1
|
Wang W, Dong G, Yang Z, Li S, Li J, Wang L, Zhu Q, Wang Y. Single-cell analysis of tumor microenvironment and cell adhesion reveals that interleukin-1 beta promotes cancer cell proliferation in breast cancer. Animal Model Exp Med 2024; 7:617-625. [PMID: 38860503 PMCID: PMC11528385 DOI: 10.1002/ame2.12445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC), which is so called because of the lack of estrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor receptor 2 (HER2) receptors on the cancer cells, accounts for 10%-15% of all breast cancers. The heterogeneity of the tumor microenvironment is high. However, the role of plasma cells controlling the tumor migration progression in TNBC is still not fully understood. METHODS We analyzed single-cell RNA sequencing data from five HER2 positive, 12 ER positive/PR positive, and nine TNBC samples. The potential targets were validated by immunohistochemistry. RESULTS Plasma cells were enriched in TNBC samples, which was consistent with validation using data from The Cancer Genome Atlas. Cell communication analysis revealed that plasma cells interact with T cells through the intercellular adhesion molecule 2-integrin-aLb2 complex, and then release interleukin 1 beta (IL1B), as verified by immunohistochemistry, ultimately promoting tumor growth. CONCLUSION Our results revealed the role of plasma cells in TNBC and identified IL1B as a new prognostic marker for TNBC.
Collapse
Affiliation(s)
- Wenyan Wang
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Ziguo Yang
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Shaoxiang Li
- Department of Pathology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Jia Li
- Department of Pathology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lin Wang
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qiang Zhu
- Department of General Surgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yuchen Wang
- Department of PharmacologyInstitute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Lu Y, Yao Y, Zhai S, Ni F, Wang J, Chen F, Zhang Y, Li H, Hu H, Zhang H, Yu B, Chen H, Huang X, Ding W, Lu D. The role of immune cell signatures in the pathogenesis of ovarian-related diseases: a causal inference based on Mendelian randomization. Int J Surg 2024; 110:6541-6550. [PMID: 38884272 PMCID: PMC11486969 DOI: 10.1097/js9.0000000000001814] [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: 05/04/2024] [Accepted: 05/19/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Immune cells play a pivotal role in maintaining ovarian function. However, the specific contributions of different immune cell phenotypes to the pathogenesis of specific ovarian-related diseases remain poorly understood. The authors aim to investigate the correlation between 731 immunophenotypes and ovarian-related diseases. MATERIALS AND METHODS Utilizing publicly available genetic data, the authors undertook a series of quality control measures to identify instrumental variables associated with exposure. Subsequently, we conducted two-sample Mendelian randomization (MR) using inverse variance weighting to explore the causal relationships between 731 immune cell features and six ovarian-related diseases: ovarian cysts, ovarian dysfunction, premature ovarian failure (POF), polycystic ovary syndrome (PCOS), benign neoplasm of ovary, and malignant neoplasm of ovary at the genetic level. Sensitivity analyses, including leave-one-out and other MR analysis models, were performed. Finally, Bayesian colocalization (COLOC) analysis was employed to identify specific co-localized genes, thereby validating the MR results. RESULTS At the significance level corrected by Bonferroni, four immune phenotypes, including CD25 on IgD- CD38- B cells, were associated with ovarian cysts; four immune phenotypes, including CD39+ CD4+ T cell Absolute Count, were associated with ovarian dysfunction; eight immune phenotypes, including SSC-A on HLA DR+ CD8+ T cells, were associated with POF; five immune phenotypes, including CD20- CD38- B cell Absolute Count, were associated with PCOS; five immune phenotypes, including CD4+ CD8dim T cell Absolute Count, were associated with benign ovarian tumors; and three immune phenotypes, including BAFF-R on IgD- CD38+ B cells, were associated with malignant ovarian tumors. Sensitivity analysis indicated robust results. COLOC analysis identified four immune cell co-localized variants (rs150386792, rs117936291, rs75926368, and rs575687159) with ovarian diseases. CONCLUSION Our study elucidates the close genetic associations between immune cells and six ovarian-related diseases, thereby providing valuable insights for future research endeavors and clinical applications.
