1
|
Guerrero-Murillo M, Rill-Hinarejos A, Trincado JL, Bataller A, Ortiz-Maldonado V, Benítez-Ribas D, Español-Rego M, González-Navarro EA, Martínez-Cibrián N, Marchese D, Martín-Martín L, Martín García-Sancho A, Rives S, Heyn H, Juan M, Urbano-Ispizúa Á, Delgado J, Orfao A, Mereu E, Bueno C, Menendez P. Integrative single-cell multi-omics of CD19-CAR pos and CAR neg T cells suggest drivers of immunotherapy response in B cell neoplasias. Cell Rep Med 2024:101803. [PMID: 39471818 DOI: 10.1016/j.xcrm.2024.101803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/31/2024] [Accepted: 10/03/2024] [Indexed: 11/01/2024]
Abstract
The impact of phenotypic, clonal, and functional heterogeneity of chimeric antigen receptor (CAR)-T cells on clinical outcome remains understudied. Here, we integrate clonal kinetics with transcriptomic heterogeneity resolved by single-cell omics to interrogate cellular dynamics of non-transduced (CARneg) and transduced (CARpos) T cells, in the infusion product (IP) and at the CAR-T cell expansion peak in five B cell acute lymphoblastic leukemia (B-ALL) patients treated with CD19CAR-T cells (varni-cel). We identify significant differences in cellular dynamics in response to therapy. CARpos T cells at IP of complete response patients exhibit a significantly higher CD4:CD8 ratio, validated in a larger cohort B-ALL patients (n = 47). Conversely, at the expansion peak, there is a clonal expansion of CD8+ effector memory and cytotoxic T cells. Cytotoxic CARpos γδ-T cells expansion correlates with treatment efficacy validated in a cohort of B-ALL (n = 18) and diffuse large B cell lymphoma (DLBCL) patients (n = 58). Our data provide insights into the complexity of T cell responses following CAR-T cell therapy and suggest drivers of immunotherapy response.
Collapse
Affiliation(s)
- Mercedes Guerrero-Murillo
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; PhD programme in Biomedicine, University of Barcelona, Barcelona, Spain
| | - Aina Rill-Hinarejos
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; PhD programme in Biomedicine, University of Barcelona, Barcelona, Spain
| | - Juan L Trincado
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Alex Bataller
- Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Valentín Ortiz-Maldonado
- Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Daniel Benítez-Ribas
- Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Español-Rego
- Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Azucena González-Navarro
- Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Doménica Marchese
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Lourdes Martín-Martín
- Cancer Research Centre (IBMCC, USAL-CSIC), Institute for Biomedical Research of Salamanca (IBSAL) and Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alejandro Martín García-Sancho
- Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), CIBERONC, University of Salamanca and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Susana Rives
- Pediatric Oncology and Hematology Department, Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Manel Juan
- Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Immunology and immunotherapy, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Álvaro Urbano-Ispizúa
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julio Delgado
- Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain
| | - Alberto Orfao
- Cancer Research Centre (IBMCC, USAL-CSIC), Institute for Biomedical Research of Salamanca (IBSAL) and Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain
| | | | - Clara Bueno
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain.
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain; Spanish Network for Advanced Therapies, RICORS-TERAV, ISCIII, Spain; Spanish Collaborative Cancer Network, CIBERONC, ISCIII, Spain; Department of Biomedicine, University of Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
2
|
Sacharczuk M, Mickael ME, Kubick N, Kamińska A, Horbańczuk JO, Atanasov AG, Religa P, Ławiński M. The Current Landscape of Hypotheses Describing the Contribution of CD4+ Heterogeneous Populations to ALS. Curr Issues Mol Biol 2024; 46:7846-7861. [PMID: 39194682 DOI: 10.3390/cimb46080465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a poorly understood and fatal disease. It has a low prevalence and a 2-4 year survival period. Various theories and hypotheses relating to its development process have been proposed, albeit with no breakthrough in its treatment. Recently, the role of the adaptive immune system in ALS, particularly CD4+ T cells, has begun to be investigated. CD4+ T cells are a heterogeneous group of immune cells. They include highly pro-inflammatory types such as Th1 and Th17, as well as highly anti-inflammatory cells such as Tregs. However, the landscape of the role of CD4+ T cells in ALS is still not clearly understood. This review covers current hypotheses that elucidate how various CD4+ T cells can contribute to ALS development. These hypotheses include the SWITCH model, which suggests that, in the early stages of the disease, Tregs are highly capable of regulating the immune response. However, in the later stages of the disease, it seems that pro-inflammatory cells such as Th1 and Th17 are capable of overwhelming Treg function. The reason why this occurs is not known. Several research groups have proposed that CD4+ T cells as a whole might experience aging. Others have proposed that gamma delta T cells might directly target Tregs. Additionally, other research groups have argued that less well-known CD4+ T cells, such as Emoes+ CD4+ T cells, may be directly responsible for neuron death by producing granzyme B. We propose that the ALS landscape is highly complicated and that there is more than one feasible hypothesis. However, it is critical to take into consideration the differences in the ability of different populations of CD4+ T cells to infiltrate the blood-brain barrier, taking into account the brain region and the time of infiltration. Shedding more light on these still obscure factors can help to create a personalized therapy capable of regaining the balance of power in the battle between the anti-inflammatory and pro-inflammatory cells in the central nervous system of ALS patients.
Collapse
Affiliation(s)
- Mariusz Sacharczuk
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-091 Warsaw, Poland
| | - Michel-Edwar Mickael
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
| | - Norwin Kubick
- Department of Biology, Institute of Plant Science and Microbiology, University of Hamburg, Ohnhorststr. 18, 22609 Hamburg, Germany
| | - Agnieszka Kamińska
- Faculty of Medicine, Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw, 01-938 Warsaw, Poland
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
| | - Atanas G Atanasov
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, 1090 Vienna, Austria
| | - Piotr Religa
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden
| | - Michał Ławiński
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland
- Department of General Surgery, Gastroenterology and Oncology, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
3
|
Chan S, Morgan B, Yong MK, Margetts M, Farchione AJ, Lucas EC, Godsell J, Giang NA, Slade CA, von Borstel A, Bryant VL, Howson LJ. Cytomegalovirus drives Vδ1 + γδ T cell expansion and clonality in common variable immunodeficiency. Nat Commun 2024; 15:4286. [PMID: 38769332 PMCID: PMC11106253 DOI: 10.1038/s41467-024-48527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/02/2024] [Indexed: 05/22/2024] Open
Abstract
The function and phenotype of γδ T cells in the context of common variable immunodeficiency (CVID) has not been explored. CVID is a primary immunodeficiency disorder characterized by impaired antibody responses resulting in increased susceptibility to infections. γδ T cells are a subset of unconventional T cells that play crucial roles in host defence against infections. In this study, we aim to determine the roles and functions of γδ T cells in CVID. We observe a higher frequency of Vδ1+ γδ T cells compared to healthy controls, particularly in older patients. We also find a higher proportion of effector-memory Vδ1+ γδ T cells and a more clonal T cell receptor (TCR) repertoire in CVID. The most significant driver of the Vδ1+ γδ T cell expansion and phenotype in CVID patients is persistent cytomegalovirus (CMV) viremia. These findings provide valuable insights into γδ T cell biology and their contribution to immune defence in CVID.
Collapse
Affiliation(s)
- Samantha Chan
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin Morgan
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Michelle K Yong
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mai Margetts
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Anthony J Farchione
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Erin C Lucas
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Jack Godsell
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Infectious Diseases, Austin Hospital, Heidelberg, VIC, Australia
| | - Nhi Ai Giang
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Charlotte A Slade
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Anouk von Borstel
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Vanessa L Bryant
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Lauren J Howson
- Immunology Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
4
|
Martín-Martín L, Gutiérrez-Herrero S, Herrero-García M, Martín García-Sancho A, Yeguas A, Martín-López AÁ, López-Corral L, Pérez-López E, García-Blázquez M, Sánchez-Guijo F, Vidriales MB, Gaipa G, Orfao A. Impact of the kinetics of circulating anti-CD19 CAR-T cells and their populations on the outcome of DLBCL patients. Blood Cancer J 2024; 14:83. [PMID: 38760376 PMCID: PMC11101460 DOI: 10.1038/s41408-024-01065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024] Open
Affiliation(s)
- Lourdes Martín-Martín
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), and University of Salamanca, Salamanca, Spain
- Flow Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Sara Gutiérrez-Herrero
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), and University of Salamanca, Salamanca, Spain
- Flow Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - María Herrero-García
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), and University of Salamanca, Salamanca, Spain
- Flow Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Alejandro Martín García-Sancho
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Ana Yeguas
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Ana-África Martín-López
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Lucía López-Corral
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Estefanía Pérez-López
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | | | - Fermín Sánchez-Guijo
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - María Belén Vidriales
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Giuseppe Gaipa
- Tettamanti Center and Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), and University of Salamanca, Salamanca, Spain.
- Flow Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain.
