, we measured functional and histological markers of muscle repair out to 14 days postmuscle injury caused by a myotoxin in wild‐type (WT) mice and CXCL10 knockout (KO) mice. Between genotypes, no significant differences were found in loss or restoration of in situ muscle force, cross‐sectional area of newly formed myofibers, or the number of embryonic myosin heavy chain‐positive myofibers. In addition, KO animals were not deficient in T‐cell accumulation in the damaged muscle following injury. Gene expression of the other two ligands (CXCL9 and 11) that bind to the same receptor as CXCL10 were also elevated in the damaged muscle of KO mice. Thus, other ligands may have compensated for the lack of CXCL10 in the KO mice. We conclude that CXCL10 is not necessary for effective muscle regeneration.
Collapse
Affiliation(s)
- Michael R Deyhle
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Paul S Hafen
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Jacob Parmley
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Coray N Preece
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Marissa Robison
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Jacob R Sorensen
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Blake Jackson
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Dennis L Eggett
- Department of Statistics, Brigham Young University, Provo, Utah
| | - Chad R Hancock
- Department of Nutrition, Dietetics & Food Science, Brigham Young University, Provo, Utah
| | - Robert D Hyldahl
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| |
Collapse
|
17
|
Celiac Disease in Children, Particularly with Accompanying Type 1 Diabetes, Is Characterized by Substantial Changes in the Blood Cytokine Balance, Which May Reflect Inflammatory Processes in the Small Intestinal Mucosa. J Immunol Res 2019; 2019:6179243. [PMID: 31214623 PMCID: PMC6535873 DOI: 10.1155/2019/6179243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/08/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Cytokines play a pivotal role in the maintenance of intestinal homeostasis inducing pro- or anti-inflammatory response and mucosal barrier function in celiac disease (CD) and type 1 diabetes (T1D). We aimed to compare the levels of pro- and anti-inflammatory cytokines in CD patients without and with coexisting T1D, as well as to evaluate its association with the presence of enteroviruses (EV), regulatory T cells (Tregs), and dendritic cells (DCs) in small bowel mucosa. Altogether, 72 patients (median age 10.1 years) who had undergone small bowel biopsy were studied. The study group consisted of 24 patients with CD (median age 6.5 years), 9 patients with CD and concomitant T1D (median age 7.0 years), two patients with T1D (median age 8.5 years), and 37 patients (median age 14.0 years) with functional gastrointestinal disorders (FGD) and a normal small bowel mucosa as controls. The levels of 33 cytokines in serum were measured by multiple analysis using the Milliplex® MAP Magnetic Bead assay. The densities of FOXP3+ Tregs, CD11c+ DC, indoleamine 2,3-dioxygenase+ (IDO+) DC, langerin+ (CD207+) DCs, and EV were evaluated by immunohistochemistry as described in our previous studies. Circulating anti-EV IgA and IgG were evaluated using ELISA. The most important finding of the study is the significant increase of the serum levels of IL-5, IL-8, IL-13, IL-15, IL-17F, IL-22, IL-27, IP-10, MIP-1β, sIL-2Rα, sTNFRII, and TNFα in CD patients compared to controls and its correlation with the degree of small bowel mucosa damage graded according to the Marsh classification. The leptin level was higher in females in all study groups. The levels of IL-2, IL-6, IL-12 (P70), IL-15, IP-10, and IFNγ correlated significantly with the density of FOXP3+ Tregs in lamina propria of the small bowel mucosa, which supports the evidence about the signaling role of these cytokines in the peripheral maintenance of FOXP3+ Tregs. At the same time, a significant negative correlation occurred between the level of IL-4 and density of FOXP3+ Tregs in controls. Another important finding of our study was the correlation of IL-17F, IP-10, sTNFRII, MCP-1, and GM-CSF with the density of EV-positive cells in the lamina propria of the small bowel mucosa. Correlation of MIP-1 (CCL-4) with CD103+ DC and langerin+ DC densities may point to their significance in the recruitment of immune cells into the lamina propria and in driving the inflammatory response in CD patients. Our results suggest the predominance of Th1 and Th17 immune responses over EV VP1 protein in CD and T1D patients. The significant elevation of Th2 cytokines, like IL-5 and IL-13, but not IL-4, in CD patients and its correlation with the degree of small bowel mucosa damage could reflect the role of these cytokines in gut defense and inflammation.
Collapse
|
18
|
Chen J, Bhattacharya S, Sirota M, Laiudompitak S, Schaefer H, Thomson E, Wiser J, Sarwal MM, Butte AJ. Assessment of Postdonation Outcomes in US Living Kidney Donors Using Publicly Available Data Sets. JAMA Netw Open 2019; 2:e191851. [PMID: 30977847 PMCID: PMC6481454 DOI: 10.1001/jamanetworkopen.2019.1851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 02/12/2019] [Indexed: 12/30/2022] Open
Abstract
Importance There are limited resources providing postdonation conditions that can occur in living donors (LDs) of solid-organ transplant. Consequently, it is difficult to visualize and understand possible postdonation outcomes in LDs. Objective To assemble an open access resource that is representative of the demographic characteristics in the US national registry, maintained by the Organ Procurement and Transplantation Network and administered by the United Network for Organ Sharing, but contains more follow-up information to help to examine postdonation outcomes in LDs. Design, Setting, and Participants Cohort study in which the data for the resource and analyses stemmed from the transplant data set derived from 27 clinical studies from the ImmPort database, which is an open access repository for clinical studies. The studies included data collected from 1963 to 2016. Data from the United Network for Organ Sharing Organ Procurement and Transplantation Network national registry collected from October 1987 to March 2016 were used to determine representativeness. Data analysis took place from June 2016 to May 2018. Data from 20 ImmPort clinical studies (including clinical trials and observational studies) were curated, and a cohort of 11 263 LDs was studied, excluding deceased donors, LDs with 95% or more missing data, and studies without a complete data dictionary. The harmonization process involved the extraction of common features from each clinical study based on categories that included demographic characteristics as well as predonation and postdonation data. Main Outcomes and Measures Thirty-six postdonation events were identified, represented, and analyzed via a trajectory network analysis. Results The curated data contained 10 869 living kidney donors (median [interquartile range] age, 39 [31-48] years; 6175 [56.8%] women; and 9133 [86.6%] of European descent). A total of 9558 living kidney donors with postdonation data were analyzed. Overall, 1406 LDs (14.7%) had postdonation events. The 4 most common events were hypertension (806 [8.4%]), diabetes (190 [2.0%]), proteinuria (171 [1.8%]), and postoperative ileus (147 [1.5%]). Relatively few events (n = 269) occurred before the 2-year postdonation mark. Of the 1746 events that took place 2 years or more after donation, 1575 (90.2%) were nonsurgical; nonsurgical conditions tended to occur in the wide range of 2 to 40 years after donation (odds ratio, 38.3; 95% CI, 4.12-1956.9). Conclusions and Relevance Most events that occurred more than 2 years after donation were nonsurgical and could occur up to 40 years after donation. Findings support the construction of a national registry for long-term monitoring of LDs and confirm the value of secondary reanalysis of clinical studies.