Collapse
Affiliation(s)
- Yangguang Lu
- The First School of Medicine, School of Information and Engineering
| | - Yingyu Yao
- Tongde Hospital of Zhejiang Province
- School of Acupuncture-Moxibustion and Tuina, School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Sijia Zhai
- The First School of Medicine, School of Information and Engineering
| | - Feitian Ni
- The Second Affiliated College, Zhejiang Chinese Medical University, Hangzhou
| | - Jingyi Wang
- The Second Affiliated College, Zhejiang Chinese Medical University, Hangzhou
| | - Feng Chen
- The Second School of Medicine, Wenzhou Medical University, Wenzhou
| | - Yige Zhang
- The First School of Medicine, School of Information and Engineering
| | - Haoyang Li
- The First School of Medicine, School of Information and Engineering
| | - Hantao Hu
- The First School of Medicine, School of Information and Engineering
| | - Hongzhi Zhang
- The First School of Medicine, School of Information and Engineering
| | - Bohuai Yu
- The First School of Medicine, School of Information and Engineering
| | - Hongbo Chen
- The First School of Medicine, School of Information and Engineering
| | - Xianfeng Huang
- The First School of Medicine, School of Information and Engineering
| | | | - Di Lu
- Tongde Hospital of Zhejiang Province
| |
Collapse
|
3
|
Fooksman DR, Jing Z, Park R. New insights into the ontogeny, diversity, maturation and survival of long-lived plasma cells. Nat Rev Immunol 2024; 24:461-470. [PMID: 38332373 DOI: 10.1038/s41577-024-00991-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Plasma cells are unique immune effectors, capable of producing large amounts of high-affinity antibodies that protect against pathogenic infections. Although most plasma cells have short lifespans, certain conditions or vaccinations can give rise to long-lived plasma cells (LLPCs) that provide individuals with lifelong protection against pathogen exposure. The nature of these LLPCs is poorly understood; however, recent studies have shed new light on the ontogeny, diversity, maturation and survival of these unique cells. Whereas LLPCs had been thought to arise preferentially from germinal centres, novel genetic tools have revealed that they can originate from various stages throughout the humoral response. Furthermore, new single-cell analyses have shown that mouse and human plasma cells are heterogeneous and may undergo further maturation in situ in the bone marrow niche. Finally, plasma cells were previously considered to be sessile cells maintained in fixed survival niches, but new data show that plasma cell subsets can differentially migrate and organize into clusters that may be associated with survival niches. These descriptive findings provide new insights into how cell-intrinsic programmes and extrinsic factors may regulate the longevity of plasma cells in various contexts, which suggest new research avenues for their functional validation.
Collapse
Affiliation(s)
- David R Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Zhixin Jing
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Rosa Park
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
4
|
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
|
5
|
Li JR, Arsang-Jang S, Cheng Y, Sun F, D'Souza A, Dhakal B, Hari P, Huang Q, Auer P, Li Y, Urrutia R, Zhan F, Shaughnessy JD, Janz S, Dong J, Cheng C. Enhancing prognostic power in multiple myeloma using a plasma cell signature derived from single-cell RNA sequencing. Blood Cancer J 2024; 14:38. [PMID: 38443358 PMCID: PMC10915134 DOI: 10.1038/s41408-024-01024-8] [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: 11/30/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024] Open
Abstract
Multiple myeloma (MM) is a heterogenous plasma cell malignancy, for which the established prognostic models exhibit limitations in capturing the full spectrum of outcome variability. Leveraging single-cell RNA-sequencing data, we developed a novel plasma cell gene signature. We evaluated and validated the associations of the resulting plasma cell malignancy (PBM) score with disease state, progression and clinical outcomes using data from five independent myeloma studies consisting of 2115 samples (1978 MM, 65 monoclonal gammopathy of undetermined significance, 35 smoldering MM, and 37 healthy controls). Overall, a higher PBM score was significantly associated with a more advanced stage within the spectrum of plasma cell dyscrasias (all p < 0.05) and a shorter overall survival in MM (hazard ratio, HR = 1.72; p < 0.001). Notably, the prognostic effect of the PBM score was independent of the International Staging System (ISS) and Revised ISS (R-ISS). The downstream analysis further linked higher PBM scores with the presence of cytogenetic abnormalities, TP53 mutations, and compositional changes in the myeloma tumor immune microenvironment. Our integrated analyses suggest the PBM score may provide an opportunity for refining risk stratification and guide decisions on therapeutic approaches to MM.
Collapse
Affiliation(s)
- Jian-Rong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Shahram Arsang-Jang
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yan Cheng
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Fumou Sun
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anita D'Souza
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Binod Dhakal
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Parameswaran Hari
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Quillan Huang
- Department of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Paul Auer
- Division of Biostatistics, Institute for Health & Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John D Shaughnessy
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Siegfried Janz
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jing Dong
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
6
|
Nguyen DC, Saney C, Hentenaar IT, Cabrera-Mora M, Capric V, Woodruff MC, Andrews J, Lonial S, Sanz I, Lee FEH. Majority of human circulating IgG plasmablasts stop blasting in a cell-free pro-survival culture. Sci Rep 2024; 14:3616. [PMID: 38350990 PMCID: PMC10864258 DOI: 10.1038/s41598-024-53977-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
Following infection or vaccination, early-minted antibody secreting cells (ASC) or plasmablasts appear in circulation transiently, and a small fraction migrates to the spleen or bone marrow (BM) to mature into long-lived plasma cells (LLPC). While LLPC, by definition, are quiescent or non-dividing, the majority of blood ASC are thought to be "blasting" or proliferative. In this study, we find > 95% nascent blood ASC in culture express Ki-67 but only 6-12% incorporate BrdU after 4 h or 24 h labeling. In contrast, < 5% BM LLPC in culture are Ki-67+ with no BrdU uptake. Due to limitations of traditional flow cytometry, we utilized a novel optofluidic technology to evaluate cell division with simultaneous functional IgG secretion. We find 11% early-minted blood ASC undergo division, and none of the terminally differentiated BM LLPC (CD19-CD38hiCD138+) divide during the 7-21 days in culture. While BM LLPC undergo complete cell cycle arrest, the process of differentiation into an ASC or plasmablasts also discourages entry into S phase. Since the majority of Ki-67+ nascent blood ASC have exited cell cycle and are no longer actively "blasting", the term "plasmablast", which traditionally refers to an ASC that still has the capacity to divide, may probably be a misnomer.