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
5
|
Peng Y, Zhang X, Tang Y, He S, Rao G, Chen Q, Xue Y, Jin H, Liu S, Zhou Z, Xiang Y. Role of autoreactive Tc17 cells in the pathogenesis of experimental autoimmune encephalomyelitis. NEUROPROTECTION 2024; 2:49-59. [DOI: 10.1002/nep3.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/16/2024] [Indexed: 07/04/2024]
Abstract
AbstractBackgroundThe pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE—an animal model of MS) is primarily mediated by T cells. However, recent studies have only focused on interleukin (IL)‐17‐secreting CD4+ T‐helper cells, also known as Th17 cells. This study aimed to compare Th17 cells and IL‐17‐secreting CD8+ T‐cytotoxic cells (Tc17) in the context of MS/EAE.MethodsFemale C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein peptides 35–55 (MOG35–55), pertussis toxin, and complete Freund's adjuvant to establish the EAE animal model. T cells were isolated from the spleen (12–14 days postimmunization). CD4+ and CD8+ T cells were purified using isolation kit and then differentiated into Th17 and Tc17, respectively, using MOG35–55 and IL‐23. The secretion levels of interferon‐γ (IFN‐γ) and IL‐17 were measured via enzyme‐linked immunosorbent assay using cultured CD4+ and CD8+ T cell supernatants. The pathogenicity of Tc17 and Th17 cells was assessed through adoptive transfer (tEAE), with the clinical course assessed using an EAE score (0–5). Hematoxylin and eosin as well as Luxol fast blue staining were used to examine the spinal cord. Purified CD8+ CD3+ and CD4+ CD3+ cells differentiated into Tc17 and Th17 cells, respectively, were stimulated with MOG35–55 peptide for proliferation assays.ResultsThe results showed that Tc17 cells (15,951 ± 1985 vs. 55,709 ± 4196 cpm; p < 0.050) exhibited a weaker response to highest dose (20 μg/mL) MOG35–55 than Th17 cells. However, this response was not dependent on Th17 cells. After the 48 h stimulation, at the highest dose (20 μg/mL) of MOG35–55. Tc17 cells secreted lower levels of IFN‐γ (280.00 ± 15.00 vs. 556.67 ± 15.28 pg/mL, p < 0.050) and IL‐17 (102.67 ± 5.86 pg/mL vs. 288.33 ± 12.58 pg/mL; p < 0.050) than Th17 cells. Similar patterns were observed for IFN‐γ secretion at 96 and 144 h. Furthermore, Tc17 cell‐induced tEAE mice exhibited similar EAE scores to Th17 cell‐induced tEAE mice and also showed similar inflammation and demyelination.ConclusionThe degree of pathogenicity of Tc17 cells in EAE is lower than that of Th17 cells. Future investigation on different immune cells and EAE models is warranted to determine the mechanisms underlying MS.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Xiuli Zhang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yandan Tang
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shunqing He
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Guilan Rao
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Quan Chen
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Yahui Xue
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Hong Jin
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shu Liu
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Ziyang Zhou
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yun Xiang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| |
Collapse
|
6
|
Verkerk T, Pappot AT, Jorritsma T, King LA, Duurland MC, Spaapen RM, van Ham SM. Isolation and expansion of pure and functional γδ T cells. Front Immunol 2024; 15:1336870. [PMID: 38426099 PMCID: PMC10902048 DOI: 10.3389/fimmu.2024.1336870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
γδ T cells are important components of the immune system due to their ability to elicit a fast and strong response against infected and transformed cells. Because they can specifically and effectively kill target cells in an MHC independent fashion, there is great interest to utilize these cells in anti-tumor therapies where antigen presentation may be hampered. Since only a small fraction of T cells in the blood or tumor tissue are γδ T cells, they require extensive expansion to allow for fundamental, preclinical and ex vivo research. Although expansion protocols can be successful, most are based on depletion of other cell types rather than γδ T cell specific isolation, resulting in unpredictable purity of the isolated fraction. Moreover, the primary focus only lies with expansion of Vδ2+ T cells, while Vδ1+ T cells likewise have anti-tumor potential. Here, we investigated whether γδ T cells directly isolated from blood could be efficiently expanded while maintaining function. γδ T cell subsets were isolated using MACS separation, followed by FACS sorting, yielding >99% pure γδ T cells. Isolated Vδ1+ and Vδ2+ T cells could effectively expand immediately after isolation or upon freeze/thawing and reached expansion ratios between 200 to 2000-fold starting from varying numbers using cytokine supported feeder stimulations. MACS/FACS isolated and PHA stimulated γδ T cells expanded as good as immobilized antibody mediated stimulated cells in PBMCs, but delivered purer cells. After expansion, potential effector functions of γδ T cells were demonstrated by IFN-γ, TNF-α and granzyme B production upon PMA/ionomycin stimulation and effective killing capacity of multiple tumor cell lines was confirmed in killing assays. In conclusion, pure γδ T cells can productively be expanded while maintaining their anti-tumor effector functions against tumor cells. Moreover, γδ T cells could be expanded from low starting numbers suggesting that this protocol may even allow for expansion of cells extracted from tumor biopsies.
Collapse
Affiliation(s)
- Tamara Verkerk
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Anouk T Pappot
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Tineke Jorritsma
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Lisa A King
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Mariël C Duurland
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Kapembwa MS, Batman PA, Fleming SC, Griffin GE. HIV enteropathy and 'Slim disease': Historical and current perspectives. Int J Infect Dis 2024; 139:86-91. [PMID: 38052315 DOI: 10.1016/j.ijid.2023.11.037] [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: 08/19/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/07/2023] Open
Abstract
OBJECTIVES Chronic diarrhoea and severe wasting associated with HIV infection were first described in East African patients as slim disease (SD) in 1985. The main histological features are flattening of the villi (villous atrophy) and crypt hyperplasia (elongated crypts), i.e., HIV enteropathy (HIVE). Selective loss of mucosal clusters of differentiation 4 (CD4)+ T helper (Th)17+ lymphocytes is the immunological hallmark of HIVE. This review explores (i) the historical background of HIVE and SD, (ii) the relationship between gut mucosal CD4+ Th17+ and intestinal-resident intra-epithelial gamma delta (IRIE) T lymphocytes in pathogenesis of HIVE, (iii) the role of cytokines in regulation of intestinal epithelial proliferation, and (iv) the role of antiretroviral therapy in HIVE. METHODS Recent studies have highlighted the role of IRIE T lymphocytes, mostly CD8+, in regulating gut epithelial regeneration. CD4+Th17+ and IRIE T cells are necessary to maintain intestinal barrier integrity and mucosal antimicrobial immune defence. However, the immunological cross-talk between such lymphocyte sub-sets culminating in HIVE is uncertain. We undertook a narrative literature review under the headings 'HIVE', 'SD', and 'Highly active antiretroviral therapy (HAART). Relevant studies were located using the electronic search engines Google Scholar and PubMed from 1984 to 2022. RESULTS Depletion of Th17+ cells in the lamina propria, attributed to low-level viraemia, is accompanied by concomitant increase in the density of gut mucosal IRIE T lymphocytes in AIDS. The latter express a broad range of cytokines (interferon-gamma, tumor necrosis factor-alpha, interleukin-17) and chemokines e.g., keratinocyte growth factor, post exposure to HIV-infected cells. Keratinocyte growth factor induces epithelial proliferation mainly in the crypts, leading to functional immaturity of enterocytes, reduced gut absorptive surface area and malabsorption in animal experiments. Of note, the absence of IRIE T cells is associated with a reduction in epithelial cell turnover. Patients with HIVE receiving early HAART show enhanced expression of mucosal repair genes and improvement of gut symptoms. CONCLUSION Multiple lines of enquiry suggest HIVE is directly related to HIV infection and is a consequence of perturbations in mucosal CD4+Th17+ and IRIE T lymphocytes. The pathological result is enterocyte immaturity and dysfunction. SD whose main features are malabsorption, diarrhoea and weight loss, is a severe clinical expression of HIVE. A better understanding of immuno-pathogenesis of HIVE opens a window of opportunity for the potential use of immunotherapy in HIV disease and other T cell-mediated enteropathies.
Collapse
Affiliation(s)
- Moses Silungwe Kapembwa
- London Northwest Teaching Hospitals NHS Trust & Imperial College of Medicine, Northwick Park & St Mark's Hospitals, Harrow, UK.
| | - Philip Anthony Batman
- Department of Histopathology, Bradford Hospitals NHS Trust, Bradford Royal Infirmary, Bradford, UK
| | | | - George Edward Griffin
- Department of Cellular and Molecular Sciences. St George's Hospital Medical School. Cranmer Terrace, London, UK
| |
Collapse
|
8
|
Pernold CPS, Lagumdzic E, Stadler M, Dolezal M, Jäckel S, Schmitt MW, Mair KH, Saalmüller A. Species comparison: human and minipig PBMC reactivity under the influence of immunomodulating compounds in vitro. Front Immunol 2024; 14:1327776. [PMID: 38264655 PMCID: PMC10803596 DOI: 10.3389/fimmu.2023.1327776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Considering the similarities between swine and humans, it is a logical consequence to use swine as a translational model in research and drug development, including non-clinical safety. Here, we compared the reactivity of peripheral blood mononuclear cells (PBMCs) from humans and minipigs under the influence of different compounds in vitro. We conducted a flow cytometry-based proliferation assay that focused on the T-cell response to three different stimuli: concanavalin A (ConA), phytohemagglutinin-L (PHA-L), and staphylococcal Enterotoxin B (SEB). Furthermore, four approved immunosuppressive drugs-abatacept, belatacept, rapamycin, and tofacitinib-which are used for the treatment of rheumatoid arthritis or rejection in transplant recipients, were combined with the different stimuli. This allowed us to study the effect of suppressive drugs in comparison with the different stimuli in both species. We examined proliferating T cells (CD3+) and investigated the presence of TCR-αβ+ and TCR-γδ+ T cells. Differences in the response of T cells of the two species under these various conditions were evident. CD4+ T cells were more activated within humans, whereas CD8+ T cells were generally more abundant in swine. The effectiveness of the used humanized antibodies is most likely related to the conserved structure of CTLA-4 as abatacept induced a much stronger reduction in swine compared with belatacept. The reduction of proliferation of rapamycin and tofacitinib was highly dependent on the used stimuli. We further investigated the effect of the immunosuppressive compounds on antigen-specific restimulation of pigs immunized against porcine circovirus 2 (PCV2). Treatment with all four compounds resulted in a clear reduction of the proliferative response, with rapamycin showing the strongest effect. In conclusion, our findings indicate that the effectiveness of suppressive compounds is highly dependent on the stimuli used and must be carefully selected to ensure accurate results. The results highlight the importance of considering the response of T cells in different species when evaluating the potential of an immunomodulatory drug.
Collapse
Affiliation(s)
- Clara P. S. Pernold
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Emil Lagumdzic
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Sven Jäckel
- Chemical and Preclinical Safety, Merck KGaA, Darmstadt, Germany
| | | | - Kerstin H. Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
9
|
Mullan KA, de Vrij N, Valkiers S, Meysman P. Current annotation strategies for T cell phenotyping of single-cell RNA-seq data. Front Immunol 2023; 14:1306169. [PMID: 38187377 PMCID: PMC10768068 DOI: 10.3389/fimmu.2023.1306169] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has become a popular technique for interrogating the diversity and dynamic nature of cellular gene expression and has numerous advantages in immunology. For example, scRNA-seq, in contrast to bulk RNA sequencing, can discern cellular subtypes within a population, which is important for heterogenous populations such as T cells. Moreover, recent advancements in the technology allow the parallel capturing of the highly diverse T-cell receptor (TCR) sequence with the gene expression. However, the field of single-cell RNA sequencing data analysis is still hampered by a lack of gold-standard cell phenotype annotation. This problem is particularly evident in the case of T cells due to the heterogeneity in both their gene expression and their TCR. While current cell phenotype annotation tools can differentiate major cell populations from each other, labelling T-cell subtypes remains problematic. In this review, we identify the common automated strategy for annotating T cells and their subpopulations, and also describe what crucial information is still missing from these tools.