Collapse
Affiliation(s)
- Jieming Chen
- Bakar Computational Health Sciences Institute, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
- Now with the Department of Bioinformatics and Computational Biology, Genentech, Inc, South San Francisco, California
| | - Sanchita Bhattacharya
- Bakar Computational Health Sciences Institute, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
| | - Sunisa Laiudompitak
- Bakar Computational Health Sciences Institute, University of California, San Francisco
| | | | | | - Jeff Wiser
- Northrop Grumman Information Systems Health IT, Rockville, Maryland
| | - Minnie M. Sarwal
- Department of Pediatrics, University of California, San Francisco
- Division of MultiOrgan Transplant, Department of Surgery and Medicine, University of California, San Francisco
| | - Atul J. Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
| |
Collapse
|
19
|
Emmerson A, Trevelin SC, Mongue-Din H, Becker PD, Ortiz C, Smyth LA, Peng Q, Elgueta R, Sawyer G, Ivetic A, Lechler RI, Lombardi G, Shah AM. Nox2 in regulatory T cells promotes angiotensin II-induced cardiovascular remodeling. J Clin Invest 2018; 128:3088-3101. [PMID: 29688896 PMCID: PMC6025997 DOI: 10.1172/jci97490] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 04/17/2018] [Indexed: 12/29/2022] Open
Abstract
The superoxide-generating enzyme Nox2 contributes to hypertension and cardiovascular remodeling triggered by activation of the renin-angiotensin system. Multiple Nox2-expressing cells are implicated in angiotensin II-induced (Ang II-induced) pathophysiology, but the importance of Nox2 in leukocyte subsets is poorly understood. Here, we investigated the role of Nox2 in T cells, particularly Tregs. Mice globally deficient in Nox2 displayed increased numbers of Tregs in the heart at baseline, whereas Ang II-induced effector T cell (Teff) infiltration was inhibited. To investigate the role of Treg Nox2, we generated a mouse line with CD4-targeted Nox2 deficiency (Nox2fl/flCD4Cre+). These animals showed inhibition of Ang II-induced hypertension and cardiac remodeling related to increased tissue-resident Tregs and reduction in infiltrating Teffs, including Th17 cells. The protection in Nox2fl/flCD4Cre+ mice was reversed by anti-CD25 antibody depletion of Tregs. Mechanistically, Nox2-/y Tregs showed higher in vitro suppression of Teff proliferation than WT Tregs, increased nuclear levels of FoxP3 and NF-κB, and enhanced transcription of CD25, CD39, and CD73. Adoptive transfer of Tregs confirmed that Nox2-deficient cells had greater inhibitory effects on Ang II-induced heart remodeling than WT cells. These results identify a previously unrecognized role of Nox2 in modulating suppression of Tregs, which acts to enhance hypertension and cardiac remodeling.
Collapse
Affiliation(s)
- Amber Emmerson
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Silvia Cellone Trevelin
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Heloise Mongue-Din
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Pablo D. Becker
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Carla Ortiz
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Lesley A. Smyth
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Qi Peng
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Raul Elgueta
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Greta Sawyer
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Aleksandar Ivetic
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Robert I. Lechler
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Giovanna Lombardi
- King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom
| | - Ajay M. Shah
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| |
Collapse
|
20
|
Wang D. The essential role of G protein-coupled receptor (GPCR) signaling in regulating T cell immunity. Immunopharmacol Immunotoxicol 2018; 40:187-192. [PMID: 29433403 DOI: 10.1080/08923973.2018.1434792] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AIM The aim of this paper is to clarify the critical role of GPCR signaling in T cell immunity. METHODS The G protein-coupled receptors (GPCRs) are the most common targets in current pharmaceutical industry, and represent the largest and most versatile family of cell surface communicating molecules. GPCRs can be activated by a diverse array of ligands including neurotransmitters, chemokines as well as sensory stimuli. Therefore, GPCRs are involved in many key cellular and physiological processes, such as sense of light, taste and smell, neurotransmission, metabolism, endocrine and exocrine secretion. In recent years, GPCRs have been found to play an important role in immune system. T cell is an important type of immune cell, which plays a central role in cell-mediated immunity. A variety of GPCRs and their signaling mediators (RGS proteins, GRKs and β-arrestin) have been found to express in T cells and involved T cell-mediated immunity. We will summarize the role of GPCR signaling and their regulatory molecules in T cell activation, homeostasis and function in this article. RESULTS GPCR signaling plays an important role in T cell activation, homeostasis and function. CONCLUSION GPCR signaling is critical in regulating T cell immunity.
Collapse
Affiliation(s)
- Dashan Wang
- a Molecular Biology Research Center, Key Medical Health Laboratory for Laboratory Medicine of Shandong Province, Department of Laboratory Medicine , Shandong Medical College , Linyi , Shandong , China
| |
Collapse
|
21
|
Alikhan MA, Huynh M, Kitching AR, Ooi JD. Regulatory T cells in renal disease. Clin Transl Immunology 2018; 7:e1004. [PMID: 29484182 PMCID: PMC5822411 DOI: 10.1002/cti2.1004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/10/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022] Open
Abstract
The kidney is vulnerable to injury, both acute and chronic from a variety of immune and metabolic insults, all of which at least to some degree involve inflammation. Regulatory T cells modulate systemic autoimmune and allogenic responses in glomerulonephritis and transplantation. Intrarenal regulatory T cells (Tregs), including those recruited to the kidney, have suppressive effects on both adaptive and innate immune cells, and probably also intrinsic kidney cells. Evidence from autoimmune glomerulonephritis implicates antigen-specific Tregs in HLA-mediated dominant protection, while in several human renal diseases Tregs are abnormal in number or phenotype. Experimentally, Tregs can protect the kidney from injury in a variety of renal diseases. Mechanisms of Treg recruitment to the kidney include via the chemokine receptors CCR6 and CXCR3 and potentially, at least in innate injury TLR9. The effects of Tregs may be context dependent, with evidence for roles for immunoregulatory roles both for endogenous Tbet-expressing Tregs and STAT-3-expressing Tregs in experimental glomerulonephritis. Most experimental work and some of the ongoing human trials in renal transplantation have focussed on unfractionated thymically derived Tregs (tTregs). However, induced Tregs (iTregs), type 1 regulatory T (Tr1) cells and in particular antigen-specific Tregs also have therapeutic potential not only in renal transplantation, but also in other kidney diseases.
Collapse
Affiliation(s)
- Maliha A Alikhan
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia
| | - Megan Huynh
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia.,Department of Nephrology Monash Health Clayton VIC Australia.,Department of Paediatric Nephrology Monash Health Clayton VIC Australia
| | - Joshua D Ooi
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia
| |
Collapse
|
22
|
Sula Karreci E, Eskandari SK, Dotiwala F, Routray SK, Kurdi AT, Assaker JP, Luckyanchykov P, Mihali AB, Maarouf O, Borges TJ, Alkhudhayri A, Patel KR, Radwan A, Ghobrial I, McGrath M, Chandraker A, Riella LV, Elyaman W, Abdi R, Lieberman J, Azzi J. Human regulatory T cells undergo self-inflicted damage via granzyme pathways upon activation. JCI Insight 2017; 2:91599. [PMID: 29093262 DOI: 10.1172/jci.insight.91599] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 09/25/2017] [Indexed: 12/13/2022] Open
Abstract
Tregs hold great promise as a cellular therapy for multiple immunologically mediated diseases, given their ability to control immune responses. The success of such strategies depends on the expansion of healthy, suppressive Tregs ex vivo and in vivo following the transfer. In clinical studies, levels of transferred Tregs decline sharply in the blood within a few days of the transfer. Tregs have a high rate of apoptosis. Here, we describe a new mechanism of Treg self-inflicted damage. We show that granzymes A and -B (GrA and GrB), which are highly upregulated in human Tregs upon stimulation, leak out of cytotoxic granules to induce cleavage of cytoplasmic and nuclear substrates, precipitating apoptosis in target cells. GrA and GrB substrates were protected from cleavage by inhibiting granzyme activity in vitro. Additionally, we show - by using cytometry by time of flight (CYTOF) - an increase in GrB-expressing Tregs in the peripheral blood and renal allografts of transplant recipients undergoing rejection. These GrB-expressing Tregs showed an activated phenotype but were significantly more apoptotic than non-GrB expressing Tregs. This potentially novel finding improves our understanding of Treg survival and suggests that manipulating Gr expression or activity might be useful for designing more effective Treg therapies.