Collapse
Affiliation(s)
- Doan C Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Celia Saney
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Ian T Hentenaar
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Monica Cabrera-Mora
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Violeta Capric
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Matthew C Woodruff
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Joel Andrews
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA.
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
7
|
Heryanto YD, Zhang YZ, Imoto S. Predicting cell types with supervised contrastive learning on cells and their types. Sci Rep 2024; 14:430. [PMID: 38172501 PMCID: PMC10764802 DOI: 10.1038/s41598-023-50185-2] [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: 09/09/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) is a powerful technique that provides high-resolution expression profiling of individual cells. It significantly advances our understanding of cellular diversity and function. Despite its potential, the analysis of scRNA-seq data poses considerable challenges related to multicollinearity, data imbalance, and batch effect. One of the pivotal tasks in single-cell data analysis is cell type annotation, which classifies cells into discrete types based on their gene expression profiles. In this work, we propose a novel modeling formalism for cell type annotation with a supervised contrastive learning method, named SCLSC (Supervised Contrastive Learning for Single Cell). Different from the previous usage of contrastive learning in single cell data analysis, we employed the contrastive learning for instance-type pairs instead of instance-instance pairs. More specifically, in the cell type annotation task, the contrastive learning is applied to learn cell and cell type representation that render cells of the same type to be clustered in the new embedding space. Through this approach, the knowledge derived from annotated cells is transferred to the feature representation for scRNA-seq data. The whole training process becomes more efficient when conducting contrastive learning for cell and their types. Our experiment results demonstrate that the proposed SCLSC method consistently achieves superior accuracy in predicting cell types compared to five state-of-the-art methods. SCLSC also performs well in identifying cell types in different batch groups. The simplicity of our method allows for scalability, making it suitable for analyzing datasets with a large number of cells. In a real-world application of SCLSC to monitor the dynamics of immune cell subpopulations over time, SCLSC demonstrates a capability to discriminate cell subtypes of CD19+ B cells that were not present in the training dataset.
Collapse
Affiliation(s)
- Yusri Dwi Heryanto
- The Institute of Medical science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yao-Zhong Zhang
- The Institute of Medical science, The University of Tokyo, Tokyo, 108-8639, Japan.
| | - Seiya Imoto
- The Institute of Medical science, The University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
8
|
Del Giudice ML, Galimberti S, Buda G. Beyond BCMA, why GPRC5D could be the right way: treatment strategies with immunotherapy at relapse after anti-BCMA agents. Cancer Immunol Immunother 2023; 72:3931-3937. [PMID: 37924369 PMCID: PMC10700430 DOI: 10.1007/s00262-023-03559-4] [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: 08/21/2023] [Accepted: 10/06/2023] [Indexed: 11/06/2023]
Abstract
Multiple Myeloma remains incurable, and there is a need for therapies with novel mechanisms of action. Recently, B cell maturation antigen targeted therapy has demonstrated deep and durable responses in a largely treated population. However, the relapse rate of myeloma patients after anti-BCMA treatment strategies is increasing worldwide, and one of the most challenging issues for them is to choose the best therapy sequencing. After anti-BCMA treatment, retreatment with anti-BCMA drugs remains an option, but new targets are emerging strongly. One of them is G protein-coupled receptor, class C group 5 member D (GPRC5D), that due to the very promising data from the use of chimeric antigen receptor T-cells (CAR-T) and bispecific antibodies (BsAb) seems to be the ideal candidate in the relay of myeloma treatment at relapse. In this literature review, we discuss data from treatment with the new drugs at relapse after anti-BCMA therapies, observing an undeniable benefit from the use of drugs directed against GPRC5D.