Collapse
Affiliation(s)
- Kerry A. Mullan
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS) Consortium, University of Antwerp, Antwerp, Belgium
| | - Nicky de Vrij
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS) Consortium, University of Antwerp, Antwerp, Belgium
- Clinical Immunology Unit, Department of Clinical Sciences, Institute for Tropical Medicine, Antwerp, Belgium
| | - Sebastiaan Valkiers
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS) Consortium, University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS) Consortium, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
10
|
Álvarez-Heredia P, Reina-Alfonso I, Domínguez-del-Castillo JJ, Gutiérrez-González C, Hassouneh F, Batista-Duharte A, Pérez AB, Tarazona R, Solana R, Pera A. Accelerated T-Cell Immunosenescence in Cytomegalovirus-Seropositive Individuals After Severe Acute Respiratory Syndrome Coronavirus 2 Infection. J Infect Dis 2023; 228:576-585. [PMID: 37103009 PMCID: PMC10469128 DOI: 10.1093/infdis/jiad119] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 04/28/2023] Open
Abstract
Cytomegalovirus (CMV) infection is a major driver of accelerated immunosenescence related to CD28null T cell expansion. CMV infection and these proatherogenic T cells have been independently associated with cardiovascular disease and coronavirus disease 2019 (COVID-19) severity. We investigated the potential contribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to immunosenescence and its relationship with CMV. Innate and adaptive immune subpopulations from individuals with mild or asymptomatic SARS-CoV-2 infection (mCOVID-19) and healthy donors were immunophenotyped. A significant increase in CD28nullCD57+CX3CR1+ T cell percentages (CD4+ [P ≤ .01], CD8+ [P ≤ .01], and TcRγδ (CD4-CD8-) [P ≤ .001]) was found in unnvaccinated CMV-seropositive mCOVID-19 individuals stable up to 12 months after infection. This expansion did not occur in CMV-seronegative mCOVID-19 individuals or in CMV-seropositive individuals infected after SARS-CoV-2 vaccination. There were no significant differences between mCOVID-19 and aortic stenosis groups. Thus, individuals coinfected with SARS-CoV-2 and CMV have accelerated T cell senescence, which might lead to an increased risk of cardiovascular disease.
Collapse
Affiliation(s)
- Pablo Álvarez-Heredia
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Irene Reina-Alfonso
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Jose Joaquín Domínguez-del-Castillo
- Cardiovascular Pathology (GA09), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Carmen Gutiérrez-González
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Fakhri Hassouneh
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Alexander Batista-Duharte
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Ana-Belén Pérez
- Microbiology Service, Reina Sofia University Hospital of Cordoba/Maimonides Biomedical Research Institute (IMIBIC)/CIBERINFEC, Córdoba, Spain
| | - Raquel Tarazona
- Department of Physiology, Immunology Unit, University of Extremadura, Cáceres, Spain
| | - Rafael Solana
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Alejandra Pera
- Immunology and Allergy Group (GC01), Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/University of Cordoba/Reina Sofia University Hospital, Córdoba, Spain
| |
Collapse
|
11
|
Darbouret- Hervier A, Assi N, Asensio MJ, Bernabe B, Lechevallier A, Iantomasi R, Rokbi B, Botelho-Nevers E, Ruiz S. Anti-staphylococcus aureus adaptive immunity is impaired in end-stage renal disease patients on hemodialysis: one-year longitudinal study. Front Immunol 2023; 14:1123160. [PMID: 37304264 PMCID: PMC10250961 DOI: 10.3389/fimmu.2023.1123160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/27/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Patients with end-stage renal disease (ESRD) display defects in adaptive and innate immunity, increasing susceptibility to infection. Staphylococcus aureus (S. aureus) is a major cause of bacteraemia in this population and is associated with increased mortality. More information on the immune response to S. aureus in these patients is needed to inform effective vaccine development. Methods A longitudinal prospective study was carried out at two medical centers and included 48 ESRD patients who started chronic hemodialysis (HD) treatment ≤3 months before inclusion. Control samples were taken from 62 consenting healthy blood donors. Blood samples were obtained from ESRD patients at each visit, on month (M) 0 (beginning of HD), M6 and M12. Around 50 immunological markers of adaptive and innate immunity were assessed to compare immune responses to S. aureus in ESRD patients versus controls to document the changes on their immune profile during HD. Results S. aureus survival in whole blood was significantly higher in ESRD patients than in controls at M0 (P=0.049), while impaired oxidative burst activity was observed in ESRD patients at all timepoints (P<0.001). S. aureus-specific immunoglobulin G (IgG) responses to iron surface determinant B (IsdB) and S. aureus α hemolysin (Hla) antigens were lower in ESRD patients than in healthy donors at M0 (P=0.003 and P=0.007, respectively) and M6 (P=0.05 and P=0.03, respectively), but were restored to control levels at M12. Moreover, S. aureus-specific T-helper cell responses were comparable to controls for IsdB but were impaired for Hla antigen at all timepoints: 10% of ESRD patients responded to Hla at M0, increasing to 30% at M12, compared with 45% of healthy donors. B-cell and T-cell concentrations in blood were significantly reduced (by 60% and 40%, respectively) compared with healthy controls. Finally, upregulation of Human Leucocyte Antigen-DR (HLA-DR) and C-C chemokine Receptor type 2 (CCR2) was impaired at M0 but was restored during the first year of HD. Conclusion All together, these results show that adaptive immunity was largely impaired in ESRD patients, whereas innate immunity was less impacted and tended to be restored by HD.
Collapse
Affiliation(s)
| | - Nada Assi
- Research Department, Sanofi, Marcy l’Etoile, France
| | | | | | | | | | - Bachra Rokbi
- Research Department, Sanofi, Marcy l’Etoile, France
| | - Elisabeth Botelho-Nevers
- Infectious Diseases Department, University Hospital, Saint-Etienne, France
- CIC Inserm, University Hospital, Saint-Etienne, France
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, University, Lyon, Université Jean Monnet, Inserm, CNRS, Saint-Etienne, France
| | - Sophie Ruiz
- Research Department, Sanofi, Marcy l’Etoile, France
| |
Collapse
|
12
|
Tuovinen EA, Pöysti S, Hamdan F, Le KM, Keskitalo S, Turunen T, Minier L, Mamia N, Heiskanen K, Varjosalo M, Cerullo V, Kere J, Seppänen MRJ, Hänninen A, Grönholm J. Characterization of Expanded Gamma Delta T Cells from Atypical X-SCID Patient Reveals Preserved Function and IL2RG-Mediated Signaling. J Clin Immunol 2023; 43:358-370. [PMID: 36260239 PMCID: PMC9892142 DOI: 10.1007/s10875-022-01375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/25/2022] [Indexed: 02/05/2023]
Abstract
Abnormally high γδ T cell numbers among individuals with atypical SCID have been reported but detailed immunophenotyping and functional characterization of these expanded γδ T cells are limited. We have previously reported atypical SCID phenotype caused by hypomorphic IL2RG (NM_000206.3) c.172C > T;p.(Pro58Ser) variant. Here, we have further investigated the index patient's abnormally large γδ T cell population in terms of function and phenotype by studying IL2RG cell surface expression, STAT tyrosine phosphorylation and blast formation in response to interleukin stimulation, immunophenotyping, TCRvγ sequencing, and target cell killing. In contrast to his ⍺β T cells, the patient's γδ T cells showed normal IL2RG cell surface expression and normal or enhanced IL2RG-mediated signaling. Vδ2 + population was proportionally increased with a preponderance of memory phenotypes and high overall tendency towards perforin expression. The patient's γδ T cells showed enhanced cytotoxicity towards A549 cancer cells. His TCRvγ repertoire was versatile but sequencing of IL2RG revealed a novel c.534C > A; p.(Phe178Leu) somatic missense variant restricted to γδ T cells. Over time this variant became predominant in γδ T cells, though initially present only in part of them. IL2RG-Pro58Ser/Phe178Leu variant showed higher cell surface expression compared to IL2RG-Pro58Ser variant in stable HEK293 cell lines, suggesting that somatic p.(Phe178Leu) variant may at least partially rescue the pathogenic effect of germline p.(Pro58Ser) variant. In conclusion, our report indicates that expansion of γδ T cells associated with atypical SCID needs further studying and cannot exclusively be deemed as a homeostatic response to low numbers of conventional T cells.
Collapse
Affiliation(s)
- Elina A Tuovinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Sakari Pöysti
- Department of Clinical Microbiology and Immunology, Turku University Hospital, Turku, Finland
| | - Firas Hamdan
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Drug Research Program Helsinki (DRP), Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
| | - Kim My Le
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Salla Keskitalo
- Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tanja Turunen
- Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Léa Minier
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Faculty of Science and Technology, University of Lille, Lille, France
| | - Nanni Mamia
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Kaarina Heiskanen
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Children's Immunodeficiency Unit, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Markku Varjosalo
- Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Drug Research Program Helsinki (DRP), Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
| | - Juha Kere
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Mikko R J Seppänen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- Rare Diseases Center and Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Arno Hänninen
- Department of Clinical Microbiology and Immunology, Turku University Hospital, Turku, Finland
| | - Juha Grönholm
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
13
|
Sulzmaier FJ, Kern N, Ahn SJ, Polovina A, Ho J, Hussain A, Cyprus G, Macedo C, Pandit R, Crago W, Rowell E, Timmer JC, Eckelman BP. INBRX-120, a CD8α-targeted detuned IL-2 that selectively expands and activates tumoricidal effector cells for safe and durable in vivo responses. J Immunother Cancer 2023; 11:jitc-2022-006116. [PMID: 36653071 PMCID: PMC9853254 DOI: 10.1136/jitc-2022-006116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND As a major driver of lymphocyte proliferation and activation interleukin 2 (IL-2) is a crucial mediator for antitumor responses. Despite promising activity in a subset of patients, wider therapeutic utility of IL-2 (aldesleukin) has been hampered by severe dose-limiting toxicities, the expansion of immunosuppressive regulatory T cells and a poor pharmacokinetic (PK) profile. Recent engineering efforts, including non-α IL-2 variants, have lowered the toxicity profile, but have yet to induce meaningful antitumor activity in a wider patient population. METHODS We engineered INBRX-120, a CD8α-targeted Cisleukin™ molecule consisting of an affinity tuned IL-2 (IL2-x) connected to two high affinity CD8α-specific single domain antibodies via an effector-silenced Fc domain. To show that this large affinity differential enables directed IL-2 cis-signaling exclusively on CD8α-expressing tumoricidal effector cell populations, INBRX-120 effects on target cell expansion, activation and antitumor activity were tested in vitro. In vivo antitumor efficacy was evaluated in syngeneic mouse models alone or in combination with programmed cell death protein-1 (PD-1) blockade. Preclinical safety, as well as pharmacodynamic (PD) and PK profiling was carried out in non-human primates. RESULTS INBRX-120 effectively expanded and enhanced the cytotoxic capacity of CD8 T cells and natural killer cells towards tumor cells without affecting regulatory T cells in vitro and in vivo. In syngeneic mouse models, INBRX-120 surrogate showed safe, potent, and durable antitumor efficacy alone and in combination with PD-1 blockade. In non-human primates, INBRX-120 expanded and activated CD8α-expressing effector cells, showed a favorable PK profile, and was well tolerated up to a dose of 1 mg/kg. CONCLUSIONS Through its unique cis-signaling activity on CD8α-expressing effector cells, INBRX-120 overcomes the major limitations of IL-2-based therapy and effectively harnesses IL-2's potent intrinsic antitumor activity. This novel therapeutic strategy promises safer clinical activity that could induce meaningful antitumor efficacy in a wider set of patients with various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | - Jason Ho
- Inhibrx Inc, La Jolla, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Corsale AM, Di Simone M, Lo Presti E, Dieli F, Meraviglia S. γδ T cells and their clinical application in colon cancer. Front Immunol 2023; 14:1098847. [PMID: 36793708 PMCID: PMC9923022 DOI: 10.3389/fimmu.2023.1098847] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
In recent years, research has focused on colorectal cancer to implement modern treatment approaches to improve patient survival. In this new era, γδ T cells constitute a new and promising candidate to treat many types of cancer because of their potent killing activity and their ability to recognize tumor antigens independently of HLA molecules. Here, we focus on the roles that γδ T cells play in antitumor immunity, especially in colorectal cancer. Furthermore, we provide an overview of small-scale clinical trials in patients with colorectal cancer employing either in vivo activation or adoptive transfer of ex vivo expanded γδ T cells and suggest possible combinatorial approaches to treat colon cancer.