Collapse
Affiliation(s)
- Esilida Sula Karreci
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Siawosh K Eskandari
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Farokh Dotiwala
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | - Sujit K Routray
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Ahmed T Kurdi
- Department of Medical Oncology, Dana-Farber Cancer Institute, and
| | - Jean Pierre Assaker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Pavlo Luckyanchykov
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Albana B Mihali
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Omar Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Thiago J Borges
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Abdullah Alkhudhayri
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Kruti R Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Amr Radwan
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Irene Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, and
| | - Martina McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Anil Chandraker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Leonardo V Riella
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Wassim Elyaman
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | - Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital
| |
Collapse
|
23
|
Sackstein R, Schatton T, Barthel SR. T-lymphocyte homing: an underappreciated yet critical hurdle for successful cancer immunotherapy. J Transl Med 2017; 97:669-697. [PMID: 28346400 PMCID: PMC5446300 DOI: 10.1038/labinvest.2017.25] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/17/2017] [Accepted: 01/22/2017] [Indexed: 12/13/2022] Open
Abstract
Advances in cancer immunotherapy have offered new hope for patients with metastatic disease. This unfolding success story has been exemplified by a growing arsenal of novel immunotherapeutics, including blocking antibodies targeting immune checkpoint pathways, cancer vaccines, and adoptive cell therapy (ACT). Nonetheless, clinical benefit remains highly variable and patient-specific, in part, because all immunotherapeutic regimens vitally hinge on the capacity of endogenous and/or adoptively transferred T-effector (Teff) cells, including chimeric antigen receptor (CAR) T cells, to home efficiently into tumor target tissue. Thus, defects intrinsic to the multi-step T-cell homing cascade have become an obvious, though significantly underappreciated contributor to immunotherapy resistance. Conspicuous have been low intralesional frequencies of tumor-infiltrating T-lymphocytes (TILs) below clinically beneficial threshold levels, and peripheral rather than deep lesional TIL infiltration. Therefore, a Teff cell 'homing deficit' may arguably represent a dominant factor responsible for ineffective immunotherapeutic outcomes, as tumors resistant to immune-targeted killing thrive in such permissive, immune-vacuous microenvironments. Fortunately, emerging data is shedding light into the diverse mechanisms of immune escape by which tumors restrict Teff cell trafficking and lesional penetrance. In this review, we scrutinize evolving knowledge on the molecular determinants of Teff cell navigation into tumors. By integrating recently described, though sporadic information of pivotal adhesive and chemokine homing signatures within the tumor microenvironment with better established paradigms of T-cell trafficking under homeostatic or infectious disease scenarios, we seek to refine currently incomplete models of Teff cell entry into tumor tissue. We further summarize how cancers thwart homing to escape immune-mediated destruction and raise awareness of the potential impact of immune checkpoint blockers on Teff cell homing. Finally, we speculate on innovative therapeutic opportunities for augmenting Teff cell homing capabilities to improve immunotherapy-based tumor eradication in cancer patients, with special focus on malignant melanoma.
Collapse
Affiliation(s)
- Robert Sackstein
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Program of Excellence in Glycosciences, Harvard Medical School, 77 Avenue Louis Pasteur, Rm 671, Boston, MA 02115, USA
| | - Tobias Schatton
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA,Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Steven R. Barthel
- Department of Dermatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Skin Disease Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA,Correspondence to: Dr. Steven R. Barthel, Harvard Institutes of Medicine, Rm. 673B, 77 Avenue Louis Pasteur, Boston, MA 02115;
| |
Collapse
|
24
|
Regulatory T Cells as Biomarkers for Rejection and Immunosuppression Tailoring in Solid Organ Transplantation. Ther Drug Monit 2016; 38 Suppl 1:S36-42. [PMID: 26977998 DOI: 10.1097/ftd.0000000000000265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The use of biomarkers to tailor immunosuppression and to predict graft and patient outcomes using biological samples obtained by non-invasive tests is one of the main objectives in solid organ transplantation. Although biopsies give the most accurate information, they are clearly invasive and are associated with potentially adverse effects. To date, regulatory T cells have been shown to play a role in allograft protection; for this reason, extensive research has been performed to define them as biomarkers. However, studies of the measurement of these cells in peripheral blood as biomarkers in solid organ transplantation have been very limited and still not validated in prospective randomized large cohorts with the use of standardized methodology. Such poor evidence has been almost exclusively obtained in renal transplantation. Available data summarized here point for their use as biomarkers in different clinical settings with discordant data in many cases.
Collapse
|
25
|
Li CX, Ling CC, Shao Y, Xu A, Li XC, Ng KTP, Liu XB, Ma YY, Qi X, Liu H, Liu J, Yeung OWH, Yang XX, Liu QS, Lam YF, Zhai Y, Lo CM, Man K. CXCL10/CXCR3 signaling mobilized-regulatory T cells promote liver tumor recurrence after transplantation. J Hepatol 2016; 65:944-952. [PMID: 27245433 DOI: 10.1016/j.jhep.2016.05.032] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/02/2016] [Accepted: 05/20/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND & AIMS Liver graft injury and tumor recurrence are the major challenges of liver transplantation for the patients with hepatocellular carcinoma (HCC). Here, we aimed to explore the role and mechanism of liver graft injury mobilizing regulatory T cells (Tregs), which lead to late phase tumor recurrence after liver transplantation. METHODS The correlation among tumor recurrence, liver graft injury and Tregs mobilization were studied in 257 liver transplant recipients with HCC and orthotopic rat liver transplantation models. The direct roles of CXCL10/CXCR3 signaling on Tregs mobilization and tumor recurrence were investigated in CXCL10-/- and CXCR3-/- mice models with hepatic IR injury. RESULTS Clinically, patients received the graft with graft weight ratio (GWR) <60% had higher HCC recurrence after liver transplantation than the recipients with GWR ⩾60% graft. More circulating Tregs and higher intragraft TLR4/CXCL10/CXCR3 levels were detected in recipients with GWR <60% graft. These results were further validated in rat transplantation model. Foxp3+ cells and expressions of TLR4, CXCL10, TGFβ, CTLA-4 and CD274 were increased in rat liver tumor tissues from small-for-size graft group. In mouse model, the mobilization and recruitment of Tregs were decreased in TLR4-/-, CXCL10-/- and CXCR3-/- mice compared to wild-type mice. Moreover, less CXCR3+ Tregs were recruited into liver in CXCL10-/- mice after hepatic IR injury. The knockout of CXCL10 and depletion of Tregs inhibited tumor recurrence after hepatic IR injury. CONCLUSION CXCL10/CXCR3 signaling upregulated at liver graft injury directly induced the mobilization and intragraft recruitment of Tregs, which further promoted HCC recurrence after transplantation. LAY SUMMARY There were positive correlation among tumor recurrence, circulating Tregs and liver graft injury after human transplantation for HCC patients. The knockout of CXCL10 decreased hepatic recruitment of CXCR3+ Tregs and late phase tumor recurrence after hepatic IR injury.
Collapse
Affiliation(s)
- Chang Xian Li
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Chang Chun Ling
- Department of Surgery, The University of Hong Kong, Hong Kong, China; Department of General Surgery, Affiliated Hospital of Nantong University, Nantong city, 226001, China
| | - Yan Shao
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Cheng Li
- Liver Transplantation Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kevin Tak-Pan Ng
- Department of Surgery, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| | - Xiao Bing Liu
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Yuen Yuen Ma
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Xiang Qi
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Hui Liu
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Jiang Liu
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | | | - Xin Xiang Yang
- Department of Surgery, The University of Hong Kong, Hong Kong, China; Liver Transplantation Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qing Sheng Liu
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Yin Fan Lam
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Yuan Zhai
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, USA
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| | - Kwan Man
- Department of Surgery, The University of Hong Kong, Hong Kong, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, China.
| |
Collapse
|
26
|
Jain NG, Wong EA, Aranyosi AJ, Boneschansker L, Markmann JF, Briscoe DM, Irimia D. Microfluidic mazes to characterize T-cell exploration patterns following activation in vitro. Integr Biol (Camb) 2016; 7:1423-31. [PMID: 26325525 DOI: 10.1039/c5ib00146c] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The migration of T-cell subsets within peripheral tissues is characteristic of inflammation and immunoregulation. In general, the lymphocyte migratory response is assumed directional and guided by local gradients of chemoattractants and/or chemorepellents. However, little is known about how cells explore their tissue environment, and whether lymphocyte activation may influence speed and exploratory patterns of migration. To probe migration patterns by T-cells we designed a microfluidic maze device that replicates critical features of a tissue-like microenvironment. We quantified the migration patterns of unstimulated and mitogen-activated human T-cells at single cell resolution and found significant differences in exploration within microfluidic mazes. While unstimulated lymphocytes migrated in a directed manner, activated T-cells migrated through large areas of the mazes in an exploratory pattern in response to the chemoattractants RANTES (CCL5) and IP-10 (CXCL10). The analysis of migration enabled by the microfluidic devices help develop new methods for determining how human circulating T-cells function in vivo to seek out antigens in health and disease states.