Collapse
Affiliation(s)
- Maria Livia Del Giudice
- Department of Clinical and Experimental Medicine, Hematology, University of Pisa, 56126, Pisa, Italy.
| | - Sara Galimberti
- Department of Clinical and Experimental Medicine, Hematology, University of Pisa, 56126, Pisa, Italy
| | - Gabriele Buda
- Department of Clinical and Experimental Medicine, Hematology, University of Pisa, 56126, Pisa, Italy
| |
Collapse
|
9
|
Polmear J, Hailes L, Olshansky M, Rischmueller M, L'Estrange‐Stranieri E, Fletcher AL, Hibbs ML, Bryant VL, Good‐Jacobson KL. Targeting BMI-1 to deplete antibody-secreting cells in autoimmunity. Clin Transl Immunology 2023; 12:e1470. [PMID: 37799772 PMCID: PMC10550498 DOI: 10.1002/cti2.1470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/13/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
Objectives B cells drive the production of autoreactive antibody-secreting cells (ASCs) in autoimmune diseases such as Systemic Lupus Erythematosus (SLE) and Sjögren's syndrome, causing long-term organ damage. Current treatments for antibody-mediated autoimmune diseases target B cells or broadly suppress the immune system. However, pre-existing long-lived ASCs are often refractory to treatment, leaving a reservoir of autoreactive cells that continue to produce antibodies. Therefore, the development of novel treatment methods targeting ASCs is vital to improve patient outcomes. Our objective was to test whether targeting the epigenetic regulator BMI-1 could deplete ASCs in autoimmune conditions in vivo and in vitro. Methods Use of a BMI-1 inhibitor in both mouse and human autoimmune settings was investigated. Lyn -/- mice, a model of SLE, were treated with the BMI-1 small molecule inhibitor PTC-028, before assessment of ASCs, serum antibody and immune complexes. To examine human ASC survival, a novel human fibroblast-based assay was established, and the impact of PTC-028 on ASCs derived from Sjögren's syndrome patients was evaluated. Results BMI-1 inhibition significantly decreased splenic and bone marrow ASCs in Lyn -/- mice. The decline in ASCs was linked to aberrant cell cycle gene expression and led to a significant decrease in serum IgG3, immune complexes and anti-DNA IgG. PTC-028 was also efficacious in reducing ex vivo plasma cell survival from both Sjögren's syndrome patients and age-matched healthy donors. Conclusion These data provide evidence that inhibiting BMI-1 can deplete ASC in a variety of contexts and thus BMI-1 is a viable therapeutic target for antibody-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Jack Polmear
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Lauren Hailes
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Moshe Olshansky
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Maureen Rischmueller
- The Queen Elizabeth Hospital and Basil Hetzel InstituteWoodville SouthSAAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSAAustralia
| | | | - Anne L Fletcher
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Margaret L Hibbs
- Department of Immunology, Alfred Research AllianceMonash UniversityMelbourneVICAustralia
| | - Vanessa L Bryant
- Immunology DivisionWalter & Eliza Hall InstituteParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- Department of Clinical Immunology & AllergyThe Royal Melbourne HospitalParkvilleVICAustralia
| | - Kim L Good‐Jacobson
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| |
Collapse
|
10
|
Nguyen DC, Saney C, Hentenaar IT, Cabrera-Mora M, Woodruff MC, Andrews J, Lonial S, Sanz I, Lee FEH. Majority of human circulating plasmablasts stop blasting: A probable misnomer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557057. [PMID: 37745615 PMCID: PMC10515790 DOI: 10.1101/2023.09.10.557057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Following infection or vaccination, early-minted antibody secreting cells (ASC) or plasmablasts appear in circulation transiently, and a small fraction migrates to the spleen or bone marrow (BM) to mature into long-lived plasma cells (LLPC). While LLPC, by definition, are quiescent or non-dividing, the majority of blood ASC are thought to be "blasting" or proliferative. In this study, we find >95% nascent blood ASC in culture express Ki-67 but only 6-12% incorporate BrdU after 4h or 24h labeling. In contrast, <5% BM LLPC in culture are Ki-67 + with no BrdU uptake. Due to limitations of traditional flow cytometry, we utilized a novel optofluidic technology to evaluate cell division with simultaneous functional Ig secretion. We find 11% early-minted blood ASC undergo division, and none of the terminally differentiated BM LLPC (CD19 - CD38 hi CD138 + ) divide during the 7-21 days in culture. While BM LLPC undergo complete cell cycle arrest, the process of differentiation into an ASC of plasmablasts discourages entry into S phase. Since the majority of Ki-67 + nascent blood ASC have exited cell cycle and are no longer actively "blasting", the term "plasmablast", which traditionally refers to an ASC that still has the capacity to divide, may probably be a misnomer.
Collapse
|
11
|
Herranz-Jusdado JG, Morel E, Ordás MC, Martín D, Docando F, González L, Sanjuán E, Díaz-Rosales P, Saura M, Fouz B, Tafalla C. Yersinia ruckeri infection activates local skin and gill B cell responses in rainbow trout. FISH & SHELLFISH IMMUNOLOGY 2023; 140:108989. [PMID: 37549876 DOI: 10.1016/j.fsi.2023.108989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Teleost fish lack organized structures in mucosal tissues such as those of mammals, but instead contain dispersed B and T cells with the capacity to respond to external stimuli. Nonetheless, there is still a great lack of knowledge regarding how B cells differentiate to plasmablasts/plasma cells in these mucosal surfaces. To contribute to a further understanding of the mechanisms through which fish mucosal B cells are activated, in the current study, we have studied the B cell responses in the skin and gills of rainbow trout (Oncorhynchus mykiss) exposed to Yersinia ruckeri. We have first analyzed the transcription levels of genes related to B cell function in both mucosal surfaces, and in spleen and kidney for comparative purposes. In a second experiment, we have evaluated how the infection affects the presence and size of B cells in both skin and gills, as well as the presence of plasmablasts secreting total or specific IgMs. The results obtained in both experiments support the local differentiation of B cells to plasmablasts/plasma cells in the skin and gills of rainbow trout in response to Y. ruckeri. Interestingly, these plasmablasts/plasma cells were shown to secrete specific IgMs as soon as 5 days after the exposure. These findings contribute to a further understanding of how B cells in the periphery respond to immune stimulation in teleost fish.