Collapse
Affiliation(s)
- Anna Maria Corsale
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| | - Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| | - Elena Lo Presti
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR)I, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnosis (Bi.N.D.) University of Palermo, Palermo, Italy
| |
Collapse
|
15
|
Abstract
ABSTRACT Multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), a pathologically similar disease used to model MS in rodents, are typical CD4+ T cell-dominated autoimmune diseases. CD4+ interleukin (IL)17+ T cells (Th17 cells) have been well studied and have shown that they play a critical role in the pathogenesis of MS/EAE. However, studies have suggested that CD8+IL17+ T cells (Tc17 cells) have a similar phenotype and cytokine and transcription factor profiles to those of Th17 cells and have been found to be crucial in the pathogenesis of autoimmune diseases, including MS/EAE, psoriasis, type I diabetes, rheumatoid arthritis, and systemic lupus erythematosus. However, the evidence for this is indirect and insufficient. Therefore, we searched for related publications and attempted to summarize the current knowledge on the role of Tc17 cells in the pathogenesis of MS/EAE, as well as in the pathogenesis of other autoimmune diseases, and to find out whether Tc17 cells or Th17 cells play a more critical role in autoimmune disease, especially in MS and EAE pathogenesis, or whether the interaction between these two cell types plays a critical role in the development of the disease.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412000, China
| | - Xiang Deng
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412000, China
| | - Qiuming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yandan Tang
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan 412000, China
| |
Collapse
|
16
|
Pernold CP, Lagumdzic E, Stadler M, Mair KH, Jäckel S, Schmitt MW, Ladinig A, Knecht C, Dürlinger S, Kreutzmann H, Martin V, Sawyer S, Saalmüller A. Characterization of the immune system of Ellegaard Göttingen Minipigs - An important large animal model in experimental medicine. Front Immunol 2022; 13:1003986. [PMID: 36203585 PMCID: PMC9531550 DOI: 10.3389/fimmu.2022.1003986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Interest in Ellegaard Göttingen Minipigs (EGMs) as a model in experimental medicine is continuously growing. The aim of this project is to increase the knowledge of the immune system of EGMs as information is still scarce. Therefore, we studied the postnatal maturation of their immune system from birth until 126 weeks of age. For the first 26 weeks of the study, animals were kept under pathogen-reduced conditions (SPF) and afterwards under conventional housing conditions. The development of the immune system was analyzed by monitoring changes in total numbers of leukocytes and lymphocytes of ten individuals and the composition of leukocyte populations by multi-color flow cytometry (FCM). We followed the presence of monocytes using monoclonal antibodies (mAbs) against CD172a+ and CD163+ and B cells based on the expression of CD79a. NK cells were distinguished as CD3-CD16+CD8α+/dim cells and further subdivided using NKp46 (CD335) expression into NKp46-, NKp46+, and NKp46high NK cells. T-cell receptor (TCR) γδ T cells were defined by the expression of TCR-γδ and different subsets were determined by their CD2 and perforin expression. TCR-αβ T cells were classified by their CD8β+ or CD4 expression. For monitoring their differentiation, expression of CD27 and perforin was investigated for CD8β++ T cells and CD8α together with CD27 for CD4+ T cells. We clearly detected a postnatal development of immune cell composition and identified phenotypes indicative of differentiation within the respective leukocyte subsets. Examination of the development of the antigen-specific immune system after transfer to different distinct housing conditions and after vaccination against common porcine pathogens such as porcine circovirus 2 (PCV2) revealed a markedly increased presence of more differentiated CD8+ and CD4+ T cells with central and effector memory T-cell phenotypes. To complement the findings, a PCV2 vaccine-specific antigen was used for in vitro restimulation experiments. We demonstrated antigen-specific proliferation of CD4+CD8α+CD27+ central and CD4+CD8α+CD27- effector memory T cells as well as antigen-specific production of TNF-α and IFN-γ. This study of postnatal immune development defines basic cellular immune parameters of EGMs and represents an important milestone for the use of EGMs for immunological questions in experimental medicine.
Collapse
Affiliation(s)
- Clara P.S. Pernold
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Emil Lagumdzic
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kerstin H. Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Christian Doppler (CD) Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sven Jäckel
- Chemical and Preclinical Safety, Merck KGaA, Darmstadt, Germany
| | | | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Christian Knecht
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sophie Dürlinger
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Heinrich Kreutzmann
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Vera Martin
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Spencer Sawyer
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
17
|
Li Y, Jiang S, Li J, Yin M, Yan F, Chen Y, Chen Y, Wu T, Cheng M, He Y, Liang H, Yu H, Qiao Q, Guo Z, Xu Y, Zhang Y, Xiang Z, Yin Z. Phenotypic Changes of Peripheral γδ T Cell and Its Subsets in Patients With Coronary Artery Disease. Front Immunol 2022; 13:900334. [PMID: 35874761 PMCID: PMC9304556 DOI: 10.3389/fimmu.2022.900334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Coronary atherosclerotic heart disease (CAD) is a chronic inflammatory cardiovascular disease with high morbidity and mortality. Growing data indicate that many immune cells are involved in the development of atherosclerosis. However, the immunological roles of γδ T cells in the initiation and progression of CAD are not fully understood. Here, we used flow cytometry to determine phenotypical changes of γδ T cells and their subpopulations in peripheral blood samples collected from 37 CAD patients. The Pearson correlation coefficient was used to analyze the relationship between the clinical parameter (serum LDL-C level) and the changes of immunophenotypes of γδ T cells. Our results demonstrated that the frequencies and absolute numbers of total γδ T cells and Vδ2+ T cells were significantly decreased in CAD patients when compared to healthy individuals. However, the proportion of Vδ1+ T cells was much lower in CAD patients than that of healthy individuals. Most importantly, a significant alteration of the Vδ1/Vδ2 ratio was found in CAD patients. In addition, a series of surface markers that are associated with costimulatory signals (CD28, CD40L, CD80, CD86), activation levels (CD69, CD25, HLA-DR), activating NK cell receptors (NKp30, NKp46, NKG2D) and inhibitory receptors (PD-1, CTLA-4, PD-1, Tim-3) were determined and then analyzed in the total γδ T cells, Vδ2+T cells and Vδ2-T cells of CAD patients and healthy individuals. The data demonstrated that immunological activities of total γδ T cells, Vδ2+T cells, and Vδ2-T cells of CAD patients were much lower than those in healthy individuals. Moreover, we found that there were positive correlations between the serum LDL-C levels and frequencies of CD3+γδ+ T cells, CD69+Vδ2+T cells, NKG2D+Vδ2+T cells, and NKp46+Vδ2+T cells. By contrast, there was an inverse correlation between the levels of serum LDL-C and the frequencies of CD69+Vδ2-T cells and NKp46+Vδ2-T cells. Accordingly, these findings could help us to better understand the roles of γδ T cells in the CAD, and shed light on the development of novel diagnostic techniques and therapeutic strategies by targeting γδ T cells for CAD patients.
Collapse
Affiliation(s)
- Yan Li
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Silin Jiang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Jiawei Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Mengzhuo Yin
- Department of Geriatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Fuxin Yan
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuyuan Chen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Yan Chen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Tongwei Wu
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengliang Cheng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihua He
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbin Liang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hang Yu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Qingqing Qiao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Xu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
- *Correspondence: Zhinan Yin, ; Zheng Xiang, ; Yan Xu, ; Yanan Zhang,
| | - Yanan Zhang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Zhinan Yin, ; Zheng Xiang, ; Yan Xu, ; Yanan Zhang,
| | - Zheng Xiang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
- *Correspondence: Zhinan Yin, ; Zheng Xiang, ; Yan Xu, ; Yanan Zhang,
| | - Zhinan Yin
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
- *Correspondence: Zhinan Yin, ; Zheng Xiang, ; Yan Xu, ; Yanan Zhang,
| |
Collapse
|
18
|
Targeting Immunosuppressive Tumor-Associated Macrophages Using Innate T Cells for Enhanced Antitumor Reactivity. Cancers (Basel) 2022; 14:cancers14112749. [PMID: 35681730 PMCID: PMC9179365 DOI: 10.3390/cancers14112749] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 01/03/2023] Open
Abstract
The field of T cell-based and chimeric antigen receptor (CAR)-engineered T (CAR-T) cell-based antitumor immunotherapy has seen substantial developments in the past decade; however, considerable issues, such as graft-versus-host disease (GvHD) and tumor-associated immunosuppression, have proven to be substantial roadblocks to widespread adoption and implementation. Recent developments in innate immune cell-based CAR therapy have opened several doors for the expansion of this therapy, especially as it relates to allogeneic cell sources and solid tumor infiltration. This study establishes in vitro killing assays to examine the TAM-targeting efficacy of MAIT, iNKT, and γδT cells. This study also assesses the antitumor ability of CAR-engineered innate T cells, evaluating their potential adoption for clinical therapies. The in vitro trials presented in this study demonstrate the considerable TAM-killing abilities of all three innate T cell types, and confirm the enhanced antitumor abilities of CAR-engineered innate T cells. The tumor- and TAM-targeting capacity of these innate T cells suggest their potential for antitumor therapy that supplements cytotoxicity with remediation of tumor microenvironment (TME)-immunosuppression.
Collapse
|
19
|
CD161 expression defines new human γδ T cell subsets. IMMUNITY & AGEING 2022; 19:11. [PMID: 35193613 PMCID: PMC8862246 DOI: 10.1186/s12979-022-00269-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/14/2022] [Indexed: 11/10/2022]
Abstract
Abstractγδ T cells are a highly versatile immune lineage involved in host defense and homeostasis, but questions remain around their heterogeneity, precise function and role during health and disease. We used multi−parametric flow cytometry, dimensionality reduction, unsupervised clustering, and self-organizing maps (SOM) to identify novel γδ T cell naïve/memory subsets chiefly defined by CD161 expression levels, a surface membrane receptor that can be activating or suppressive. We used middle-to-old age individuals given immune blockade is commonly used in this population. Whilst most Vδ1+subset cells exhibited a terminal differentiation phenotype, Vδ1− subset cells showed an early memory phenotype. Dimensionality reduction revealed eight γδ T cell clusters chiefly diverging through CD161 expression with CD4 and CD8 expression limited to specific subpopulations. Comparison of matched healthy elderly individuals to bronchiectasis patients revealed elevated Vδ1+ terminally differentiated effector memory cells in patients potentially linking this population with chronic proinflammatory disease.