Collapse
Affiliation(s)
- Namrata G Jain
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Transplant Research Program, Boston Children's Hospital, Boston, MA 02115, USA.
| | - Elisabeth A Wong
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Alexander J Aranyosi
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Leo Boneschansker
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Transplant Research Program, Boston Children's Hospital, Boston, MA 02115, USA.
| | - James F Markmann
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Ave, MA 02139, USA
| | - David M Briscoe
- Transplant Research Program, Boston Children's Hospital, Boston, MA 02115, USA. and Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Ave, MA 02139, USA
| | - Daniel Irimia
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
27
|
Chemoattractant Signals and Adhesion Molecules Promoting Human Regulatory T Cell Recruitment to Porcine Endothelium. Transplantation 2016; 100:753-62. [PMID: 26720299 DOI: 10.1097/tp.0000000000001034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Human CD4+CD25+Foxp3+ T regulatory cells (huTreg) suppress CD4+ T cell-mediated antipig xenogeneic responses in vitro and might therefore be used to induce xenograft tolerance. The present study investigated the role of the adhesion molecules, their porcine ligands, and the chemoattractant factors that may promote the recruitment of huTreg to porcine aortic endothelial cells (PAEC) and their capacity to regulate antiporcine natural killer (NK) cell responses. METHODS Interactions between ex vivo expanded huTreg and PAEC were studied by static chemotaxis assays and flow-based adhesion and transmigration assays. In addition, the suppressive function of huTreg on human antiporcine NK cell responses was analyzed. RESULTS The TNFα-activated PAEC released factors that induce huTreg chemotaxis, partially inhibited by antihuman CXCR3 blocking antibodies. Coating of PAEC with human CCL17 significantly increased the transmigration of CCR4+ huTreg under physiological shear stress. Under static conditions, transendothelial Treg migration was inhibited by blocking integrin sub-units (CD18, CD49d) on huTreg, or their respective porcine ligands intercellular adhesion molecule 2 (CD102) and vascular cell adhesion molecule 1 (CD106). Finally, huTreg partially suppressed xenogeneic human NK cell adhesion, NK cytotoxicity and degranulation (CD107 expression) against PAEC; however, this inhibition was modest, and there was no significant change in the production of IFNγ. CONCLUSIONS Recruitment of huTreg to porcine endothelium depends on particular chemokine receptors (CXCR3, CCR4) and integrins (CD18 and CD49d) and was increased by CCL17 coating. These results will help to develop new strategies to enhance the recruitment of host huTreg to xenogeneic grafts to regulate cell-mediated xenograft rejection including NK cell responses.
Collapse
|
28
|
Nosko A, Kluger MA, Diefenhardt P, Melderis S, Wegscheid C, Tiegs G, Stahl RAK, Panzer U, Steinmetz OM. T-Bet Enhances Regulatory T Cell Fitness and Directs Control of Th1 Responses in Crescentic GN. J Am Soc Nephrol 2016; 28:185-196. [PMID: 27297951 DOI: 10.1681/asn.2015070820] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 04/05/2016] [Indexed: 12/11/2022] Open
Abstract
Th1 cells are central pathogenic mediators of crescentic GN (cGN). Mechanisms responsible for Th1 cell downregulation, however, remain widely unknown. Recently, it was proposed that activation of the Th1-characteristic transcription factor T-bet optimizes Foxp3+ regulatory T (Treg) cells to counteract Th1-type inflammation. Because very little is known about the role of T-bet+ Treg1 cells in inflammatory diseases, we studied the function of these cells in the nephrotoxic nephritis (NTN) model of cGN. The percentage of Treg1 cells progressively increased in kidneys of nephritic wild-type mice during the course of NTN, indicating their functional importance. Notably, naïve Foxp3CrexT-betfl/fl mice, lacking Treg1 cells, showed spontaneous skewing toward Th1 immunity. Furthermore, absence of Treg1 cells resulted in aggravated NTN with selectively dysregulated renal and systemic Th1 responses. Detailed analyses of Treg cells from Foxp3CrexT-betfl/fl mice revealed unaltered cytokine production and suppressive capacity. However, in competitive cotransfer experiments, wild-type Treg cells outcompeted T-bet-deficient Treg cells in terms of population expansion and expression levels of Foxp3, indicating that T-bet expression is crucial for general Treg fitness. Additionally, T-bet-deficient Treg cells lacked expression of the Th1-characteristic trafficking receptor CXCR3, which correlated with significant impairment of renal Treg infiltration. In summary, our data indicate a new subtype of Treg cells in cGN. These Treg1 cells are characterized by activation of the transcription factor T-bet, which enhances the overall fitness of these cells and optimizes their capacity to downregulate Th1 responses by inducing chemokine receptor CXCR3 expression.
Collapse
Affiliation(s)
| | | | | | | | - Claudia Wegscheid
- Institut für experimentelle Immunologie und Hepatologie, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - Gisa Tiegs
- Institut für experimentelle Immunologie und Hepatologie, Universitätsklinikum Eppendorf, Hamburg, Germany
| | | | | | | |
Collapse
|
29
|
Wang P, Han W, Ma D. Electronic Sorting of Immune Cell Subpopulations Based on Highly Plastic Genes. THE JOURNAL OF IMMUNOLOGY 2016; 197:665-73. [PMID: 27288532 DOI: 10.4049/jimmunol.1502552] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/17/2016] [Indexed: 12/14/2022]
Abstract
Immune cells are highly heterogeneous and plastic with regard to gene expression and cell phenotype. In this study, we categorized genes into those with low and high gene plasticity, and those categories revealed different functions and applications. We proposed that highly plastic genes could be suited for the labeling of immune cell subpopulations; thus, novel immune cell subpopulations could be identified by gene plasticity analysis. For this purpose, we systematically analyzed highly plastic genes in human and mouse immune cells. In total, 1,379 human and 883 mouse genes were identified as being extremely plastic. We also expanded our previous immunoinformatic method, electronic sorting, which surveys big data to perform virtual analysis. This approach used correlation analysis and took dosage changes into account, which allowed us to identify the differentially expressed genes. A test with human CD4(+) T cells supported the method's feasibility, effectiveness, and predictability. For example, with the use of human nonregulatory T cells, we found that FOXP3(hi)CD4(+) T cells were highly expressive of certain known molecules, such as CD25 and CTLA4, and that this process of investigation did not require isolating or inducing these immune cells in vitro. Therefore, the sorting process helped us to discover the potential signature genes or marker molecules and to conduct functional evaluations for immune cell subpopulations. Finally, in human CD4(+) T cells, 747 potential immune cell subpopulations and their candidate signature genes were identified, which provides a useful resource for big data-driven knowledge discoveries.