Collapse
Affiliation(s)
- J G Herranz-Jusdado
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - E Morel
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - M C Ordás
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - D Martín
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - F Docando
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - L González
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - E Sanjuán
- Institute of Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Burjassot, Valencia, Spain
| | - P Díaz-Rosales
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain
| | - M Saura
- Animal Breeding Department, National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - B Fouz
- Institute of Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Burjassot, Valencia, Spain
| | - C Tafalla
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos-Alalpardo, Madrid, Spain.
| |
Collapse
|
12
|
Herranz-Jusdado JG, Morel E, Simón R, Díaz-Rosales P, Tafalla C. Teleost IgD +IgM - B cells in gills and skin have a plasmablast profile, but functionally and phenotypically differ from IgM +IgD - B cells in these sites. iScience 2023; 26:107434. [PMID: 37593459 PMCID: PMC10428129 DOI: 10.1016/j.isci.2023.107434] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/15/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Although most B cells in teleost systemic compartments co-express IgM and IgD on the surface, cells exclusively expressing either of the two Igs are common in fish mucosal tissues, providing us with a unique opportunity to further characterize IgD+IgM- B cells, an intriguing B cell subset. Hence, we compared the phenotype of IgD+IgM- cells to that of IgM+IgD- B cells in rainbow trout gills and skin, also establishing the response of these subsets to immune stimulation. The transcriptional profile and secreting capacity of IgD+IgM- B cells corresponded to that of cells that have started a differentiation program toward plasmablasts, similarly to IgM+IgD- B cells. Yet, IgM+IgD- B cells retained high levels of surface MHC II and antigen-processing abilities, while these were much lower in IgD+IgM- cells, suggesting important differences in their antigen-presenting capacities. Our findings contribute to a deeper understanding of the enigmatic role of IgD in mucosal surfaces.
Collapse
Affiliation(s)
- J. Germán Herranz-Jusdado
- Animal Health Research Center (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos 28130 Madrid, Spain
| | - Esther Morel
- Animal Health Research Center (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos 28130 Madrid, Spain
| | - Rocío Simón
- Animal Health Research Center (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos 28130 Madrid, Spain
| | - Patricia Díaz-Rosales
- Animal Health Research Center (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos 28130 Madrid, Spain
| | - Carolina Tafalla
- Animal Health Research Center (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos 28130 Madrid, Spain
| |
Collapse
|
13
|
Brownlie RJ, Kennedy R, Wilson EB, Milanovic M, Taylor CF, Wang D, Davies JR, Owston H, Adams EJ, Stephenson S, Caeser R, Gewurz BE, Giannoudis PV, Scuoppo C, McGonagle D, Hodson DJ, Tooze RM, Doody GM, Cook G, Westhead DR, Klein U. Cytokine receptor IL27RA is an NF-κB-responsive gene involved in CD38 upregulation in multiple myeloma. Blood Adv 2023; 7:3874-3890. [PMID: 36867577 PMCID: PMC10405202 DOI: 10.1182/bloodadvances.2022009044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/27/2023] [Indexed: 03/04/2023] Open
Abstract
Multiple myeloma (MM) shows constitutive activation of canonical and noncanonical nuclear factor κB (NF-κB) signaling via genetic mutations or tumor microenvironment (TME) stimulations. A subset of MM cell lines showed dependency for cell growth and survival on the canonical NF-κB transcription factor RELA alone, suggesting a critical role for a RELA-mediated biological program in MM pathogenesis. Here, we determined the RELA-dependent transcriptional program in MM cell lines and found the expression of the cell surface molecules interleukin-27 receptor-α (IL-27Rα) and the adhesion molecule JAM2 to be responsive to RELA at the messenger RNA and protein levels. IL-27Rα and JAM2 were expressed on primary MM cells at higher levels than on healthy long-lived plasma cells (PCs) in the bone marrow. IL-27 activated STAT1, and to a lesser extent STAT3, in MM cell lines and in PCs generated from memory B cells in an IL-21-dependent in vitro PC differentiation assay. Concomitant activity of IL-21 and IL-27 enhanced differentiation into PCs and increased the cell-surface expression of the known STAT target gene CD38. In accordance, a subset of MM cell lines and primary MM cells cultured with IL-27 upregulated CD38 cell-surface expression, a finding with potential implications for enhancing the efficacy of CD38-directed monoclonal antibody therapies by increasing CD38 expression on tumor cells. The elevated expression of IL-27Rα and JAM2 on MM cells compared with that on healthy PCs may be exploited for the development of targeted therapeutic strategies that modulate the interaction of MM cells with the TME.