Collapse
|
20
|
Lee SI, Jeong CG, Ul Salam Mattoo S, Nazki S, Prasad Aganja R, Kim SC, Khatun A, Oh Y, Noh SH, Lee SM, Kim WI. Protective immunity induced by concurrent intradermal injection of porcine circovirus type 2 and Mycoplasma hyopneumoniae inactivated vaccines in pigs. Vaccine 2021; 39:6691-6699. [PMID: 34538524 DOI: 10.1016/j.vaccine.2021.07.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/25/2021] [Accepted: 07/16/2021] [Indexed: 11/15/2022]
Abstract
Vaccines against porcine circovirus type 2 (PCV2) and Mycoplasma hyopneumoniae (Mhp) are routinely used by intramuscular injection. However, since intramuscular vaccination causes stress and increases the risk of cross-contamination among pigs, research on intradermal vaccination is currently being actively conducted. This study was designed to evaluate the efficacy of intradermally administered inactivated vaccines against PCV2 and Mhp in pigs. Three-week-old specific pathogen-free pigs were divided into three groups (5 pigs per group). Pigs in the two groups were intradermally vaccinated with the PCV2 or Mhp vaccine using a needle-free injector. Pigs in the third group were kept as nonvaccinated controls. At 21 days post-vaccination, pigs in one of these vaccinated groups and the nonvaccinated group were intranasally challenged with PCV2b and Mhp, while the other vaccinated group pigs were maintained as vaccine controls. Vaccine efficacy was evaluated by observing weight gain, pathogen load, pathological changes, and humoral or cellular immune responses. As a result, vaccinated pigs revealed significantly higher body weight gain, with lower clinical scores. Vaccinated pigs also showed higher antibody responses but lower PCV2b or Mhp loads in sera, nasal swabs, or lungs than nonvaccinated pigs. Intriguingly, vaccinated pigs upregulated cytotoxic T cells (CTLs), helper T type 1 cells (Th1 cells), and helper T type 17 cells (Th17 cells) after immunization and showed significantly higher levels of CTLs, Th1 and Th17 cells at 14 days post-challenge than nonvaccinated and challenged pigs. This study demonstrated that protective immune responses against PCV2 and Mhp could be efficiently induced in pigs using a relatively small volume of intradermal vaccines, probably due to effective antigen delivery to antigen-presenting cells in the dermis.
Collapse
Affiliation(s)
- Sim-In Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| | - Chang-Gi Jeong
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| | | | - Salik Nazki
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea; The Pirbright Institute, Ash Road, Pirbright-GU24 0NF, Woking, United Kingdom.
| | - Ram Prasad Aganja
- Division of Biotechnology, Jeonbuk National University, Iksan, Republic of Korea.
| | - Seung-Chai Kim
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| | - Amina Khatun
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea; Department of Pathology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Sher-e-Bangla Nagar, Dhaka 1207, Bangladesh.
| | - Yeonsu Oh
- Department of Veterinary Pathology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Sang-Hyun Noh
- MSD Animal Health Korea Ltd., Seoul 04637, Republic of Korea.
| | - Sang-Myeong Lee
- Division of Biotechnology, Jeonbuk National University, Iksan, Republic of Korea.
| | - Won-Il Kim
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| |
Collapse
|
21
|
Keddache S, Laheurte C, Boullerot L, Laurent L, Dalphin JC, Adotevi O, Soumagne T. Inflammatory and immunological profile in COPD secondary to organic dust exposure. Clin Immunol 2021; 229:108798. [PMID: 34280578 DOI: 10.1016/j.clim.2021.108798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/22/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Inflammatory response in patients with COPD secondary to organic dust exposure (OD-COPD) is poorly understood. We therefore aimed to characterize inflammatory and immune profile from peripheral blood mononuclear cells (PBMC) in a group of patients with mild-to-moderate COPD secondary to organic dust exposure (OD-COPD), tobacco smoking (T-COPD), or both. We compared T, B and NK cells distribution and inflammatory (TNF-α, Il-1β, IL-6), type 1 (IFN-γ), type 2 (IL-4, IL-13) and type 3 (IL-17) immunity related cytokines at baseline, and after stimulation with LPS, flagellin and CD3/CD28 beads in all COPD groups. OD-COPD displayed significantly lower NK cells and CD8+ T cells compared with controls. After flagellin stimulation, T-COPD had significantly lower IL-13 levels than OD-COPD and controls (p < 0.05) whereas IFN-γ tended to be lower in OD-COPD. All COPD groups displayed higher IL-1β and IL-17 than controls after CD3/CD28 stimulation. Inflammatory responses in OD-COPD were different from T-COPD. OD-COPD displayed higher levels of type 2 immunity related cytokines.
Collapse
Affiliation(s)
- Sophia Keddache
- Service de Pneumologie, Oncologie Thoracique et Allergologie Respiratoire, CHRU de Besançon, France; Service de Pneumologie, Hôpital Bicêtre, AP-, HP, Le Kremlin-Bicêtre, France
| | - Caroline Laheurte
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Université de Bourgogne Franche-Comté, Besançon, France; EFS Bourgogne Franche Comté, Plateforme de BioMonitoring, INSERM CIC-1431/UMR1098, 25000 Besançon, France
| | - Laura Boullerot
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Université de Bourgogne Franche-Comté, Besançon, France; EFS Bourgogne Franche Comté, Plateforme de BioMonitoring, INSERM CIC-1431/UMR1098, 25000 Besançon, France
| | - Lucie Laurent
- Service de Pneumologie, Oncologie Thoracique et Allergologie Respiratoire, CHRU de Besançon, France
| | - Jean-Charles Dalphin
- Service de Pneumologie, Oncologie Thoracique et Allergologie Respiratoire, CHRU de Besançon, France
| | - Olivier Adotevi
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Université de Bourgogne Franche-Comté, Besançon, France; EFS Bourgogne Franche Comté, Plateforme de BioMonitoring, INSERM CIC-1431/UMR1098, 25000 Besançon, France
| | - Thibaud Soumagne
- Service de Pneumologie, Oncologie Thoracique et Allergologie Respiratoire, CHRU de Besançon, France.
| |
Collapse
|
22
|
Knight V, Heimall JR, Chong H, Nandiwada SL, Chen K, Lawrence MG, Sadighi Akha AA, Kumánovics A, Jyonouchi S, Ngo SY, Vinh DC, Hagin D, Forbes Satter LR, Marsh RA, Chiang SCC, Willrich MAV, Frazer-Abel AA, Rider NL. A Toolkit and Framework for Optimal Laboratory Evaluation of Individuals with Suspected Primary Immunodeficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:3293-3307.e6. [PMID: 34033983 DOI: 10.1016/j.jaip.2021.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022]
Abstract
Knowledge related to the biology of inborn errors of immunity and associated laboratory testing methods continues to expand at a tremendous rate. Despite this, many patients with inborn errors of immunity suffer for prolonged periods of time before identification of their underlying condition, thereby delaying appropriate care. Understanding that test selection and optimal evaluation for patients with recurrent infections or unusual patterns of inflammation can be unclear, we present a document that distills relevant clinical features of immunologic disease due to inborn errors of immunity and related appropriate and available test options. This document is intended to serve the practicing clinical immunologist and, in turn, patients by describing best available test options for initial and expanded immunologic evaluations across the disease spectrum. Our goal is to demystify the process of evaluating patients with suspected immune dysfunction and to enable more rapid and accurate diagnosis of such individuals.
Collapse
Affiliation(s)
- Vijaya Knight
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado School of Medicine, Aurora, Colo
| | - Jennifer R Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Perlman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | - Hey Chong
- Division of Pulmonary Medicine, Allergy and Immunology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pa
| | - Sarada L Nandiwada
- The Texas Children's Hospital, Section of Immunology, Allergy and Retrovirology, The Baylor College of Medicine and the William T. Shearer Center for Human Immunobiology, Houston, Tex
| | - Karin Chen
- Department of Immunology, University of Washington and Seattle Children's Hospital, Seattle, Wash
| | - Monica G Lawrence
- Division of Asthma, Allergy and Clinical Immunology, University of Virginia, Charlottesville, Va
| | - Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - Attila Kumánovics
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - Soma Jyonouchi
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Perlman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | - Suzanne Y Ngo
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado School of Medicine, Aurora, Colo
| | - Donald C Vinh
- Division of Infectious Diseases, Allergy & Clinical Immunology, Department of Medical Microbiology and Human Genetics, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lisa R Forbes Satter
- The Texas Children's Hospital, Section of Immunology, Allergy and Retrovirology, The Baylor College of Medicine and the William T. Shearer Center for Human Immunobiology, Houston, Tex
| | - Rebecca A Marsh
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Samuel C C Chiang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Maria A V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - Ashley A Frazer-Abel
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colo
| | - Nicholas L Rider
- The Texas Children's Hospital, Section of Immunology, Allergy and Retrovirology, The Baylor College of Medicine and the William T. Shearer Center for Human Immunobiology, Houston, Tex.
| |
Collapse
|
23
|
Hanson ED, Bates LC, Bartlett DB, Campbell JP. Does exercise attenuate age- and disease-associated dysfunction in unconventional T cells? Shining a light on overlooked cells in exercise immunology. Eur J Appl Physiol 2021; 121:1815-1834. [PMID: 33822261 DOI: 10.1007/s00421-021-04679-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
Unconventional T Cells (UTCs) are a unique population of immune cells that links innate and adaptive immunity. Following activation, UTCs contribute to a host of immunological activities, rapidly responding to microbial and viral infections and playing key roles in tumor suppression. Aging and chronic disease both have been shown to adversely affect UTC numbers and function, with increased inflammation, change in body composition, and physical inactivity potentially contributing to the decline. One possibility to augment circulating UTCs is through increased physical activity. Acute exercise is a potent stimulus leading to the mobilization of immune cells while the benefits of exercise training may include anti-inflammatory effects, reductions in fat mass, and improved fitness. We provide an overview of age-related changes in UTCs, along with chronic diseases that are associated with altered UTC number and function. We summarize how UTCs respond to acute exercise and exercise training and discuss potential mechanisms that may lead to improved frequency and function.
Collapse
Affiliation(s)
- Erik D Hanson
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Lauren C Bates
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA.,Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David B Bartlett
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC, USA
| | | |
Collapse
|
24
|
Wu J, Abraham SN. The Roles of T cells in Bladder Pathologies. Trends Immunol 2021; 42:248-260. [PMID: 33536141 PMCID: PMC7914211 DOI: 10.1016/j.it.2021.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/26/2022]
Abstract
T lymphocytes play important roles in the skin and mucosal surfaces such as the gut and lung. Until recently the contributions of T cells to mammalian bladder immunity were largely unknown. With newer techniques, including single-cell RNA sequencing and reporter mice, an understanding is emerging of T cell roles in bladder diseases (bacterial infections, bladder cancer, chronic inflammation). In these pathologies, many bladder T cell responses can be harmful to the host through suboptimal clearance of bacteria or cancer cells, or by modulating autoinflammation. Recent findings suggest that T cell behavior might be influenced by resident T cell interactions with the bladder microbiota and other immunostimulants. Thus, regulating bladder T cell functions could emerge as a putative immunotherapy to treat some bladder diseases.
