1
|
Yu Z, Xu Z, Fu T, Liu S, Cui J, Zhang B, Liang J, Pang C, Ke Y, Wang R, Tang Z, Gao Y, Du B, Feng Y, Zhao H, Xue G, Yan C, Gan L, Feng J, Fan Z, Yang Y, Huang L, Zhao S, Ying S, Gu Q, Yuan J. Parallel comparison of T cell and B cell subpopulations of adenoid hypertrophy and tonsil hypertrophy of children. Nat Commun 2025; 16:3516. [PMID: 40229254 PMCID: PMC11997228 DOI: 10.1038/s41467-025-58094-w] [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: 05/09/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
The adenoids and tonsils are important immune organs of the nasopharynx that often become hypertrophic in childhood because of recurrent pathogen infection. However, the differences in the immune microenvironment of adenoid hypertrophy (AH) and tonsil hypertrophy (TH) are unclear. Here, we show the epidemiological characteristics and peripheral blood cell indices of 1209 pediatric patients (1-15 years old) diagnosed with AH, and find that AH is often accompanied by TH and characterized by specific changes in immune cell types. Single-cell RNA sequencing analysis show that 12 paired AH and TH samples contain large numbers of B, T cells and some exhausted effector memory CD4+ T cells. Compared with matched TH, AH have more naïve B cells and regulatory CD4+ T cells and less plasma B cells. Weaker antigen presentation and more significant immunosuppression are also observed in AH. In contrast, the number and cytotoxicity of cytotoxic CD8+ T cells decrease with AH grade. These findings will help our understanding of the immune response to nasopharyngeal infection.
Collapse
Affiliation(s)
- Zihui Yu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziying Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Tongtong Fu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shiyu Liu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Military supplies and energy quality supervision station of Bejing, Beijing, 100071, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Zhang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Jieqiong Liang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Chong Pang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ruikun Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Zhijie Tang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Yagang Gao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zheng Fan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yang Yang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lijuan Huang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shuo Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Sun Ying
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qinglong Gu
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China.
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
2
|
Huang W, de Vries C, Sharma RK, Wangriatisak K, Chatzidionysiou K, Malmström V, Grönwall C. JAK Inhibitors and B Cell Function: A Comparative Study of Their Impact on Plasma Cell Differentiation, Cytokine Production, and Naïve B Cell Activation. Eur J Immunol 2025; 55:e202451437. [PMID: 40098342 PMCID: PMC11914861 DOI: 10.1002/eji.202451437] [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: 08/05/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025]
Abstract
B cells play a crucial role in autoimmune diseases, as evidenced by autoantibody responses and the effectiveness of B cell-targeted therapies. Janus kinase inhibitors (JAKi), which target downstream signaling of cytokine receptors, are potent rheumatic disease-modifying drugs. However, besides reducing inflammation, JAKi may impact the adaptive immune system. In this study, we examined the effects of JAKi on B-cell function using in vitro cultures and multiparameter flow cytometry. The results show a JAKi-mediated reduction in plasma cell differentiation, primarily by inhibition of memory B-cell stimulation and proliferation. JAKi exposure resulted in stalling R848, IL-2, and IL-21 stimulated B cells in an intermediate activated state with elevated naïve cells displaying increased expression of CXCR5, CD71, CD22, and CD20. In addition, the data demonstrate a moderate JAKi-mediated reduction of B cell TNF and IL-8 cytokine expression following stimulation. Importantly, the efficacy varied greatly between drugs; tofacitinib and upadacitinib (pan JAKi; JAK1i) exhibited the strongest impact, while baricitinib (JAK1/JAK2i) showed donor-dependent variation, and filgotinib (JAK1i) had no effect. All JAKi, except filgotinib, inhibited IL-2 or IL-21-induced STAT3 phosphorylation. Still, filgotinib demonstrated similar inhibition of phospho-STAT5 as other JAKi following IL-21. These findings underscore the therapeutic impact of JAKi through the modulation of B-cell functions.