Collapse
Affiliation(s)
- Rebecca J. Brownlie
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Ruth Kennedy
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Erica B. Wilson
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Maja Milanovic
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY
| | - Claire F. Taylor
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Dapeng Wang
- Leeds Omics, University of Leeds, Leeds, United Kingdom
| | - John R. Davies
- Bioinformatics Group, School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Heather Owston
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Emma J. Adams
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Sophie Stephenson
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Rebecca Caeser
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Peter V. Giannoudis
- Leeds Orthopaedic & Trauma Sciences, Leeds General Infirmary, and Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Claudio Scuoppo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Daniel J. Hodson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Reuben M. Tooze
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Gina M. Doody
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Gordon Cook
- CRUK Clinical Trials Unit, Leeds Institute of Clinical Trial Research, University of Leeds, Leeds, United Kingdom
| | - David R. Westhead
- Bioinformatics Group, School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Ulf Klein
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
14
|
Verstegen NJM, Pollastro S, Unger PPA, Marsman C, Elias G, Jorritsma T, Streutker M, Bassler K, Haendler K, Rispens T, Schultze JL, ten Brinke A, Beyer M, van Ham SM. Single-cell analysis reveals dynamics of human B cell differentiation and identifies novel B and antibody-secreting cell intermediates. eLife 2023; 12:83578. [PMID: 36861964 PMCID: PMC10005767 DOI: 10.7554/elife.83578] [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: 09/20/2022] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Differentiation of B cells into antibody-secreting cells (ASCs) is a key process to generate protective humoral immunity. A detailed understanding of the cues controlling ASC differentiation is important to devise strategies to modulate antibody formation. Here, we dissected differentiation trajectories of human naive B cells into ASCs using single-cell RNA sequencing. By comparing transcriptomes of B cells at different stages of differentiation from an in vitro model with ex vivo B cells and ASCs, we uncovered a novel pre-ASC population present ex vivo in lymphoid tissues. For the first time, a germinal-center-like population is identified in vitro from human naive B cells and possibly progresses into a memory B cell population through an alternative route of differentiation, thus recapitulating in vivo human GC reactions. Our work allows further detailed characterization of human B cell differentiation into ASCs or memory B cells in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Niels JM Verstegen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Sabrina Pollastro
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Peter-Paul A Unger
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Casper Marsman
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - George Elias
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Tineke Jorritsma
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Marij Streutker
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Kevin Bassler
- Genomics and Immunoregulation, University of BonnBonnGermany
| | - Kristian Haendler
- Genomics and Immunoregulation, University of BonnBonnGermany
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of BonnBonnGermany
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Joachim L Schultze
- Genomics and Immunoregulation, University of BonnBonnGermany
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of BonnBonnGermany
| | - Anja ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
| | - Marc Beyer
- Genomics and Immunoregulation, University of BonnBonnGermany
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of BonnBonnGermany
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative DiseasesBonnGermany
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of AmsterdamAmsterdamNetherlands
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
15
|
The oxidative phosphorylation inhibitor IM156 suppresses B-cell activation by regulating mitochondrial membrane potential and contributes to the mitigation of systemic lupus erythematosus. Kidney Int 2023; 103:343-356. [PMID: 36332729 DOI: 10.1016/j.kint.2022.09.031] [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: 08/03/2021] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/15/2022]
Abstract
Current treatment strategies for autoimmune diseases may not sufficiently control aberrant metabolism in B-cells. To address this concern, we investigated a biguanide derivative, IM156, as a potential regulator for B-cell metabolism in vitro and in vivo on overactive B-cells stimulated by the pro-inflammatory receptor TLR-9 agonist CpG oligodeoxynucleotide, a mimic of viral/bacterial DNA. Using RNA sequencing, we analyzed the B-cell transcriptome expression, identifying the major molecular pathways affected by IM156 in vivo. We also evaluated the anti-inflammatory effects of IM156 in lupus-prone NZB/W F1 mice. CD19+B-cells exhibited higher mitochondrial mass and mitochondrial membrane potential compared to T-cells and were more susceptible to IM156-mediated oxidative phosphorylation inhibition. In vivo, IM156 inhibited mitochondrial oxidative phosphorylation, cell cycle progression, plasmablast differentiation, and activation marker levels in CpG oligodeoxynucleotide-stimulated mouse spleen B-cells. Interestingly, IM156 treatment significantly increased overall survival, reduced glomerulonephritis and inhibited B-cell activation in the NZB/W F1 mice. Thus, our data indicated that IM156 suppressed the mitochondrial membrane potentials of activated B-cells in mice, contributing to the mitigation of lupus activity. Hence, IM156 may represent a therapeutic alternative for autoimmune disease mediated by B-cell hyperactivity.