Collapse
Affiliation(s)
- Jianxuan Wu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Soman N Abraham
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore.
| |
Collapse
|
25
|
Craven KE, Gökmen-Polar Y, Badve SS. CIBERSORT analysis of TCGA and METABRIC identifies subgroups with better outcomes in triple negative breast cancer. Sci Rep 2021; 11:4691. [PMID: 33633150 PMCID: PMC7907367 DOI: 10.1038/s41598-021-83913-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Studies have shown that the presence of tumor infiltrating lymphocytes (TILs) in Triple Negative Breast Cancer (TNBC) is associated with better prognosis. However, the molecular mechanisms underlying these immune cell differences are not well delineated. In this study, analysis of hematoxylin and eosin images from The Cancer Genome Atlas (TCGA) breast cancer cohort failed to show a prognostic benefit of TILs in TNBC, whereas CIBERSORT analysis, which quantifies the proportion of each immune cell type, demonstrated improved overall survival in TCGA TNBC samples with increased CD8 T cells or CD8 plus CD4 memory activated T cells and in Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) TNBC samples with increased gamma delta T cells. Twenty-five genes showed mutational frequency differences between the TCGA high and low T cell groups, and many play important roles in inflammation or immune evasion (ATG2B, HIST1H2BC, PKD1, PIKFYVE, TLR3, NOTCH3, GOLGB1, CREBBP). Identification of these mutations suggests novel mechanisms by which the cancer cells attract immune cells and by which they evade or dampen the immune system during the cancer immunoediting process. This study suggests that integration of mutations with CIBERSORT analysis could provide better prediction of outcomes and novel therapeutic targets in TNBC cases.
Collapse
Affiliation(s)
- Kelly E Craven
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yesim Gökmen-Polar
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sunil S Badve
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, 46202, USA.
| |
Collapse
|
26
|
Mohtashami M, Razavi A, Abolhassani H, Aghamohammadi A, Yazdani R. Primary Immunodeficiency and Thrombocytopenia. Int Rev Immunol 2021; 41:135-159. [PMID: 33464134 DOI: 10.1080/08830185.2020.1868454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Primary immunodeficiency (PID) or Inborn errors of immunity (IEI) refers to a heterogeneous group of disorders characterized by immune system impairment. Although patients with IEI manifest highly variable symptoms, the most common clinical manifestations are recurrent infections, autoimmunity and malignancies. Some patients present hematological abnormality including thrombocytopenia due to different pathogenic mechanisms. This review focuses on primary and secondary thrombocytopenia as a complication, which can occur in IEI. Based on the International Union of Immunological Societies phenotypic classification for IEI, the several innate and adaptive immunodeficiency disorders can lead to thrombocytopenia. This review, for the first time, describes manifestation, mechanism and therapeutic modalities for thrombocytopenia in different classes of IEI.
Collapse
Affiliation(s)
- Maryam Mohtashami
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.,Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadehsadat Razavi
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran.,Department of Animal Biology, Faculty of Biology Sciences, University of Kharazmi, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Phan TS, Schink L, Mann J, Merk VM, Zwicky P, Mundt S, Simon D, Kulms D, Abraham S, Legler DF, Noti M, Brunner T. Keratinocytes control skin immune homeostasis through de novo-synthesized glucocorticoids. SCIENCE ADVANCES 2021; 7:7/5/eabe0337. [PMID: 33514551 PMCID: PMC7846173 DOI: 10.1126/sciadv.abe0337] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/10/2020] [Indexed: 05/06/2023]
Abstract
Glucocorticoids (GC), synthesized by the 11β-hydroxylase (Cyp11b1), control excessive inflammation through immunosuppressive actions. The skin was proposed to regulate homeostasis by autonomous GC production in keratinocytes. However, their immunosuppressive capacity and clinical relevance remain unexplored. Here, we demonstrate the potential of skin-derived GC and their role in the regulation of physiological and prevalent inflammatory skin conditions. In line with 11β-hydroxylase deficiency in human inflammatory skin disorders, genetic in vivo Cyp11b1 ablation and long-term GC deficiency in keratinocytes primed the murine skin immune system resulting in spontaneous skin inflammation. Deficient skin GC in experimental models for inflammatory skin disorders led to exacerbated contact hypersensitivity and psoriasiform skin inflammation accompanied by decreased regulatory T cells and the involvement of unconventional T cells. Our findings provide insights on how skin homeostasis and pathology are critically regulated by keratinocyte-derived GC, emphasizing the immunoregulatory potential of endogenous GC in the regulation of epithelial immune microenvironment.
Collapse
Affiliation(s)
- Truong San Phan
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Leonhard Schink
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Jasmin Mann
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Verena M Merk
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital University Hospital, Bern, Switzerland
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, TU-Dresden, Dresden, Germany
| | - Susanne Abraham
- Experimental Dermatology, Department of Dermatology, TU-Dresden, Dresden, Germany
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Mario Noti
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
28
|
Castro CD, Boughter CT, Broughton AE, Ramesh A, Adams EJ. Diversity in recognition and function of human γδ T cells. Immunol Rev 2020; 298:134-152. [PMID: 33136294 DOI: 10.1111/imr.12930] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/17/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
As interest increases in harnessing the potential power of tissue-resident cells for human health and disease, γδ T cells have been thrust into the limelight due to their prevalence in peripheral tissues, their sentinel-like phenotypes, and their unique antigen recognition capabilities. This review focuses primarily on human γδ T cells, highlighting their distinctive characteristics including antigen recognition, function, and development, with an emphasis on where they differ from their αβ T cell comparators, as well as from γδ T cell populations in the mouse. We review the antigens that have been identified thus far to regulate members of the human Vδ1 population and discuss what players are involved in transducing phosphoantigen-mediated signals to human Vγ9Vδ2 T cells. We also briefly review distinguishing features of these cells in terms of TCR signaling, use of coreceptor and costimulatory molecules and their development. These cells have great potential to be harnessed in a clinical setting, but caution must be taken to understand their unique capabilities and how they differ from the populations to which they are commonly compared.
Collapse
Affiliation(s)
- Caitlin D Castro
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Christopher T Boughter
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL, USA
| | - Augusta E Broughton
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Amrita Ramesh
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| | - Erin J Adams
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, USA
| |
Collapse
|
29
|
Magnoumba M, Singh A, Ogongo P, Roider J, Asowata O, Fehlings M, Karim F, Ndung'u T, Anderson F, Leslie A, Kløverpris H. Unbiased Profiling Reveals Compartmentalization of Unconventional T-Cells Within the Intestinal Mucosa Irrespective of HIV Infection. Front Immunol 2020; 11:579743. [PMID: 33117384 PMCID: PMC7561384 DOI: 10.3389/fimmu.2020.579743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022] Open
Abstract
The intestinal mucosa is enriched for unconventional T-cells, including mucosal associated invariant T-cells (MAIT), invariant natural killer T-cells (iNKT) and γδ T-cells. These cells are activated by bacterial metabolites, lipid antigens and cytokines, and are important for intestinal barrier integrity. The loss of gut homeostasis observed in HIV infection is central to disease pathogenesis, and studies have highlighted impairment of particular unconventional T-cell subsets within a specific gut compartment. However, although the small and large intestine are distinct niches, the overall impact of HIV on unconventional T-cells across the gut mucosal has not been well-studied. We hypothesized that compartment specific differences in the unconventional T-cell repertoire would exist between the small and large intestine, due to increasing bacterial loads and microbial diversity; and that the impact of HIV infection might differ depending on the compartment examined. We used mass cytometry, flow cytometry and unbiased T-cell receptor profiling to quantify unconventional T-cells in blood and tissue from the small (duodenum) and large (colon) intestine in HIV infected and uninfected participants undergoing examination for a range of intestinal conditions. Overall, we find distinct compartmentalisation of T-cells between blood, duodenum and colon, with iNKT cells significantly enriched in the duodenum and δ-1 expressing γδ T-cells in the colon. In addition, we observe greater clonal expansion of conventional TCRs in the duodenum, suggestive of stronger adaptive immunity in this compartment. Conversely, we find evidence of an expanded unconventional TCR repertoire in the colon, which contained far more overlapping “donor unrestricted” sequences than the duodenum. Twelve of these TCRs were highly “MAIT-like” and 3 were unique to the colon, suggesting an enrichment of donor unrestricted T-cells (DURTs) in this compartment. Unexpectedly, however, no significant impact of HIV infection on any of the unconventional T-cell subsets measured was observed in either mucosal site in terms of frequency or TCR repertoire. Further studies are required to investigate the importance of these unconventional T-cell subsets to intestinal homeostasis within the different gut compartments and determine if they are functionally impaired during HIV infection.
Collapse
Affiliation(s)
- Magalli Magnoumba
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Alveera Singh
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Paul Ogongo
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa.,Institute of Primate Research, National Museums of Kenya, Nairobi, Kenya
| | - Julia Roider
- Department of Infectious Diseases, Medizinische Klinik IV, Ludwig-Maximilians-University Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Osaretin Asowata
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa
| | | | - Farina Karim
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Thumbi Ndung'u
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa.,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.,Division of Infection and Immunity, University College London (UCL), London, United Kingdom
| | - Frank Anderson
- Department of Surgery, Inkosi Albert Luthuli Hospital, Durban, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa.,Division of Infection and Immunity, University College London (UCL), London, United Kingdom
| | - Henrik Kløverpris
- Africa Health Research Institute (AHRI), University of KwaZulu-Natal (UKZN), Durban, South Africa.,Division of Infection and Immunity, University College London (UCL), London, United Kingdom.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Zernecke A, Winkels H, Cochain C, Williams JW, Wolf D, Soehnlein O, Robbins CS, Monaco C, Park I, McNamara CA, Binder CJ, Cybulsky MI, Scipione CA, Hedrick CC, Galkina EV, Kyaw T, Ghosheh Y, Dinh HQ, Ley K. Meta-Analysis of Leukocyte Diversity in Atherosclerotic Mouse Aortas. Circ Res 2020; 127:402-426. [PMID: 32673538 PMCID: PMC7371244 DOI: 10.1161/circresaha.120.316903] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The diverse leukocyte infiltrate in atherosclerotic mouse aortas was recently analyzed in 9 single-cell RNA sequencing and 2 mass cytometry studies. In a comprehensive meta-analysis, we confirm 4 known macrophage subsets-resident, inflammatory, interferon-inducible cell, and Trem2 (triggering receptor expressed on myeloid cells-2) foamy macrophages-and identify a new macrophage subset resembling cavity macrophages. We also find that monocytes, neutrophils, dendritic cells, natural killer cells, innate lymphoid cells-2, and CD (cluster of differentiation)-8 T cells form prominent and separate immune cell populations in atherosclerotic aortas. Many CD4 T cells express IL (interleukin)-17 and the chemokine receptor CXCR (C-X-C chemokine receptor)-6. A small number of regulatory T cells and T helper 1 cells is also identified. Immature and naive T cells are present in both healthy and atherosclerotic aortas. Our meta-analysis overcomes limitations of individual studies that, because of their experimental approach, over- or underrepresent certain cell populations. Mass cytometry studies demonstrate that cell surface phenotype provides valuable information beyond the cell transcriptomes. The present analysis helps resolve some long-standing controversies in the field. First, Trem2+ foamy macrophages are not proinflammatory but interferon-inducible cell and inflammatory macrophages are. Second, about half of all foam cells are smooth muscle cell-derived, retaining smooth muscle cell transcripts rather than transdifferentiating to macrophages. Third, Pf4, which had been considered specific for platelets and megakaryocytes, is also prominently expressed in the main population of resident vascular macrophages. Fourth, a new type of resident macrophage shares transcripts with cavity macrophages. Finally, the discovery of a prominent innate lymphoid cell-2 cluster links the single-cell RNA sequencing work to recent flow cytometry data suggesting a strong atheroprotective role of innate lymphoid cells-2. This resolves apparent discrepancies regarding the role of T helper 2 cells in atherosclerosis based on studies that predated the discovery of innate lymphoid cells-2 cells.