Collapse
Affiliation(s)
- Wenqi Huang
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Charlotte de Vries
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Immunology and Rheumatology, All India Institute of Medical Sciences, Bilaspur, India
| | - Kittikorn Wangriatisak
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Katerina Chatzidionysiou
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Grönwall
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Krause R, Ogongo P, Tezera L, Ahmed M, Mbano I, Chambers M, Ngoepe A, Magnoumba M, Muema D, Karim F, Khan K, Lumamba K, Nargan K, Madansein R, Steyn A, Shalek AK, Elkington P, Leslie A. B cell heterogeneity in human tuberculosis highlights compartment-specific phenotype and functional roles. Commun Biol 2024; 7:584. [PMID: 38755239 PMCID: PMC11099031 DOI: 10.1038/s42003-024-06282-7] [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: 10/16/2023] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
B cells are important in tuberculosis (TB) immunity, but their role in the human lung is understudied. Here, we characterize B cells from lung tissue and matched blood of patients with TB and found they are decreased in the blood and increased in the lungs, consistent with recruitment to infected tissue, where they are located in granuloma associated lymphoid tissue. Flow cytometry and transcriptomics identify multiple B cell populations in the lung, including those associated with tissue resident memory, germinal centers, antibody secretion, proinflammatory atypical B cells, and regulatory B cells, some of which are expanded in TB disease. Additionally, TB lungs contain high levels of Mtb-reactive antibodies, specifically IgM, which promotes Mtb phagocytosis. Overall, these data reveal the presence of functionally diverse B cell subsets in the lungs of patients with TB and suggest several potential localized roles that may represent a target for interventions to promote immunity or mitigate immunopathology.
Collapse
Affiliation(s)
- Robert Krause
- Africa Health Research Institute, Durban, South Africa.
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Paul Ogongo
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Institute of Primate Research, National Museums of Kenya, Nairobi, Kenya
| | - Liku Tezera
- National Institute for Health Research Southampton Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- Division of Infection and Immunity, University College London, London, UK
| | - Mohammed Ahmed
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ian Mbano
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mark Chambers
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | - Magalli Magnoumba
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Daniel Muema
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Farina Karim
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Khadija Khan
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | | | - Rajhmun Madansein
- Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Adrie Steyn
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for AIDS Research and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alex K Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paul Elkington
- National Institute for Health Research Southampton Biomedical Research Centre, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Al Leslie
- Africa Health Research Institute, Durban, South Africa.
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
4
|
Flippot R, Teixeira M, Rey-Cardenas M, Carril-Ajuria L, Rainho L, Naoun N, Jouniaux JM, Boselli L, Naigeon M, Danlos FX, Escudier B, Scoazec JY, Cassard L, Albiges L, Chaput N. B cells and the coordination of immune checkpoint inhibitor response in patients with solid tumors. J Immunother Cancer 2024; 12:e008636. [PMID: 38631710 PMCID: PMC11029261 DOI: 10.1136/jitc-2023-008636] [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] [Accepted: 03/31/2024] [Indexed: 04/19/2024] Open
Abstract
Immunotherapy profoundly changed the landscape of cancer therapy by providing long-lasting responses in subsets of patients and is now the standard of care in several solid tumor types. However, immunotherapy activity beyond conventional immune checkpoint inhibition is plateauing, and biomarkers are overall lacking to guide treatment selection. Most studies have focused on T cell engagement and response, but there is a growing evidence that B cells may be key players in the establishment of an organized immune response, notably through tertiary lymphoid structures. Mechanisms of B cell response include antibody-dependent cellular cytotoxicity and phagocytosis, promotion of CD4+ and CD8+ T cell activation, maintenance of antitumor immune memory. In several solid tumor types, higher levels of B cells, specific B cell subpopulations, or the presence of tertiary lymphoid structures have been associated with improved outcomes on immune checkpoint inhibitors. The fate of B cell subpopulations may be widely influenced by the cytokine milieu, with versatile roles for B-specific cytokines B cell activating factor and B cell attracting chemokine-1/CXCL13, and a master regulatory role for IL-10. Roles of B cell-specific immune checkpoints such as TIM-1 are emerging and could represent potential therapeutic targets. Overall, the expanding field of B cells in solid tumors of holds promise for the improvement of current immunotherapy strategies and patient selection.