Collapse
|
16
|
Nielsen CM, Barrett JR, Davis C, Fallon JK, Goh C, Michell AR, Griffin C, Kwok A, Loos C, Darko S, Laboune F, Tekman M, Diouf A, Miura K, Francica JR, Ransier A, Long CA, Silk SE, Payne RO, Minassian AM, Lauffenburger DA, Seder RA, Douek DC, Alter G, Draper SJ. Delayed boosting improves human antigen-specific Ig and B cell responses to the RH5.1/AS01B malaria vaccine. JCI Insight 2023; 8:e163859. [PMID: 36692019 PMCID: PMC9977309 DOI: 10.1172/jci.insight.163859] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/30/2022] [Indexed: 01/24/2023] Open
Abstract
Modifications to vaccine delivery that increase serum antibody longevity are of great interest for maximizing efficacy. We have previously shown that a delayed fractional (DFx) dosing schedule (0-1-6 month) - using AS01B-adjuvanted RH5.1 malaria antigen - substantially improves serum IgG durability as compared with monthly dosing (0-1-2 month; NCT02927145). However, the underlying mechanism and whether there are wider immunological changes with DFx dosing were unclear. Here, PfRH5-specific Ig and B cell responses were analyzed in depth through standardized ELISAs, flow cytometry, systems serology, and single-cell RNA-Seq (scRNA-Seq). Data indicate that DFx dosing increases the magnitude and durability of circulating PfRH5-specific B cells and serum IgG1. At the peak antibody magnitude, DFx dosing was distinguished by a systems serology feature set comprising increased FcRn binding, IgG avidity, and proportion of G2B and G2S2F IgG Fc glycans, alongside decreased IgG3, antibody-dependent complement deposition, and proportion of G1S1F IgG Fc glycan. Concomitantly, scRNA-Seq data show a higher CDR3 percentage of mutation from germline and decreased plasma cell gene expression in circulating PfRH5-specific B cells. Our data, therefore, reveal a profound impact of DFx dosing on the humoral response and suggest plausible mechanisms that could enhance antibody longevity, including improved FcRn binding by serum Ig and a potential shift in the underlying cellular response from circulating short-lived plasma cells to nonperipheral long-lived plasma cells.
Collapse
Affiliation(s)
| | | | - Christine Davis
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
| | - Jonathan K. Fallon
- Ragon Institute of Massachusetts General Hospital (MGH), MIT and Harvard, Boston, Massachusetts, USA
| | - Cyndi Goh
- University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Ashlin R. Michell
- Ragon Institute of Massachusetts General Hospital (MGH), MIT and Harvard, Boston, Massachusetts, USA
| | - Catherine Griffin
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
| | - Andrew Kwok
- University of Oxford, Oxford, Oxfordshire, United Kingdom
- Wellcome Center for Human Genetics, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Carolin Loos
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Massachusetts General Hospital (MGH), MIT and Harvard, Boston, Massachusetts, USA
| | - Samuel Darko
- Vaccine Research Center, NIAID/NIH, Bethesda, Maryland, USA
| | - Farida Laboune
- Vaccine Research Center, NIAID/NIH, Bethesda, Maryland, USA
| | - Mehmet Tekman
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | | | - Amy Ransier
- Vaccine Research Center, NIAID/NIH, Bethesda, Maryland, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Sarah E. Silk
- University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Ruth O. Payne
- University of Oxford, Oxford, Oxfordshire, United Kingdom
| | | | | | | | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital (MGH), MIT and Harvard, Boston, Massachusetts, USA
| | | |
Collapse
|
17
|
Morel E, Herranz-Jusdado JG, Simón R, Abós B, Perdiguero P, Martín-Martín A, Andrés G, Muñoz-Atienza E, Guerra Rodriguez M, Díaz-Rosales P, Tafalla C. Endoplasmic reticulum expansion throughout the differentiation of teleost B cells to plasmablasts. iScience 2022; 26:105854. [PMID: 36619985 PMCID: PMC9812713 DOI: 10.1016/j.isci.2022.105854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/18/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
The differentiation of B cells into antibody-secreting cells is fundamental for the generation of humoral immunity. In mammals, this process involves a series of metabolic and intracellular changes, not studied to date in teleost fish, where a clear distinction between naive B cells and plasmablasts/plasma cells (PCs) is still missing. Thus, in the current study, we have established that upon activation, teleost B cells undergo an expansion of the endoplasmic reticulum (ER) but experience no significant changes in mitochondria content. In parallel, the transcription of genes implicated in B cell differentiation increases, while that of mitochondrial genes decreases. In this context, ER monitoring has allowed us to distinguish between small cells with low amounts of ER (FSCloERlo B cells), that correspond to undifferentiated cells, and large cells with expanded ER (FSChiERhi B cells), characterized as plasmablasts. The results shed new light on the B cell differentiation process in teleosts and provide us with novel tools to study B cell function in these species.