Collapse
Affiliation(s)
- Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Holger Winkels
- Heart Center, University Hospital Cologne, Cologne, Germany
- Clinic III for Internal Medicine, Department of Cardiology, University of Cologne, Cologne, Germany
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Wüzburg, Germany
| | - Jesse W. Williams
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN USA
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN USA
| | - Dennis Wolf
- Department of Cardiology and Angiology I, University Heart Center, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Klinikum LMU Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Stockholm, Sweden
| | - Clint S. Robbins
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S1A1, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S1A1, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Toronto, ON M5G1L7, Canada
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Inhye Park
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, USA
- Division of Cardioascular Medicine, University of Virginia School of Medicine, Charlottesville, USA
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Myron I. Cybulsky
- Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Corey A. Scipione
- Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Elena V. Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA USA
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Huy Q. Dinh
- La Jolla Institute for Immunology, La Jolla, CA USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA USA
- Department of Bioengineering, University of California San Diego, CA, USA
| |
Collapse
|
31
|
Beucke N, Wistuba-Hamprecht K. Accurate determination of γδ T cells in multi-channel mass and flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:288-289. [PMID: 32469457 DOI: 10.1002/cyto.b.21885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/22/2020] [Indexed: 11/12/2022]
Affiliation(s)
- Nicola Beucke
- Division of Dermatooncology, Department of Dermatology, University Medical Center, Tübingen, Germany
| | - Kilian Wistuba-Hamprecht
- Division of Dermatooncology, Department of Dermatology, University Medical Center, Tübingen, Germany
| |
Collapse
|
32
|
Siedel H, Roers A, Rösen-Wolff A, Luksch H. Type I interferon-independent T cell impairment in a Tmem173 N153S/WT mouse model of STING associated vasculopathy with onset in infancy (SAVI). Clin Immunol 2020; 216:108466. [PMID: 32470544 DOI: 10.1016/j.clim.2020.108466] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/15/2020] [Accepted: 05/16/2020] [Indexed: 12/23/2022]
Abstract
STING-associated vasculopathy with onset in infancy (SAVI) is an autoimmune disease caused by heterozygous gain of function mutations of STING (stimulator of interferon genes) that had initially been classified as a type I interferonopathy. We recently reported a genetically engineered mouse strain carrying a common SAVI-associated STING mutation. These STING N153S/WT mice reproduce key features of SAVI, including lung inflammation, loss of T cells in spleen and blood, splenomegaly and thymic hypoplasia. Here we show that αβ T lymphocytopenia is due to disrupted T cell development and is associated with impaired T cell activation and a relative increase in γδ T cell numbers. These alterations were not rescued by additional knockout of the type I IFN receptor (IFNAR1). Collectively, our findings consolidate the concept that constitutive STING signalling leads to a SCID-like phenotype in STING N153S/WT mice.
Collapse
Affiliation(s)
- Hannah Siedel
- Department of Pediatrics, University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Axel Roers
- Institute of Immunology, Medical Faculty TU Dresden, Dresden, Germany
| | - Angela Rösen-Wolff
- Department of Pediatrics, University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany.
| | - Hella Luksch
- Department of Pediatrics, University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
| |
Collapse
|
33
|
Xu W, Lau ZWX, Fulop T, Larbi A. The Aging of γδ T Cells. Cells 2020; 9:E1181. [PMID: 32397491 PMCID: PMC7290956 DOI: 10.3390/cells9051181] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
In the coming decades, many developed countries in the world are expecting the "greying" of their populations. This phenomenon poses unprecedented challenges to healthcare systems. Aging is one of the most important risk factors for infections and a myriad of diseases such as cancer, cardiovascular and neurodegenerative diseases. A common denominator that is implicated in these diseases is the immune system. The immune system consists of the innate and adaptive arms that complement each other to provide the host with a holistic defense system. While the diverse interactions between multiple arms of the immune system are necessary for its function, this complexity is amplified in the aging immune system as each immune cell type is affected differently-resulting in a conundrum that is especially difficult to target. Furthermore, certain cell types, such as γδ T cells, do not fit categorically into the arms of innate or adaptive immunity. In this review, we will first introduce the human γδ T cell family and its ligands before discussing parallels in mice. By covering the ontogeny and homeostasis of γδ T cells during their lifespan, we will better capture their evolution and responses to age-related stressors. Finally, we will identify knowledge gaps within these topics that can advance our understanding of the relationship between γδ T cells and aging, as well as age-related diseases such as cancer.
Collapse
Affiliation(s)
- Weili Xu
- Biology of Aging Program and Immunomonitoring Platform, Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore 138648, Singapore; (W.X.); (Z.W.X.L.)
| | - Zandrea Wan Xuan Lau
- Biology of Aging Program and Immunomonitoring Platform, Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore 138648, Singapore; (W.X.); (Z.W.X.L.)
| | - Tamas Fulop
- Department of Geriatrics, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada;
| | - Anis Larbi
- Biology of Aging Program and Immunomonitoring Platform, Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore 138648, Singapore; (W.X.); (Z.W.X.L.)
- Department of Geriatrics, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada;
- Department of Microbiology, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
34
|
Chruściel E, Urban-Wójciuk Z, Arcimowicz Ł, Kurkowiak M, Kowalski J, Gliwiński M, Marjański T, Rzyman W, Biernat W, Dziadziuszko R, Montesano C, Bernardini R, Marek-Trzonkowska N. Adoptive Cell Therapy-Harnessing Antigen-Specific T Cells to Target Solid Tumours. Cancers (Basel) 2020; 12:E683. [PMID: 32183246 PMCID: PMC7140076 DOI: 10.3390/cancers12030683] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
In recent years, much research has been focused on the field of adoptive cell therapies (ACT) that use native or genetically modified T cells as therapeutic tools. Immunotherapy with T cells expressing chimeric antigen receptors (CARs) demonstrated great success in the treatment of haematologic malignancies, whereas adoptive transfer of autologous tumour infiltrating lymphocytes (TILs) proved to be highly effective in metastatic melanoma. These encouraging results initiated many studies where ACT was tested as a treatment for various solid tumours. In this review, we provide an overview of the challenges of T cell-based immunotherapies of solid tumours. We describe alternative approaches for choosing the most efficient T cells for cancer treatment in terms of their tumour-specificity and phenotype. Finally, we present strategies for improvement of anti-tumour potential of T cells, including combination therapies.
Collapse
Affiliation(s)
- Elżbieta Chruściel
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Zuzanna Urban-Wójciuk
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Łukasz Arcimowicz
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Jacek Kowalski
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
- Department of Pathomorphology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Mateusz Gliwiński
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Tomasz Marjański
- Department of Thoracic Surgery, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (T.M.); (W.R.)
| | - Witold Rzyman
- Department of Thoracic Surgery, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (T.M.); (W.R.)
| | - Wojciech Biernat
- Department of Pathomorphology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Rafał Dziadziuszko
- Department of Oncology and Radiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Carla Montesano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy;
| | - Roberta Bernardini
- Department of Biology and Interdepartmental Center CIMETA, University of Rome "Tor Vergata", 00133 Rome, Italy;
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
35
|
Yang S, Kang Q, Hou Y, Wang L, Li L, Liu S, Liao H, Cao Z, Yang L, Xiao Z. Mutant BCL11B in a Patient With a Neurodevelopmental Disorder and T-Cell Abnormalities. Front Pediatr 2020; 8:544894. [PMID: 33194885 PMCID: PMC7641641 DOI: 10.3389/fped.2020.544894] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/28/2020] [Indexed: 11/13/2022] Open
Abstract
Background: BCL11B encodes B-cell lymphoma/leukemia 11B, a transcription factor that participates in the differentiation and migration of neurons and lymphocyte cells. De novo mutations of BCL11B have been associated with neurodevelopmental disorder and immunodeficiency, such as immunodeficiency 49 (IMD49) and intellectual developmental disorder with speech delay, dysmorphic facies, and T-cell abnormalities (IDDSFTA). However, the pathogenesis of the neurodevelopmental disorder and T-cell deficiency is still mysterious. The strategy to distinguish these two diseases in detail is also unclear. Methods: A patient with unique clinical features was identified. Multiple examinations were applied for evaluation. Whole-exome sequencing (WES) and Sanger sequencing were also performed for the identification of the disease-causing mutation. Results: We reported a 17-month-old girl with intellectual disability, speech impairment, and delay in motor development. She presented with mild dysmorphic facial features and weak functional movement. MRI indicated the abnormal myelination of the white matter. Immunological analysis showed normal levels of RTEs and γδT cells but a deficiency of naive T cells. Genetic sequencing identified a de novo heterozygous frameshift mutation c.1192_1196delAGCCC in BCL11B. Conclusions: An IDDSFTA patient of East Asian origin was reported. The unreported neurological display, immunophenotype, and a novel disease-causing mutation of the patient extended the spectrum of clinical features and genotypes of IDDSFTA.
Collapse
Affiliation(s)
- Sai Yang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Qingyun Kang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | | | - Lili Wang
- Research Institute of Pediatrics, Hunan Children's Hospital, Changsha, China
| | - Liping Li
- Research Institute of Pediatrics, Hunan Children's Hospital, Changsha, China
| | - Shulei Liu
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Hongmei Liao
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | | | - Liming Yang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Zhenghui Xiao
- Department of Neurology, Hunan Children's Hospital, Changsha, China.,Research Institute of Pediatrics, Hunan Children's Hospital, Changsha, China.,Hunan Provincial Key Laboratory of Children's Emergency Medicine, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
36
|
Exploring single-cell data with deep multitasking neural networks. Nat Methods 2019; 16:1139-1145. [PMID: 31591579 PMCID: PMC10164410 DOI: 10.1038/s41592-019-0576-7] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 08/19/2019] [Indexed: 01/22/2023]
Abstract
It is currently challenging to analyze single-cell data consisting of many cells and samples, and to address variations arising from batch effects and different sample preparations. For this purpose, we present SAUCIE, a deep neural network that combines parallelization and scalability offered by neural networks, with the deep representation of data that can be learned by them to perform many single-cell data analysis tasks. Our regularizations (penalties) render features learned in hidden layers of the neural network interpretable. On large, multi-patient datasets, SAUCIE's various hidden layers contain denoised and batch-corrected data, a low-dimensional visualization and unsupervised clustering, as well as other information that can be used to explore the data. We analyze a 180-sample dataset consisting of 11 million T cells from dengue patients in India, measured with mass cytometry. SAUCIE can batch correct and identify cluster-based signatures of acute dengue infection and create a patient manifold, stratifying immune response to dengue.