Collapse
Affiliation(s)
- Ronan Flippot
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Marcus Teixeira
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Macarena Rey-Cardenas
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Lucia Carril-Ajuria
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
- Medical Oncology, CHU Brugmann, Brussels, Belgium
| | - Larissa Rainho
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Natacha Naoun
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Jean-Mehdi Jouniaux
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Lisa Boselli
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Marie Naigeon
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Francois-Xavier Danlos
- LRTI, INSERM U1015, Gustave Roussy, Villejuif, France
- Drug Development Department, Gustave Roussy, Villejuif, France
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | | | - Lydie Cassard
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Nathalie Chaput
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
5
|
Korosec CS, Farhang-Sardroodi S, Dick DW, Gholami S, Ghaemi MS, Moyles IR, Craig M, Ooi HK, Heffernan JM. Long-term durability of immune responses to the BNT162b2 and mRNA-1273 vaccines based on dosage, age and sex. Sci Rep 2022; 12:21232. [PMID: 36481777 PMCID: PMC9732004 DOI: 10.1038/s41598-022-25134-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
The lipid nanoparticle (LNP)-formulated mRNA vaccines BNT162b2 and mRNA-1273 are a widely adopted multi vaccination public health strategy to manage the COVID-19 pandemic. Clinical trial data has described the immunogenicity of the vaccine, albeit within a limited study time frame. Here, we use a within-host mathematical model for LNP-formulated mRNA vaccines, informed by available clinical trial data from 2020 to September 2021, to project a longer term understanding of immunity as a function of vaccine type, dosage amount, age, and sex. We estimate that two standard doses of either mRNA-1273 or BNT162b2, with dosage times separated by the company-mandated intervals, results in individuals losing more than 99% humoral immunity relative to peak immunity by 8 months following the second dose. We predict that within an 8 month period following dose two (corresponding to the original CDC time-frame for administration of a third dose), there exists a period of time longer than 1 month where an individual has lost more than 99% humoral immunity relative to peak immunity, regardless of which vaccine was administered. We further find that age has a strong influence in maintaining humoral immunity; by 8 months following dose two we predict that individuals aged 18-55 have a four-fold humoral advantage compared to aged 56-70 and 70+ individuals. We find that sex has little effect on the immune response and long-term IgG counts. Finally, we find that humoral immunity generated from two low doses of mRNA-1273 decays at a substantially slower rate relative to peak immunity gained compared to two standard doses of either mRNA-1273 or BNT162b2. Our predictions highlight the importance of the recommended third booster dose in order to maintain elevated levels of antibodies.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| | - Suzan Farhang-Sardroodi
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Department of Mathematics, University of Manitoba, 186 Dysart Road, Winnipeg, MB, R3T 2N2, Canada
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Sameneh Gholami
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal & Sainte-Justine University Hospital Research Centre, 3175, ch. Côte Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
6
|
Spear S, Candido JB, McDermott JR, Ghirelli C, Maniati E, Beers SA, Balkwill FR, Kocher HM, Capasso M. Discrepancies in the Tumor Microenvironment of Spontaneous and Orthotopic Murine Models of Pancreatic Cancer Uncover a New Immunostimulatory Phenotype for B Cells. Front Immunol 2019; 10:542. [PMID: 30972056 PMCID: PMC6445859 DOI: 10.3389/fimmu.2019.00542] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/28/2019] [Indexed: 12/25/2022] Open
Abstract
B cells are salient features of pancreatic ductal adenocarcinoma (PDAC) tumors, yet their role in this disease remains controversial. Murine studies have indicated a protumoral role for B cells, whereas clinical data show tumor-infiltrating B cells are a positive prognostic factor, both in PDAC and other cancers. This disparity needs to be clarified in order to develop effective immunotherapies. In this study, we provide new evidence that reconcile human and mouse data and highlight the importance of using relevant preclinical tumor models when assessing B cell function. We compared B cell infiltration and activation in both a genetic model of murine PDAC (KPC mouse) and an injectable orthotopic model. A pronounced B cell infiltrate was only observed in KPC tumors and correlated with T cell infiltration, mirroring human disease. In contrast, orthotopic tumors exhibited a relative paucity of B cells. Accordingly, KPC-derived B cells displayed markers of B cell activation (germinal center entry, B cell memory, and plasma cell differentiation) accompanied by significant intratumoral immunoglobulin deposition, a feature markedly weaker in orthotopic tumors. Tumor immunoglobulins, however, did not appear to form immune complexes. Furthermore, in contrast to the current paradigm that tumor B cells are immunosuppressive, when assessed as a bulk population, intratumoral B cells upregulated several proinflammatory and immunostimulatory genes, a distinctly different phenotype to that of splenic-derived B cells; further highlighting the importance of studying tumor-infiltrating B cells over B cells from secondary lymphoid organs. In agreement with the current literature, genetic deletion of B cells (μMT mice) resulted in reduced orthotopic tumor growth, however, this was not recapitulated by treatment with B-cell-depleting anti-CD20 antibody and, more importantly, was not observed in anti-CD20-treated KPC mice. This suggests the result from B cell deficient mice might be caused by their altered immune system, rather than lack of B cells. Therefore, our data indicate B cells do not favor tumor progression. In conclusion, our analysis of relevant preclinical models shows B cells to be active members of the tumor microenvironment, producing immunostimulatory factors that might support the adaptive antitumor immune response, as suggested by human PDAC studies.