Collapse
Affiliation(s)
- Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China; and Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China; and Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Dalong Ma
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China; and Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| |
Collapse
|
30
|
Hu M, Wang YM, Wang Y, Zhang GY, Zheng G, Yi S, O'Connell PJ, Harris DCH, Alexander SI. Regulatory T cells in kidney disease and transplantation. Kidney Int 2016; 90:502-14. [PMID: 27263492 DOI: 10.1016/j.kint.2016.03.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/06/2016] [Accepted: 03/17/2016] [Indexed: 01/03/2023]
Abstract
Regulatory T cells (Tregs) have been shown to be important in maintaining immune homeostasis and preventing autoimmune disease, including autoimmune kidney disease. It is also likely that they play a role in limiting kidney transplant rejection and potentially in promoting transplant tolerance. Although other subsets of Tregs exist, the most potent and well-defined Tregs are the Foxp3 expressing CD4(+) Tregs derived from the thymus or generated peripherally. These CD4(+)Foxp3(+) Tregs limit autoimmune renal disease in animal models, especially chronic kidney disease, and kidney transplantation. Furthermore, other subsets of Tregs, including CD8 Tregs, may play a role in immunosuppression in kidney disease. The development and protective mechanisms of Tregs in kidney disease and kidney transplantation involve multiple mechanisms of suppression. Here we review the development and function of CD4(+)Foxp3(+) Tregs. We discuss the specific application of Tregs as a therapeutic strategy to prevent kidney disease and to limit kidney transplant rejection and detail clinical trials in this area of transplantation.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Geoff Y Zhang
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Shounan Yi
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Philip J O'Connell
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - David C H Harris
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
31
|
Emaminejad S, Paik KH, Tabard-Cossa V, Javanmard M. Portable Cytometry Using Microscale Electronic Sensing. SENSORS AND ACTUATORS. B, CHEMICAL 2016; 224:275-281. [PMID: 27647950 PMCID: PMC5026313 DOI: 10.1016/j.snb.2015.08.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
In this manuscript, we present three different micro-impedance sensing architectures for electronic counting of cells and beads. The first method of sensing is based on using an open circuit sensing electrode integrated in a micro-pore, which measures the shift in potential as a micron-sized particle passes through. Our micro-pore, based on a funnel shaped microchannel, was fabricated in PDMS and was bound covalently to a glass substrate patterned with a gold open circuit electrode. The amplification circuitry was integrated onto a battery-powered custom printed circuit board. The second method is based on a three electrode differential measurement, which opens up the potential of using signal processing techniques to increase signal to noise ratio post measurement. The third architecture uses a contactless sensing approach, which significantly minimizes the cost of the consumable component of the impedance cytometer. We demonstrated proof of concept for the three sensing architectures by measuring the detected signal due to the passage of micron sized beads through the pore.
Collapse
Affiliation(s)
- Sam Emaminejad
- Electrical Engineering and Computer Science Department, University of California Berkeley, Berkeley, CA, USA; School of Medicine, Stanford University, Stanford, CA, USA
| | - Kee-Hyun Paik
- School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Mehdi Javanmard
- Electrical and Computer Engineering Department, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
32
|
Choi J, Selmi C, Leung PSC, Kenny TP, Roskams T, Gershwin ME. Chemokine and chemokine receptors in autoimmunity: the case of primary biliary cholangitis. Expert Rev Clin Immunol 2016; 12:661-72. [PMID: 26821815 DOI: 10.1586/1744666x.2016.1147956] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemokines represent a major mediator of innate immunity and play a key role in the selective recruitment of cells during localized inflammatory responses. Beyond critical extracellular mediators of leukocyte trafficking, chemokines and their cognate receptors are expressed by a variety of resident and infiltrating cells (monocytes, lymphocytes, NK cells, mast cells, and NKT cells). Chemokines represent ideal candidates for mechanistic studies (particularly in murine models) to better understand the pathogenesis of chronic inflammation and possibly become biomarkers of disease. Nonetheless, therapeutic approaches targeting chemokines have led to unsatisfactory results in rheumatoid arthritis, while biologics against pro-inflammatory cytokines are being used worldwide with success. In this comprehensive review we will discuss the evidence supporting the involvement of chemokines and their specific receptors in mediating the effector cell response, utilizing the autoimmune/primary biliary cholangitis setting as a paradigm.
Collapse
Affiliation(s)
- Jinjung Choi
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA.,b Division of Rheumatology , CHA University Medical Center , Bundang , Korea
| | - Carlo Selmi
- c Rheumatology and Clinical Immunology , Humanitas Research Hospital , Rozzano , Italy.,d BIOMETRA Department , University of Milan , Milano , Italy
| | - Patrick S C Leung
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA
| | - Thomas P Kenny
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA
| | - Tania Roskams
- e Translational Cell and Tissue Research , University of Leuven , Leuven , Belgium
| | - M Eric Gershwin
- a Division of Rheumatology, Allergy and Clinical Immunology , University of California Davis , Davis , CA , USA
| |
Collapse
|
33
|
Hall BM, Tran GT, Robinson CM, Hodgkinson SJ. Induction of antigen specific CD4+CD25+Foxp3+T regulatory cells from naïve natural thymic derived T regulatory cells. Int Immunopharmacol 2015; 28:875-86. [DOI: 10.1016/j.intimp.2015.03.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
|
34
|
Abstract
Chemokines are chemotactic cytokines that control the migration of cells between tissues and the positioning and interactions of cells within tissue. The chemokine superfamily consists of approximately 50 endogenous chemokine ligands and 20 G protein-coupled seven-transmembrane spanning signaling receptors. Chemokines mediate the host response to cancer by directing the trafficking of leukocytes into the tumor microenvironment. This migratory response is complex and consists of diverse leukocyte subsets with both antitumor and protumor activities. Although chemokines were initially appreciated as important mediators of immune cell migration, we now know that they also play important roles in the biology of nonimmune cells important for tumor growth and progression. Chemokines can directly modulate the growth of tumors by inducing the proliferation of cancer cells and preventing their apoptosis. They also direct tumor cell movement required for metastasis. Chemokines can also indirectly modulate tumor growth through their effects on tumor stromal cells and by inducing the release of growth and angiogenic factors from cells in the tumor microenvironment. In this Masters of Immunology primer, we focus on recent advances in understanding the complex nature of the chemokine system in tumor biology with a focus on how the chemokine system could be used to augment cancer immunotherapeutic strategies to elicit a more robust and long-lasting host antitumor immune response.
Collapse
Affiliation(s)
- Melvyn T Chow
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
35
|
Bhattacharya S, Andorf S, Gomes L, Dunn P, Schaefer H, Pontius J, Berger P, Desborough V, Smith T, Campbell J, Thomson E, Monteiro R, Guimaraes P, Walters B, Wiser J, Butte AJ. ImmPort: disseminating data to the public for the future of immunology. Immunol Res 2015; 58:234-9. [PMID: 24791905 DOI: 10.1007/s12026-014-8516-1] [Citation(s) in RCA: 540] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The immunology database and analysis portal (ImmPort) system is the archival repository and dissemination vehicle for clinical and molecular datasets created by research consortia funded by the National Institute of Allergy and Infectious Diseases Division of Allergy, Immunology, and Transplantation. With nearly 100 datasets now publicly available and hundreds of downloads per month, ImmPort is an important source for raw data and protocols from clinical trials, mechanistic studies, and novel methods for cellular and molecular measurements. To facilitate data transfer, templates for data representation and standard operating procedures have also been created and are also publicly available. ImmPort facilitates transparency and reproducibility in immunology research, serves as an important resource for education, and enables newly generated hypotheses and data-driven science.