Collapse
|
37
|
Hong H, Song HK, Hwang ES, Lee AR, Han DS, Kim SE, Oh ES. Up-regulation of syndecan-2 in proximal colon correlates with acute inflammation. FASEB J 2019; 33:11381-11395. [PMID: 31311305 DOI: 10.1096/fj.201900561r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We previously reported that syndecan-2 expression is increased on the colonic epithelium during chronic inflammation. Here, we report that syndecan-2 exhibits a different pattern of site-specific colonic expression during acute inflammation. Syndecan-2 expression was up-regulated predominantly in the proximal colon of dextran sulfate sodium-induced colitis mice. The colitis-associated up-regulation of syndecan-2 was barely detected in Rag-1-/- (recombination activating gene 1 knockout) mice under colitis-inducing conditions. Increased syndecan-2 expression correlated with increased levels of infiltrated CD4+ IL-17A+ T cells in the proximal colon. Serum levels of IL-17A were increased during the acute inflammatory response in normal mice but not Rag-1-/- mice. IL-17A directly induced IL-17 receptor (IL-17RA) and syndecan-2 expression in ex vivo-cultured proximal colon tissues and adenoma cell lines from proximal colon. IL-17RA knockdown reduced the IL-17A-mediated syndecan-2 expression in SNU1235 cells. No elevation of syndecan-2 or IL-17RA was observed in colonic tissues from IL-17A-/- mice during colitis induction. Finally, increased expression of syndecan-2 and IL-17RA was observed in the proximal colons of cecal ligation and puncture-induced sepsis mice and infectious pan colitis patients. Together, these data suggest that acute inflammation induces syndecan-2 expression predominantly in the proximal colon via IL-17A-IL-17RA signaling during the early stage of the inflammatory response and that proximal colonic syndecan-2 might be a biomarker for acute inflammation.-Hong, H., Song, H.-K., Hwang, E. S., Lee, A. R., Han, D. S., Kim, S.-E., Oh, E.-S. Up-regulation of syndecan-2 in proximal colon correlates with acute inflammation.
Collapse
Affiliation(s)
- Heejeong Hong
- Department of Life Sciences, Ewha Womans University, Seoul, South Korea.,Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Hyun-Kuk Song
- Department of Life Sciences, Ewha Womans University, Seoul, South Korea.,Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Eun Sook Hwang
- Department of Pharmacy, Ewha Womans University, Seoul, South Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - A Reum Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Guri, South Korea
| | - Dong Soo Han
- Department of Internal Medicine, Hanyang University College of Medicine, Guri, South Korea
| | - Seong-Eun Kim
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Institute, Seoul, South Korea
| | - Eok-Soo Oh
- Department of Life Sciences, Ewha Womans University, Seoul, South Korea.,Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
38
|
Glatzová D, Cebecauer M. Dual Role of CD4 in Peripheral T Lymphocytes. Front Immunol 2019; 10:618. [PMID: 31001252 PMCID: PMC6454155 DOI: 10.3389/fimmu.2019.00618] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/08/2019] [Indexed: 01/07/2023] Open
Abstract
The interaction of T-cell receptors (TCRs) with self- and non-self-peptides in the major histocompatibility complex (MHC) stimulates crucial signaling events, which in turn can activate T lymphocytes. A variety of accessory molecules further modulate T-cell signaling. Of these, the CD4 and CD8 coreceptors make the most critical contributions to T cell sensitivity in vivo. Whereas, CD4 function in T cell development is well-characterized, its role in peripheral T cells remains incompletely understood. It was originally suggested that CD4 stabilizes weak interactions between TCRs and peptides in the MHC and delivers Lck kinases to that complex. The results of numerous experiments support the latter role, indicating that the CD4-Lck complex accelerates TCR-triggered signaling and controls the availability of the kinase for TCR in the absence of the ligand. On the other hand, extremely low affinity of CD4 for MHC rules out its ability to stabilize the receptor-ligand complex. In this review, we summarize the current knowledge on CD4 in T cells, with a special emphasis on the spatio-temporal organization of early signaling events and the relevance for CD4 function. We further highlight the capacity of CD4 to interact with the MHC in the absence of TCR. It drives the adhesion of T cells to the cells that express the MHC. This process is facilitated by the CD4 accumulation in the tips of microvilli on the surface of unstimulated T cells. Based on these observations, we suggest an alternative model of CD4 role in T-cell activation.
Collapse
Affiliation(s)
- Daniela Glatzová
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czechia
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Marek Cebecauer
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
39
|
Mousset CM, Hobo W, Woestenenk R, Preijers F, Dolstra H, van der Waart AB. Comprehensive Phenotyping of T Cells Using Flow Cytometry. Cytometry A 2019; 95:647-654. [PMID: 30714682 DOI: 10.1002/cyto.a.23724] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 12/15/2022]
Abstract
The T cell compartment can form a powerful defense against extrinsic (e.g., pathogens) and intrinsic danger (e.g., malignant cells). At the same time, specific subsets of T cells control this process to keep the immune system in check and prevent autoimmunity. A wide variety in T cell functionalities exists, which is dependent on the differentiation and maturation state of the T cells. In this review, we report an overview for the identification of CD4+ T-αβ cells (T-helper (Th)1, Th2, Th9, Th17, Th22, and CD4+ regulatory T cells), CD8+ T-αβ cells (cytotoxic T lymphocyte (Tc)1, Tc2, Tc9, Tc17, and CD8+ regulatory T cells), and their additional effector memory status (naïve, stem cell memory, central memory, effector memory, and effector) using flow cytometry. These different subsets can be discriminated based on selective extracellular markers, in combination with intracellular transcription factor and/or cytokine stainings. Additionally, identification of very small subsets, including antigen-specific T cells, and important technical considerations of flow cytometry are discussed. Together, this overview can be used for comprehensive phenotyping of a T cell subset of interest. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Charlotte M Mousset
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob Woestenenk
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank Preijers
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anniek B van der Waart
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Maffucci P, Chavez J, Jurkiw TJ, O’Brien PJ, Abbott JK, Reynolds PR, Worth A, Notarangelo LD, Felgentreff K, Cortes P, Boisson B, Radigan L, Cobat A, Dinakar C, Ehlayel M, Ben-Omran T, Gelfand EW, Casanova JL, Cunningham-Rundles C. Biallelic mutations in DNA ligase 1 underlie a spectrum of immune deficiencies. J Clin Invest 2018; 128:5489-5504. [PMID: 30395541 PMCID: PMC6264644 DOI: 10.1172/jci99629] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/04/2018] [Indexed: 12/30/2022] Open
Abstract
We report the molecular, cellular, and clinical features of 5 patients from 3 kindreds with biallelic mutations in the autosomal LIG1 gene encoding DNA ligase 1. The patients exhibited hypogammaglobulinemia, lymphopenia, increased proportions of circulating γδT cells, and erythrocyte macrocytosis. Clinical severity ranged from a mild antibody deficiency to a combined immunodeficiency requiring hematopoietic stem cell transplantation. Using engineered LIG1-deficient cell lines, we demonstrated chemical and radiation defects associated with the mutant alleles, which variably impaired the DNA repair pathway. We further showed that these LIG1 mutant alleles are amorphic or hypomorphic, and exhibited variably decreased enzymatic activities, which lead to premature release of unligated adenylated DNA. The variability of the LIG1 genotypes in the patients was consistent with that of their immunological and clinical phenotypes. These data suggest that different forms of autosomal recessive, partial DNA ligase 1 deficiency underlie an immunodeficiency of variable severity.
Collapse
Affiliation(s)
- Patrick Maffucci
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
- Graduate School of Biomedical Sciences, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jose Chavez
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
| | - Thomas J. Jurkiw
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick J. O’Brien
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jordan K. Abbott
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Paul R. Reynolds
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Austen Worth
- Department of Pediatric Medicine, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kerstin Felgentreff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Patricia Cortes
- Department of Molecular, Cellular and Biomedical Science, CUNY School of Medicine, City College of New York, New York, New York, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Paris Descartes University, Imagine Institute, Paris, France
| | - Lin Radigan
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
| | - Aurélie Cobat
- Paris Descartes University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
| | - Chitra Dinakar
- Allergy, Asthma & Immunodeficiency, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Mohammad Ehlayel
- Section of Pediatric Allergy-Immunology, Department of Pediatrics, Weill Cornell Medical College, Hamad Medical Corporation, Doha, Qatar
| | - Tawfeg Ben-Omran
- Department of Clinical and Metabolic Genetics, Department of Pediatrics, Weill Cornell Medical College, Hamad Medical Corporation, Doha, Qatar
| | - Erwin W. Gelfand
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Paris Descartes University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris, France
| | - Charlotte Cunningham-Rundles
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
- Graduate School of Biomedical Sciences, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
41
|
Marin AV, Jiménez-Reinoso A, Briones AC, Muñoz-Ruiz M, Aydogmus C, Pasick LJ, Couso J, Mazariegos MS, Alvarez-Prado AF, Blázquez-Moreno A, Cipe FE, Haskologlu S, Dogu F, Morín M, Moreno-Pelayo MA, García-Sánchez F, Gil-Herrera J, Fernández-Malavé E, Reyburn HT, Ramiro AR, Ikinciogullari A, Recio MJ, Regueiro JR, Garcillán B. Primary T-cell immunodeficiency with functional revertant somatic mosaicism in CD247. J Allergy Clin Immunol 2017; 139:347-349.e8. [DOI: 10.1016/j.jaci.2016.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 05/03/2016] [Accepted: 06/02/2016] [Indexed: 01/25/2023]
|
42
|
Christopoulos P, Bukatz D, Kock S, Malkovsky M, Finke J, Fisch P. Improved analysis of TCRγδ variable region expression in humans. J Immunol Methods 2016; 434:66-72. [DOI: 10.1016/j.jim.2016.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/20/2016] [Accepted: 04/20/2016] [Indexed: 01/13/2023]
|
43
|
Kabelitz D, Déchanet-Merville J. Editorial: "Recent Advances in Gamma/Delta T Cell Biology: New Ligands, New Functions, and New Translational Perspectives". Front Immunol 2015; 6:371. [PMID: 26257738 PMCID: PMC4508528 DOI: 10.3389/fimmu.2015.00371] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/06/2015] [Indexed: 01/12/2023] Open
Affiliation(s)
- Dieter Kabelitz
- Institute of Immunology, University of Kiel , Kiel , Germany
| | | |
Collapse
|