Collapse
Affiliation(s)
- Sarah Spear
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Juliana B. Candido
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jacqueline R. McDermott
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- Department of Pathology, University College London Hospital, London, United Kingdom
| | - Cristina Ghirelli
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Eleni Maniati
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Frances R. Balkwill
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Hemant M. Kocher
- Centre for Tumor Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Melania Capasso
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
7
|
Hähnlein JS, Ramwadhdoebe TH, Semmelink JF, Choi IY, Berger FH, Maas M, Gerlag DM, Tak PP, Geijtenbeek TBH, van Baarsen LGM. Distinctive expression of T cell guiding molecules in human autoimmune lymph node stromal cells upon TLR3 triggering. Sci Rep 2018; 8:1736. [PMID: 29379035 PMCID: PMC5789053 DOI: 10.1038/s41598-018-19951-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/09/2018] [Indexed: 12/22/2022] Open
Abstract
Infections are implicated in autoimmunity. Autoantibodies are produced in lymphoid tissue where lymph node stromal cells (LNSCs) regulate lymphocyte function. Infections can alter the interaction between LNSCs and lymphocytes resulting in defective immune responses. In rheumatoid arthritis (RA) autoantibody production precedes clinical disease allowing identification of at risk individuals. We investigated the ability of human LNSCs derived from RA, RA-risk and healthy individuals to sense and respond to pathogens. Human LNSCs cultured directly from freshly collected lymph node biopsies expressed TLR1-9 with exception of TLR7. In all donors TLR3 triggering induced expression of ISGs, IL-6 and adhesion molecules like VCAM-1 and ICAM-1. Strikingly, T cell guiding chemokines CCL19 and IL-8 as well as the antiviral gene MxA were less induced upon TLR3 triggering in autoimmune LNSCs. This observed decrease, found already in LNSCs of RA-risk individuals, may lead to incorrect positioning of lymphocytes and aberrant immune responses during viral infections.
Collapse
Affiliation(s)
- Janine S Hähnlein
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara H Ramwadhdoebe
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johanna F Semmelink
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ivy Y Choi
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ferco H Berger
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mario Maas
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Danielle M Gerlag
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, UK
| | - Paul P Tak
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Ghent University, Ghent, Belgium
- University of Cambridge, Cambridge, UK
- GlaxoSmithKline, Stevenage, UK
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lisa G M van Baarsen
- Amsterdam Rheumatology & immunology Center (ARC), Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Das S, Li J, Hirano M, Sutoh Y, Herrin BR, Cooper MD. Evolution of two prototypic T cell lineages. Cell Immunol 2015; 296:87-94. [PMID: 25958271 DOI: 10.1016/j.cellimm.2015.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/14/2015] [Accepted: 04/24/2015] [Indexed: 01/06/2023]
Abstract
Jawless vertebrates, which occupy a unique position in chordate phylogeny, employ leucine-rich repeat (LRR)-based variable lymphocyte receptors (VLR) for antigen recognition. During the assembly of the VLR genes (VLRA, VLRB and VLRC), donor LRR-encoding sequences are copied in a step-wise manner into the incomplete germ-line genes. The assembled VLR genes are differentially expressed by discrete lymphocyte lineages: VLRA- and VLRC-producing cells are T-cell like, whereas VLRB-producing cells are B-cell like. VLRA(+) and VLRC(+) lymphocytes resemble the two principal T-cell lineages of jawed vertebrates that express the αβ or γδ T-cell receptors (TCR). Reminiscent of the interspersed nature of the TCRα/TCRδ locus in jawed vertebrates, the close proximity of the VLRA and VLRC loci facilitates sharing of donor LRR sequences during VLRA and VLRC assembly. Here we discuss the insight these findings provide into vertebrate T- and B-cell evolution, and the alternative types of anticipatory receptors they use for adaptive immunity.
Collapse
Affiliation(s)
- Sabyasachi Das
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, GA 30322, United States
| | - Jianxu Li
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, GA 30322, United States
| | - Masayuki Hirano
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, GA 30322, United States
| | - Yoichi Sutoh
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, GA 30322, United States
| | - Brantley R Herrin
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, GA 30322, United States
| | - Max D Cooper
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, GA 30322, United States.