Collapse
Affiliation(s)
- Sanchita Bhattacharya
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 1265 Welch Road MSOB X163, Stanford, CA, 94305-5415, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pharmacodynamic Monitoring of Mammalian Target of Rapamycin Inhibition by Phosphoflow Cytometric Determination of p70S6 Kinase Activity. Transplantation 2015; 99:210-9. [DOI: 10.1097/tp.0000000000000273] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
37
|
Maliniemi P, Hahtola S, Ovaska K, Jeskanen L, Väkevä L, Jäntti K, Stadler R, Michonneau D, Fraitag S, Hautaniemi S, Ranki A. Molecular characterization of subcutaneous panniculitis-like T-cell lymphoma reveals upregulation of immunosuppression- and autoimmunity-associated genes. Orphanet J Rare Dis 2014; 9:160. [PMID: 25928531 PMCID: PMC4320460 DOI: 10.1186/s13023-014-0160-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/09/2014] [Indexed: 12/21/2022] Open
Abstract
Background Subcutaneous panniculitis-like T cell lymphomas represent a rare and difficult to diagnose entity of cutaneous T cell lymphomas. SPTL affects predominantly young adults and presents with multifocal subcutaneous nodules and frequently associated autoimmune features. The pathogenesis of SPTL is not completely understood. Methods The aim of this study was to unravel molecular pathways critical to the SPTL pathogenesis. Therefore, we analyzed 23 skin samples from 20 newly diagnosed SPTL patients and relevant control samples of adipose and non-malignant panniculitis tissue by using gene expression microarray, quantitative PCR, and two-colour immunohistochemistry. Results Interestingly, indoleamine 2,3-dioxygenase (IDO-1), an immunotolerance-inducing enzyme, was among the most highly overexpressed genes in all comparisons. The expression of Th1-specific cytokines, known to be associated with autoimmune inflammation (i.e. IFNG, CXCR3, CXCL9, CXCL10, CXCL11, and CCL5), were also significantly increased. Confirmed using immunohistochemistry, the morphologically malignant lymphocytes expressed CXCR3 and CXCL9. IDO-1 expression was found both in some morphologically malignant lymphocytes rimming the adipocytes and in surrounding CD11c− CD68− cells but not in CD11c+ dendritic cells in the microenvironment. The proportion of FoxP3+ cells in SPTL exceeded that in the benign panniculitis samples. Conclusions Our results indicate that the up regulation of the tolerogenic IDO-1 together with the up regulation of IFNG, CXCR3 ligands, and CCL5 are features of SPTL lesions. We anticipate that the IFNG-inducible IDO-1 expression contributes to the formation of an immunosuppressive microenvironment, favorable for the malignant T cells. This study provides a relevant molecular basis for further studies exploring novel therapeutic means for subcutaneous T cell lymphoma. Electronic supplementary material The online version of this article (doi:10.1186/s13023-014-0160-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pilvi Maliniemi
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Sonja Hahtola
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Kristian Ovaska
- Systems Biology Laboratory, Institute of Biomedicine and Genome-Scale Biology Program, University of Helsinki, Helsinki, Finland.
| | - Leila Jeskanen
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Liisa Väkevä
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Kirsi Jäntti
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Rudolf Stadler
- Johannes-Wesling-Klinikum Minden, Akademisches Lehrkrankenhaus der Medizinischen Hochschule Hannover, Minden, Germany.
| | - David Michonneau
- Institut Pasteur, Département d'immunologie, Equipe Dynamique des réponses immunes, 25 rue du Docteur Roux, 75015, Paris, France.
| | - Sylvie Fraitag
- Service d'anatomie et de cytologie pathologiques, Hôpital Necker-Enfants-Malades, AP-HP, 149, rue de Sèvres, 75743, Paris Cedex 15, France.
| | - Sampsa Hautaniemi
- Systems Biology Laboratory, Institute of Biomedicine and Genome-Scale Biology Program, University of Helsinki, Helsinki, Finland.
| | - Annamari Ranki
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| |
Collapse
|
38
|
Wang J, Vodovotz Y, Fan L, Li Y, Liu Z, Namas R, Barclay D, Zamora R, Billiar TR, Wilson MA, Fan J, Jiang Y. Injury-induced MRP8/MRP14 stimulates IP-10/CXCL10 in monocytes/macrophages. FASEB J 2014; 29:250-62. [PMID: 25342131 DOI: 10.1096/fj.14-255992] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Trauma/hemorrhagic shock is associated with morbidity and mortality due to dysregulated inflammation, which is driven in part by monocytes/macrophages stimulated by injury-induced release of damage-associated molecular pattern (DAMP) molecules. MRP8/MRP14 is an endogenous DAMP involved in various inflammatory diseases, though its mechanism of action is unclear. Circulating MRP8/MRP14 levels in human blunt trauma nonsurvivors were significantly lower than those of survivors (P < 0.001). Human monocytic THP-1 cells stimulated with MRP8/MRP14 expressed the chemokine IFN-γ inducible protein 10 (IP-10)/CXCL10. Circulating IP-10 levels in human blunt trauma patients were correlated positively with MRP8/MRP14 levels (r = 0.396, P < 0.001), and were significantly lower in trauma nonsurvivors than in survivors (P < 0.001). We therefore sought to determine the mechanisms by which MRP8/MRP14 stimulates IP-10 in monocytes/macrophages, and found that induction of IP-10 by MRP8/MRP14 required Toll-like receptor 4 and TRIF but not MyD88. Full induction of IP-10 by MRP8/MRP14 required synergy between the transcription factors NF-κB and IFN regulatory factor 3 (IRF3). The receptor for IP-10 is CXCR3, and MRP8/MRP14-induced chemotaxis of CXCR3(+) cells was dependent on the production of IP-10 in monocytes/macrophages. Furthermore, in vivo study with a mouse trauma/hemorrhagic shock model showed that administration of neutralizing antibody against MRP8 prevented activation of NF-κB and IRF3 as well as IP-10 production. Thus, the current study identified a novel signaling mechanism that controls IP-10 expression in monocytes/macrophages by MRP8/MRP14, which may play an important role in injury-induced inflammation.
Collapse
Affiliation(s)
- Juan Wang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Transcriptomics and Proteomics, Ministry of Education of China, Key Laboratory of Proteomics of Guangdong Province, Southern Medical University, Guangzhou, China; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Liyan Fan
- University of Pittsburgh School of Arts and Science, Pittsburgh, Pennsylvania, USA; and
| | - Yuehua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zheng Liu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Transcriptomics and Proteomics, Ministry of Education of China, Key Laboratory of Proteomics of Guangdong Province, Southern Medical University, Guangzhou, China
| | - Rami Namas
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark A Wilson
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Jie Fan
- State Key Laboratory of Organ Failure Research, Key Laboratory of Transcriptomics and Proteomics, Ministry of Education of China, Key Laboratory of Proteomics of Guangdong Province, Southern Medical University, Guangzhou, China; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yong Jiang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Transcriptomics and Proteomics, Ministry of Education of China, Key Laboratory of Proteomics of Guangdong Province, Southern Medical University, Guangzhou, China;
| |
Collapse
|
39
|
Junkin M, Tay S. Microfluidic single-cell analysis for systems immunology. LAB ON A CHIP 2014; 14:1246-60. [PMID: 24503696 DOI: 10.1039/c3lc51182k] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The immune system constantly battles infection and tissue damage, but exaggerated immune responses lead to allergies, autoimmunity and cancer. Discrimination of self from foreign and the fine-tuning of immunity are achieved by information processing pathways, whose regulatory mechanisms are little understood. Cell-to-cell variability and stochastic molecular interactions result in diverse cellular responses to identical signaling inputs, casting doubt on the reliability of traditional population-averaged analyses. Furthermore, dynamic molecular and cellular interactions create emergent properties that change over multiple time scales. Understanding immunity in the face of complexity and noisy dynamics requires time-dependent analysis of single-cells in a proper context. Microfluidic systems create precisely defined microenvironments by controlling fluidic and surface chemistries, feature sizes, geometries and signal input timing, and thus enable quantitative multi-parameter analysis of single cells. Such qualities allow observable dynamic environments approaching in vivo levels of biological complexity. Seamless parallelization of functional units in microfluidic devices allows high-throughput measurements, an essential feature for statistically meaningful analysis of naturally variable biological systems. These abilities recapitulate diverse scenarios such as cell-cell signaling, migration, differentiation, antibody and cytokine production, clonal selection, and cell lysis, thereby enabling accurate and meaningful study of immune behaviors in vitro.