| |
Collapse
|
9
|
Sigurdardottir SL, Thorleifsdottir RH, Guzman AM, Gudmundsson GH, Valdimarsson H, Johnston A. The anti-microbial peptide LL-37 modulates immune responses in the palatine tonsils where it is exclusively expressed by neutrophils and a subset of dendritic cells. Clin Immunol 2012; 142:139-49. [DOI: 10.1016/j.clim.2011.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 09/26/2011] [Accepted: 09/29/2011] [Indexed: 11/24/2022]
|
10
|
Saha NR, Smith J, Amemiya CT. Evolution of adaptive immune recognition in jawless vertebrates. Semin Immunol 2010; 22:25-33. [PMID: 20056434 DOI: 10.1016/j.smim.2009.12.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 12/04/2009] [Indexed: 11/18/2022]
Abstract
All extant vertebrates possess an adaptive immune system wherein diverse immune receptors are created and deployed in specialized blood cell lineages. Recent advances in DNA sequencing and developmental resources for basal vertebrates have facilitated numerous comparative analyses that have shed new light on the molecular and cellular bases of immune defense and the mechanisms of immune receptor diversification in the "jawless" vertebrates. With data from these key species in hand, it is becoming possible to infer some general aspects of the early evolution of vertebrate adaptive immunity. All jawed vertebrates assemble their antigen-receptor genes through combinatorial recombination of different "diversity" segments into immunoglobulin or T-cell receptor genes. However, the jawless vertebrates employ an analogous, but independently derived set of immune receptors in order to recognize and bind antigens: the variable lymphocyte receptors (VLRs). The means by which this locus generates receptor diversity and achieves antigen specificity is of considerable interest because these mechanisms represent a completely independent strategy for building a large immune repertoire. Therefore, studies of the VLR system are providing insight into the fundamental principles and evolutionary potential of adaptive immune recognition systems. Here we review and synthesize the wealth of data that have been generated towards understanding the evolution of the adaptive immune system in the jawless vertebrates.
Collapse
Affiliation(s)
- Nil Ratan Saha
- Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA.
| | | | | |
Collapse
|
11
|
Noronha AM, Liang Y, Hetzel JT, Hasturk H, Kantarci A, Stucchi A, Zhang Y, Nikolajczyk BS, Farraye FA, Ganley-Leal LM. Hyperactivated B cells in human inflammatory bowel disease. J Leukoc Biol 2009; 86:1007-16. [PMID: 19589946 DOI: 10.1189/jlb.0309203] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IBD is characterized by a chronic, dysregulated immune response to intestinal bacteria. Past work has focused on the role of T cells and myeloid cells in mediating chronic gastrointestinal and systemic inflammation. Here, we show that circulating and tissue B cells from CD patients demonstrate elevated basal levels of activation. CD patient B cells express surface TLR2, spontaneously secrete high levels of IL-8, and contain increased ex vivo levels of phosphorylated signaling proteins. CD clinical activity correlates directly with B cell expression of IL-8 and TLR2, suggesting a positive relationship between these B cell inflammatory mediators and disease pathogenesis. In contrast, B cells from UC patients express TLR2 but generally do not demonstrate spontaneous IL-8 secretion; however, significant IL-8 production is inducible via TLR2 stimulation. Furthermore, UC clinical activity correlates inversely with levels of circulating TLR2+ B cells, which is opposite to the association observed in CD. In conclusion, TLR2+ B cells are associated with clinical measures of disease activity and differentially associated with CD- and UC-specific patterns of inflammatory mediators, suggesting a formerly unappreciated role of B cells in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Ansu Mammen Noronha
- Section of Infectious Disease, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Guo P, Hirano M, Herrin BR, Li J, Yu C, Sadlonova A, Cooper MD. Dual nature of the adaptive immune system in lampreys. Nature 2009; 459:796-801. [PMID: 19474790 PMCID: PMC2714547 DOI: 10.1038/nature08068] [Citation(s) in RCA: 248] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Accepted: 04/14/2009] [Indexed: 12/31/2022]
Abstract
Jawless vertebrates use variable lymphocyte receptors (VLR) comprised of leucine-rich-repeat (LRR) segments as counterparts of the immunoglobulin-based receptors that jawed vertebrates use for antigen recognition. Highly diverse VLR genes are somatically assembled by the insertion of variable LRR sequences into incomplete germline VLRA and VLRB genes. Here we show that in sea lampreys (Petromyzon marinus) VLRA and VLRB anticipatory receptors are expressed by separate lymphocyte populations by monoallelic VLRA or VLRB assembly, together with expression of cytosine deaminase 1 (CDA1) or 2 (CDA2), respectively. Distinctive gene expression profiles for VLRA(+) and VLRB(+) lymphocytes resemble those of mammalian T and B cells. Although both the VLRA and the VLRB cells proliferate in response to antigenic stimulation, only the VLRB lymphocytes bind native antigens and differentiate into VLR antibody-secreting cells. Conversely, VLRA lymphocytes respond preferentially to a classical T-cell mitogen and upregulate the expression of the pro-inflammatory cytokine genes interleukin-17 (IL-17) and macrophage migration inhibitory factor (MIF). The finding of T-like and B-like lymphocytes in lampreys offers new insight into the evolution of adaptive immunity.