Collapse
Affiliation(s)
- Michael Junkin
- Department of Biosystems Science and Engineering, ETH Zürich, Switzerland.
| | | |
Collapse
|
40
|
Kim SM, Kim BY, Lee SA, Eo SK, Yun Y, Kim CD, Kim K. 27-Hydroxycholesterol and 7alpha-hydroxycholesterol trigger a sequence of events leading to migration of CCR5-expressing Th1 lymphocytes. Toxicol Appl Pharmacol 2014; 274:462-70. [DOI: 10.1016/j.taap.2013.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/10/2013] [Accepted: 12/11/2013] [Indexed: 12/13/2022]
|
41
|
Yoshikawa M, Wada K, Yoshimura T, Asaka D, Okada N, Matsumoto K, Moriyama H. Increased CXCL10 expression in nasal fibroblasts from patients with refractory chronic rhinosinusitis and asthma. Allergol Int 2013; 62:495-502. [PMID: 24153332 DOI: 10.2332/allergolint.13-oa-0572] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 07/03/2013] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is characterized by local inflammation of the sinonasal tissues. CRS patients with nasal polyps and asthma often develop acute exacerbation of sinonasal symptoms after upper respiratory tract infections. However, the influence of concomitant asthma on the nasal immune response to viral infection remains unclear. METHODS Specimens of nasal polyp and mucosal tissues were obtained from 3 groups of CRS patients (n = 14 per group): 1) patients without asthma (CRS group), 2) patients with aspirin-tolerant asthma (ATA group), and 3) patients with aspirin-intolerant asthma (AIA group). Nasal fibroblasts isolated from the specimens were stimulated with poly I:C. CXCL10 expression was analyzed by the quantitative real-time polymerase chain reaction and enzyme-linked immunoadsorbent assay. Biopsy specimens from CRS patients without asthma were subjected to immunohistochemistry for detection of T-bet and GATA-3 expression in CD3+ T cells by double labeling. RESULTS Nasal fibroblasts from the ATA and AIA groups showed significantly enhanced expression of CXCL10 mRNA and protein after poly I:C stimulation compared with cells from the CRS group and the control group (normal nasal mucosa). In addition to T helper (Th)2 cells, there was more abundant infiltration of Th1 cells into tissues from the AIA and ATA groups. CONCLUSIONS Our findings suggest that CRS associated with asthma may become intractable through the over-production of CXCL10 in response to viral infection.
Collapse
Affiliation(s)
- Mamoru Yoshikawa
- Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Organ transplantation appears today to be the best alternative to replace the loss of vital organs induced by various diseases. Transplants can, however, also be rejected by the recipient. In this review, we provide an overview of the mechanisms and the cells/molecules involved in acute and chronic rejections. T cells and B cells mainly control the antigen-specific rejection and act either as effector, regulatory, or memory cells. On the other hand, nonspecific cells such as endothelial cells, NK cells, macrophages, or polymorphonuclear cells are also crucial actors of transplant rejection. Last, beyond cells, the high contribution of antibodies, chemokines, and complement molecules in graft rejection is discussed in this article. The understanding of the different components involved in graft rejection is essential as some of them are used in the clinic as biomarkers to detect and quantify the level of rejection.
Collapse
Affiliation(s)
- Aurélie Moreau
- INSERM UMR 1064, Center for Research in Transplantation and Immunology-ITUN, CHU de Nantes 44093, France
| | | | | | | |
Collapse
|
43
|
Hu M, Wang C, Zhang GY, Saito M, Wang YM, Fernandez MA, Wang Y, Wu H, Hawthorne WJ, Jones C, O'Connell PJ, Sparwasser T, Bishop GA, Sharland AF, Alexander SI. Infiltrating Foxp3(+) regulatory T cells from spontaneously tolerant kidney allografts demonstrate donor-specific tolerance. Am J Transplant 2013; 13:2819-30. [PMID: 24102948 DOI: 10.1111/ajt.12445] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/16/2013] [Accepted: 07/27/2013] [Indexed: 01/25/2023]
Abstract
Foxp3(+) regulatory T cells (Tregs) have an essential role in immune and allograft tolerance. However, in both kidney and liver transplantation in humans, FOXP3(+) Tregs have been associated with clinical rejection. Therefore, the role and function of graft infiltrating Tregs have been of great interest. In the studies outlined, we demonstrated that Foxp3(+) Tregs were expanded in tolerant kidney allografts and in draining lymph nodes in the DBA/2 (H-2(d) ) to C57BL/6 (H-2(b) ) mouse spontaneous kidney allograft tolerance model. Kidney allograft tolerance was abrogated after deletion of Foxp3(+) Tregs in DEpletion of REGulatory T cells (DEREG) mice. Kidney allograft infiltrating Foxp3(+) Tregs (K-Tregs) expressed elevated levels of TGF-β, IL-10, interferon gamma (IFN-γ), the transcriptional repressor B lymphocyte-induced maturation protein-1 (Blimp-1) and chemokine receptor 3 (Cxcr3). These K-Tregs had the capacity to transfer dominant tolerance and demonstrate donor alloantigen-specific tolerance to skin allografts. This study demonstrated the crucial role, potency and specificity of graft infiltrating Foxp3(+) Tregs in the maintenance of spontaneously induced kidney allograft tolerance.
Collapse
Affiliation(s)
- M Hu
- Centre for Kidney Research, Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia; Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney, Westmead, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hoang AN, Jones CN, Dimisko L, Hamza B, Martel J, Kojic N, Irimia D. Measuring neutrophil speed and directionality during chemotaxis, directly from a droplet of whole blood. TECHNOLOGY 2013; 1:49. [PMID: 24809064 PMCID: PMC4010229 DOI: 10.1142/s2339547813500040] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Neutrophil chemotaxis is critical for defense against infections and its alterations could lead to chronic inflammation and tissue injury. The central role that transient alterations of neutrophil chemotaxis could have on patient outcomes calls for its quantification in the clinic. However, current methods for measuring neutrophil chemotaxis require large volumes of blood and are time consuming. To address the need for rapid and robust assays, we designed a microfluidic device that measures neutrophil chemotaxis directly from a single droplet of blood. We validated the assay by comparing neutrophil chemotaxis from finger prick, venous blood and purified neutrophil samples. We found consistent average velocity of (19 ± 6 μm/min) and directionality (91.1%) between the three sources. We quantified the variability in neutrophil chemotaxis between healthy donors and found no significant changes over time. We also validated the device in the clinic and documented temporary chemotaxis deficiencies after burn injuries.
Collapse
Affiliation(s)
- Anh N. Hoang
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, USA
- Correspondence should be addressed to D.I. ()
| | - Caroline N. Jones
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, USA
- Correspondence should be addressed to D.I. ()
| | - Laurie Dimisko
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, USA
| | - Bashar Hamza
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, USA
| | - Joseph Martel
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard University School of Engineering and Applied Sciences, Boston, USA
| | - Nikola Kojic
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, USA
| | - Daniel Irimia
- The BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, USA
| |
Collapse
|
45
|
Schmitz K, Pickert G, Wijnvoord N, Häussler A, Tegeder I. Dichotomy of CCL21 and CXCR3 in nerve injury-evoked and autoimmunity-evoked hyperalgesia. Brain Behav Immun 2013; 32:186-200. [PMID: 23643685 DOI: 10.1016/j.bbi.2013.04.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 04/20/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023] Open
Abstract
The chemokine CCL21 is released from injured neurons and acts as a ligand of the chemokine receptor, CXCR3, which likely contributes to pro-inflammatory adaptations and secondary neuronal damage. CCL21-CXCR3 signalling may therefore impact on the development of neuropathic pain. By using the respective knockout mice we show that deficiency of CCL19/21 in plt/plt mice attenuates nerve injury evoked pain but not the hyperalgesia evoked by autoimmune encephalomyelitis (EAE). Oppositely, CXCR3-deficiency had no protective effect after traumatic nerve injury but reduced EAE-evoked hyperalgesia and was associated with reduced clinical EAE scores, a reduction of the pro-inflammatory cell infiltration and reduced upregulation of interferon gamma and interleukin-17 in the spinal cord. In contrast, microglia activation in the spinal cord after traumatic sciatic nerve injury was neither attenuated in CXCR3(-/-) nor plt/plt mice, nor in double knockouts. However, the severity of EAE, but not the hyperalgesia, was also reduced in plt/plt mice, which was associated with reduced infiltration of the spinal cord with CCR7+ T-cells, an increase of CD25+ T-cells and reduced upregulation of CXCL9 and 10, CCL11 and 12. The data show that CCL21 and CXCR3 have dichotomous functions in traumatic and EAE-evoked neuropathic pain suggesting diverse mechanisms likely requiring diverse treatments although both types of neuropathic pain are mediated in part through the immune activation.