Collapse
Affiliation(s)
- Peng Guo
- Emory Vaccine Center and Department of Pathology and Laboratory Medicine, Emory University, 1462 Clifton Road North-East, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Holm BC, Svensson J, Akesson C, Arvastsson J, Ljungberg J, Lynch K, Ivarsson SA, Lernmark A, Cilio CM. Evidence for immunological priming and increased frequency of CD4+ CD25+ cord blood T cells in children born to mothers with type 1 diabetes. Clin Exp Immunol 2007; 146:493-502. [PMID: 17100770 PMCID: PMC1810404 DOI: 10.1111/j.1365-2249.2006.03243.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Maternal transmission of islet autoantibodies to children born to mothers with type 1 diabetes (T1D) has been shown to protect from autoantibodies and diabetes development later in life. However, the factors conferring disease protection are poorly understood. The aim of this study was to evaluate comparatively proinflammatory cytokines, autoantibodies and lymphocyte subsets in cord blood (CB) of children born to mothers with either T1D (n = 13), gestational diabetes (GDM) (n = 32) or healthy mothers (n = 81) in relation to transplacental passage of autoantibodies. The results are consistent with early priming of the fetal immune system only in children born to mothers with T1D. Levels of interleukin (IL)-1beta (P = 0.022), tumour necrosis factor (TNF)-alpha (P = 0.002) and IL-8 (P = 0.0012), as well as the frequency of CD4(+) CD25(+) T cells (P < 0.01) were significantly increased, and the increased levels correlated positively with anti-GAD65 autoantibody (GADA) levels. Moreover, CD4(+) CD25(+) T cells of children born to T1D mothers exhibited a more pronounced memory phenotype with increased CCR4 expression and down-regulation of CD62L. These data suggest that early activation of the fetal immune system as a consequence of maternal autoimmunity and transplacental passage of GADA may influence the generation and expansion of fetal regulatory T cells. This might induce an early antigen-specific immunological tolerance that could protect against T1D later in life.
Collapse
Affiliation(s)
- B C Holm
- Cellular Autoimmunity Unit, Malmö University Hospital, Lund University Hospital, Lund University, Malmö, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Teleshova N, Kenney J, Van Nest G, Marshall J, Lifson JD, Sivin I, Dufour J, Bohm R, Gettie A, Robbiani M. Local and systemic effects of intranodally injected CpG-C immunostimulatory-oligodeoxyribonucleotides in macaques. THE JOURNAL OF IMMUNOLOGY 2007; 177:8531-41. [PMID: 17142751 DOI: 10.4049/jimmunol.177.12.8531] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunostimulatory CpG-C oligodeoxyribonucleotides (ISS-ODNs) represent a promising strategy to enhance vaccine efficacy. We have shown that the CpG-C ISS-ODN C274 stimulates macaque blood dendritic cells (DCs) and B cells and augments SIV-specific IFN-gamma responses in vitro. To further explore the potential of C274 for future vaccine studies, we assessed the in vivo effects of locally administered C274 (in naive and healthy infected macaques). Costimulatory molecules were marginally increased on DCs and B cells within cells isolated from C274-injected lymph nodes (LNs). However, cells from C274-injected LNs exhibited heightened responsiveness to in vitro culture. This was particularly apparent at the level of CD80 (less so CD86) expression by CD123(+) plasmacytoid DCs and was further boosted in the presence of additional C274 in vitro. Notably, cells from C274-injected LNs secreted significantly elevated levels of several cytokines and chemokines upon in vitro culture. This was more pronounced when cells were exposed to additional stimuli in vitro, producing IFN-alpha, IL-3, IL-6, IL-12, TNF-alpha, CCL2, CCL3, CCL5, and CXCL8. Following C274 administration in the absence of additional SIV Ag, endogenous IFN-gamma secretion was elevated in LN cells of infected animals, but SIV-specific responses were unchanged. Endogenous and SIV-specific responses decreased in blood, before the SIV-specific responses rebounded by 2 wk after C274 treatment. Elevated IFN-alpha, CCL2, and CCL5 were also detected in the plasma after C274 injection. Thus, locally administered C274 has local and systemic activities, supporting the potential for CpG-C ISS-ODNs to boost immune function to enhance anti-HIV vaccine immunogenicity.
Collapse
Affiliation(s)
- Natalia Teleshova
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ganley-Leal LM, Liu X, Wetzler LM. Toll-like receptor 2-mediated human B cell differentiation. Clin Immunol 2006; 120:272-84. [PMID: 16766226 DOI: 10.1016/j.clim.2006.04.571] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 04/12/2006] [Accepted: 04/20/2006] [Indexed: 10/24/2022]
Abstract
Human B cells likely have a major role in the adjuvant activity of Toll-like receptor (TLR) 9 agonists by enhancing innate and adaptive immune responses. As several TLR2 ligands are promising vaccine adjuvant candidates, our aim was to characterize the effects of TLR2 stimulation on human B cell activation and differentiation using cells derived from healthy peripheral blood (PB), spleen, and diseased tonsils. We found a subset of partially differentiated TLR2+ PB and splenic B cells which responds to TLR2 agonists by mediating events involved in germinal center formation, such as upregulating CD77 and secreting chemokines. Furthermore, we show that TLR2-activated monocytes induce B cells to secrete significant quantities of IgM. Finally, activated TLR2+ B cells from tonsils are induced to secrete IgM directly by TLR2 ligands. Thus, TLR2 is likely involved in specific B cell-mediated functions and may be a viable vaccine adjuvant target in humans.
Collapse
Affiliation(s)
- Lisa M Ganley-Leal
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Evans Biomedical Research Center, 650 Albany Street, Boston, MA 02118, USA.
| | | | | |
Collapse
|
16
|
Teleshova N, Kenney J, Williams V, Van Nest G, Marshall J, Lifson JD, Sivin I, Dufour J, Bohm R, Gettie A, Pope M. CpG-C ISS-ODN activation of blood-derived B cells from healthy and chronic immunodeficiency virus-infected macaques. J Leukoc Biol 2006; 79:257-67. [PMID: 16443827 DOI: 10.1189/jlb.0205084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cytosine-phosphate-guanine class C (CpG-C) immunostimulatory sequence oligodeoxynucleotides (ISS-ODNs) activate human B cells and dendritic cells (DCs), properties that suggest potential use as a novel adjuvant to enhance vaccine efficacy. After demonstrating that the CpG-C ISS-ODN C274 activates macaque DCs, we examined in vitro activation of macaque B cells by C274 as a prelude to evaluation of this molecule as an adjuvant in the testing of candidate human immunodeficiency virus vaccines in the rhesus macaque-simian immunodeficiency virus (SIV) model. C274 induced macaque CD20(+) B cells to proliferate more strongly than CD40 ligand or CpG-B ISS-ODN. C274 enhanced B cell survival; increased viability was most evident after 3-7 days of culture. Increased expression of CD40, CD80, and CD86 by B cells was apparent within 24 h of exposure to C274 and persisted for up to 1 week. C274-stimulated, B cell-enriched and peripheral blood mononuclear cell suspensions from naïve and immunodeficiency virus-infected monkeys secreted several cytokines [e.g., interleukin (IL)-3, IL-6, IL-12, interferon-alpha] and chemokines [e.g., monocyte chemoattractant protein-1/CC chemokine ligand 2 (CCL2), macrophage-inflammatory protein-1alpha/CCL3, IL-8/CXC chemokine ligand 8]. In comparison, exposure of macaque B cells to SIV had minimal impact on surface phenotype, despite inducing cytokine and chemokine production in cells from infected and uninfected animals. These observations emphasize the need to identify strategies to optimally boost immune function, as immunodeficiency viruses themselves only partially activate B cells and DCs. The ability of C274 to stimulate B cells and DCs in healthy and infected monkeys suggests its possible use as a broad-acting adjuvant to be applied in the rhesus macaque model for the development of preventative and therapeutic vaccines.
Collapse
Affiliation(s)
- N Teleshova
- Population Council, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ek S, Björck E, Högerkorp CM, Nordenskjöld M, Porwit-MacDonald A, Borrebaeck CAK. Mantle cell lymphomas acquire increased expression of CCL4, CCL5 and 4-1BB-L implicated in cell survival. Int J Cancer 2006; 118:2092-7. [PMID: 16287062 DOI: 10.1002/ijc.21579] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We have analyzed mantle cell lymphomas (MCLs), using high-density DNA microarrays, and confirmed the expression of differentially regulated antigens, using flow cytometry and immunohistochemistry. The results show that MCLs acquire expression of molecules that normally are involved in interaction with other immune cells and, thus, might affect the ability of the tumor to survive. The MCL signature is represented by the overexpression of the chemokine CCL4 (MIP-1beta), implicated in the recruitment of regulatory T cells, as well as CCL5 and 4-1BB-L. The latter molecules are normally involved in chemotaxis of T cells and B cell activation, respectively. Signaling through 4-1BB-L allows B cells to proliferate and the expression of its ligand, by the intra-tumoral mesh of follicular dendritic cells (FDC), could thus serve as a paracrine loop facilitating growth and survival of MCL cells.