Collapse
Affiliation(s)
- Katja Schmitz
- Pharmazentrum Frankfurt, Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | | | | | | | | |
Collapse
|
46
|
Wu J, Wu X, Lin F. Recent developments in microfluidics-based chemotaxis studies. LAB ON A CHIP 2013; 13:2484-99. [PMID: 23712326 DOI: 10.1039/c3lc50415h] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Microfluidic devices can better control cellular microenvironments compared to conventional cell migration assays. Over the past few years, microfluidics-based chemotaxis studies showed a rapid growth. New strategies were developed to explore cell migration in manipulated chemical gradients. In addition to expanding the use of microfluidic devices for a broader range of cell types, microfluidic devices were used to study cell migration and chemotaxis in complex environments. Furthermore, high-throughput microfluidic chemotaxis devices and integrated microfluidic chemotaxis systems were developed for medical and commercial applications. In this article, we review recent developments in microfluidics-based chemotaxis studies and discuss the new trends in this field observed over the past few years.
Collapse
Affiliation(s)
- Jiandong Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | | | |
Collapse
|
47
|
Suga H, Sugaya M, Miyagaki T, Ohmatsu H, Okochi H, Sato S. CXCR3 deficiency prolongs Th1-type contact hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2013; 190:6059-70. [PMID: 23656737 DOI: 10.4049/jimmunol.1201606] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Sensitization and challenge using dinitrofluorobenzene (DNFB) induce contact hypersensitivity (CHS) with Th1 cell infiltration, whereas those using FITC generate CHS with Th2 cell infiltration. In this study, we attempted to determine the role of CXCR3, a chemokine receptor, in Th1- and Th2-type CHS induced by DNFB or FITC using CXCR3-deficient (CXCR3(-/-)) mice. Ear swelling was prolonged after DNFB challenge in CXCR3(-/-) mice, which was accompanied by increased Th1 cytokines and decreased TGF-β and IL-10 expression at a late time point of CHS, whereas there was no significant difference between wild-type and CXCR3(-/-) mice in FITC-induced CHS. In Th1-type CHS, the number of regulatory T cells (Tregs) was decreased in the challenged ear of CXCR3(-/-) mice compared with that of wild-type mice, suggesting that CXCR3 would be important in migration of Tregs into the site of inflammation. Moreover, we examined the characteristics of CXCR3(+) Tregs both in vitro and in vivo, revealing that CXCR3(+) Tregs expressed high levels of TGF-β and IL-10 as well as IFN-γ compared with CXCR3(-) Tregs. When CXCR3(-/-) mice were injected with CXCR3(+) Tregs, the prolonged ear swelling induced by DNFB was normalized. Taken together, our results suggest that CXCR3(+) Tregs play a key role for quenching Th1-type CHS.
Collapse
Affiliation(s)
- Hiraku Suga
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Afshar R, Strassner JP, Seung E, Causton B, Cho JL, Harris RS, Hamilos DL, Medoff BD, Luster AD. Compartmentalized chemokine-dependent regulatory T-cell inhibition of allergic pulmonary inflammation. J Allergy Clin Immunol 2013; 131:1644-52. [PMID: 23632297 DOI: 10.1016/j.jaci.2013.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 02/08/2013] [Accepted: 03/04/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND Induction of endogenous regulatory T (Treg) cells represents an exciting new potential modality for treating allergic diseases, such as asthma. Treg cells have been implicated in the regulation of asthma, but the anatomic location in which they exert their regulatory function and the mechanisms controlling the migration necessary for their suppressive function in asthma are not known. Understanding these aspects of Treg cell biology will be important for harnessing their power in the clinic. OBJECTIVE We sought to determine the anatomic location at which Treg cells exert their regulatory function in the sensitization and effector phases of allergic asthma and to determine the chemokine receptors that control the migration of Treg cells to these sites in vivo in both mice and human subjects. METHODS The clinical efficacy and anatomic location of adoptively transferred chemokine receptor-deficient CD4(+)CD25(+) forkhead box protein 3-positive Treg cells was determined in the sensitization and effector phases of allergic airway inflammation in mice. The chemokine receptor expression profile was determined on Treg cells recruited into the human airway after bronchoscopic segmental allergen challenge of asthmatic patients. RESULTS We show that CCR7, but not CCR4, is required on Treg cells to suppress allergic airway inflammation during the sensitization phase. In contrast, CCR4, but not CCR7, is required on Treg cells to suppress allergic airway inflammation during the effector phase. Consistent with our murine studies, human subjects with allergic asthma had an increase in CCR4-expressing functional Treg cells in the lungs after segmental allergen challenge. CONCLUSION The location of Treg cell function differs during allergic sensitization and allergen-induced recall responses in the lung, and this differential localization is critically dependent on differential chemokine function.
Collapse
Affiliation(s)
- Roshi Afshar
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
T regulatory cell therapy in transplantation: stability, localization and functional specialization. Curr Opin Organ Transplant 2013; 17:343-8. [PMID: 22790068 DOI: 10.1097/mot.0b013e328355aaaf] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW There is great hope that cellular therapy with regulatory T cells (Tregs) will be an effective way to induce alloantigen specific tolerance, ultimately allowing for reduction or elimination of nonspecific immunosuppression. In the past, considerable effort was focused on defining the optimal ways to isolate and expand Tregs from peripheral or cord blood. Now that expansion of therapeutically relevant numbers of Tregs is feasible, we need to consider what is going to happen to the cells when they are transferred in vivo. RECENT FINDINGS For optimal function, Tregs must be able to traffic to the correct location(s) and, despite the presence of immunosuppressive therapy, live long enough to transfer their regulatory function to recipient T cells. Within the Treg pool, there are also functionally specialized subsets, identified by chemokine receptor expression and/or cytokine production, which control their trafficking and relative ability to suppress different types of T helper cells, respectively. Recent findings imply that the plasticity of appropriately obtained populations of Tregs may not be of as great concern as previously suggested. Experimental data have also provided evidence as to how one might design adjunctive treatment that best supports the viability and function of Tregs after transfer. SUMMARY Knowledge of how Tregs work in transplantation comes from studies that do not recapitulate how these cells will be used in humans. There is a need to develop better preclinical models to study how the in-vivo function of human Tregs can be optimized to ensure they can meet the challenge of inducing transplantation tolerance.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Immunological barriers still preclude clinical xenotransplantation. The protective role of CD4(+)CD25(+)Foxp3(+) T-regulatory cells (Treg) in allotransplantation is well described and, therefore, could represent a promising therapeutical tool for xenotransplantation. This review addresses the latest findings on Treg in xenotransplantation research. RECENT FINDINGS In vivo, costimulation blockade-based strategies including anti-CD154 monoclonal antibodies (mAbs) in combination with rapamycin or anti-LFA-1 mAb prolonged both concordant and discordant islets xenografts survival in a Treg-dependent manner. In vitro, IL-10 secretion was shown to be critical for the suppression of xenogeneic responses mediated by Treg. Moreover, transgenic expression of inducible costimulator-immunoglobulin or PD-L1 on porcine endothelial cells inhibited human T-cell proliferation in vitro and was associated with the induction of Treg and IL-10 secretion. CXCR3 mediated the recruitment of Treg to pig endothelium. Finally, the recruitment of human Treg was enhanced by the immobilization of human CCL17 on pig endothelium. SUMMARY There is increasing evidence for the potential of CD4(+)CD25(+)Foxp3(+) Treg to protect xenografts. Induction of Treg in recipients and/or recruitment of human Treg to pig endothelium may represent novel strategies to prevent cell-mediated rejection in pig-to-human xenotransplantation.
Collapse
|