Collapse
Affiliation(s)
- Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | | | | | | |
Collapse
|
18
|
Sims-Mourtada JC, Bruce S, McKeller MR, Rangel R, Guzman-Rojas L, Cain K, Lopez C, Zimonjic DB, Popescu NC, Gordon J, Wilkinson MF, Martinez-Valdez H. The human AKNA gene expresses multiple transcripts and protein isoforms as a result of alternative promoter usage, splicing, and polyadenylation. DNA Cell Biol 2005; 24:325-38. [PMID: 15869410 DOI: 10.1089/dna.2005.24.325] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
We previously showed that the human AKNA gene encodes an AT-hook transcription factor that regulates the expression of costimulatory cell surface molecules on lymphocytes. However, AKNA cDNA probes hybridize with multiple transcripts, suggesting either the existence of other homologous genes or a complex regulation operating on a single gene. Here we report evidence for the latter, as we find that AKNA is encoded by a single gene that spans a 61-kb locus of 24 exons on the fragile FRA9E region of human chromosome 9q32. This gene gives rise to at least nine distinct transcripts, most of which are expressed in a tissue-specific manner in lymphoid organs. Many of the AKNA transcripts originate from alternative splicing; others appear to derive from differential polyadenylation and promoter usage. The alternative AKNA transcripts are predicted to encode overlapping protein isoforms, some of which (p70 and p100) are readily detectable using a polyclonal anti-AKNA antisera that we generated. We also find that AKNA PEST-dependent cleavage into p50 polypeptides is targeted to mature B cells and appears to be required for CD40 upregulation. The unusual capacity of the AKNA gene to generate multiple transcripts and proteins may reflect its functional diversity, and it may also provide a fail-safe mechanism that preserves AKNA expression.
Collapse
Affiliation(s)
- Jennifer C Sims-Mourtada
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kwakkenbos MJ, Pouwels W, Matmati M, Stacey M, Lin HH, Gordon S, van Lier RAW, Hamann J. Expression of the largest CD97 and EMR2 isoforms on leukocytes facilitates a specific interaction with chondroitin sulfate on B cells. J Leukoc Biol 2004; 77:112-9. [PMID: 15498814 DOI: 10.1189/jlb.0704402] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The EGF-TM7 receptors CD97 and EMR2 are heptahelical molecules predominantly expressed on leukocytes. A characteristic of these receptors is their ability to interact with cellular ligands via the N-terminal epidermal growth factor (EGF)-like domains. The first two EGF domains of CD97 (but not EMR2) bind CD55 (decay-accelerating factor), while the fourth EGF domain of both CD97 and EMR2 interacts with the glycosaminoglycan chondroitin sulfate (CS). Using fluorescent beads coated with soluble recombinant CD97 and EMR2 protein, and isoform-specific monoclonal antibodies, we have determined the cellular and molecular characteristics of the interaction with CS. The fourth EGF domain of CD97 and EMR2 is expressed on activated lymphocytes and myeloid cells, whereas the ligand is specifically found on B cells within the peripheral blood. The interaction between CD97/EMR2 and CS may therefore play a role in the interaction of activated T cells, dendritic cells, and macrophages with B cells.
Collapse
Affiliation(s)
- Mark J Kwakkenbos
- Laboratory of Expermential Immunology, Academic Medical Centre, University of Amsterdam, PO Box 22700, 1100 DE Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Kluger Y, Tuck DP, Chang JT, Nakayama Y, Poddar R, Kohya N, Lian Z, Ben Nasr A, Halaban HR, Krause DS, Zhang X, Newburger PE, Weissman SM. Lineage specificity of gene expression patterns. Proc Natl Acad Sci U S A 2004; 101:6508-13. [PMID: 15096607 PMCID: PMC404075 DOI: 10.1073/pnas.0401136101] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The hematopoietic system offers many advantages as a model for understanding general aspects of lineage choice and specification. Using oligonucleotide microarrays, we compared gene expression patterns of multiple purified hematopoietic cell populations, including neutrophils, monocytes, macrophages, resting, centrocytic, and centroblastic B lymphocytes, dendritic cells, and hematopoietic stem cells. Some of these cells were studied under both resting and stimulated conditions. We studied the collective behavior of subsets of genes derived from the Biocarta database of functional pathways, hand-tuned groupings of genes into broad functional categories based on the Gene Ontology database, and the metabolic pathways in the Kyoto Encyclopedia of Genes and Genomes database. Principal component analysis revealed strikingly pervasive differences in relative levels of gene expression among cell lineages that involve most of the subsets examined. These results indicate that many processes in these cells behave differently in different lineages. Much of the variation among lineages was captured by the first few principal components. Principal components biplots were found to provide a convenient visual display of the contributions of the various genes within the subsets in lineage discrimination. Moreover, by applying tree-constructing methodologies borrowed from phylogenetics to the expression data from differentiated cells and stem cells, we reconstructed a tree of relationships that resembled the established hematopoietic program of lineage development. Thus, the mRNA expression data implicitly contained information about developmental relationships among cell types.
Collapse
Affiliation(s)
- Yuval Kluger
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|