Collapse
Affiliation(s)
- Esther Morel
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain
| | | | - Rocío Simón
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain
| | - Beatriz Abós
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain
| | - Pedro Perdiguero
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain,Department of Genetics, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid (UCM), Madrid 28040, Spain
| | - Alba Martín-Martín
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain
| | - Germán Andrés
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain,Centre for Molecular Biology Severo Ochoa (CBMSO), Madrid 28049, Spain
| | | | | | | | - Carolina Tafalla
- Animal Health Research Center (CISA-INIA), Valdeolmos, Madrid 28130, Spain,Corresponding author
| |
Collapse
|
18
|
Wu L, Yang Y, Gao A, Li J, Ye J. Teleost fish IgM+ plasma-like cells possess IgM-secreting, phagocytic, and antigen-presenting capacities. Front Immunol 2022; 13:1016974. [PMID: 36225937 PMCID: PMC9550268 DOI: 10.3389/fimmu.2022.1016974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
Plasma cells are terminally differentiated antibody-secreting B lymphocytes that contribute to humoral immunity by producing large numbers of antibodies. Increasing evidence suggests that teleost fish B cells share certain characteristics with mammalian B1 B cells, including antibody-secreting, phagocytic, and antigen-presenting capacities. However, the difference between mature B cells and plasma cells remains unclear. In this study, we found that, based on their light-scattering characteristics, tilapia anterior kidney (AK) leukocytes can be categorized into two IgM+ B-cell subsets: the lymphoid (L) gate and granulocyte–monocyte/macrophage (G-M) subsets. G-M gate cells are more numerous than L-gate cells and have higher mean fluorescence, but lower forward scatter and side scatter. We analyzed the morphological and ultrastructural features of sorted IgM+ cells and found that L-gate IgM+ cells have a high nucleus–cytoplasm ratio and lymphocyte-like morphology, whereas G-M gate IgM+ cells have a small nucleus, more abundant endoplasmic reticulum, and a larger number of mitochondria, and have a plasma cell-like or macrophage-like morphology. To further characterize the cell types, we examined the specific patterns of expression of B-cell- and T-cell-related genes. We found that B-cell-specific genes were expressed by both L-gate and G-M gate IgM+ cells, and that G-M gate IgM+ cells secreted extremely high levels of IgM. However, T-cell-related genes were highly expressed only in L-gate IgM– cells. These results suggest that G-M gate IgM+ cells are similar to plasma-like cells, with high antibody-secreting capacity. Given that G-M gate cells include the granulocyte, monocyte, and macrophage cell types, but not B cells, monocyte/macrophage markers were used to investigate the cell types further. A macrophage receptor with a collagenous structure was frequently observed, and macrophage-expressed gene-1 was highly expressed, in the G-M gate IgM+ cells. Phagocytic capacity, as determined by ingestion of beads or bacteria, was significantly higher in G-M gate IgM+ cells than in L-gate IgM+ cells, as was antigen-processing capacity. Our findings show that tilapia AK leukocytes can be divided into two IgM+ B-cell subsets and that G-M gate IgM+ cells resemble plasma-like cells, having high antibody-secreting, phagocytic, and antigen-presenting capacities. Thus, this study increases our understanding of the functions of teleost fish plasma-like cells.
Collapse
Affiliation(s)
- Liting Wu
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yanjian Yang
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Along Gao
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jun Li
- Laboratory for Marine Fisheries Science and Food Production Processes, Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- School of Science and Medicine, Lake Superior State University, Sault Sainte Marie, MI, United States
| | - Jianmin Ye
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou, China
- *Correspondence: Jianmin Ye,
| |
Collapse
|
19
|
Cai H, Kakiuchi-Kiyota S, Hendricks R, Zhong S, Liu L, Adedeji AO, Chan P, Schutten MM, Kamath AV, Ovacik MA. Nonclinical Pharmacokinetics, Pharmacodynamics, and Translational Model of RO7297089, A Novel Anti-BCMA/CD16A Bispecific Tetravalent Antibody for the Treatment of Multiple Myeloma. AAPS J 2022; 24:100. [PMID: 36127472 DOI: 10.1208/s12248-022-00744-8] [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: 05/27/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023] Open
Abstract
RO7297089, an anti-B-cell maturation antigen (BCMA)/CD16A bispecific tetravalent antibody, is being developed as a multiple myeloma (MM) therapeutic. This study characterized nonclinical pharmacokinetics (PK), pharmacodynamics (PD), soluble BCMA (sBCMA), and soluble CD16 (sCD16) changes following administration of RO7297089 to support clinical trials. Unbound and total RO7297089 concentrations were measured in cynomolgus monkeys. RO7297089 exhibited a bi-phasic systemic concentration-time profile, similar to a typical human immunoglobulin 1 antibody. Target engagement by RO7297089 led to a robust increase (~100-fold) in total systemic sBCMA levels and relatively mild increase (~2-fold) in total sCD16 levels. To describe the relationship of nonclinical PK/PD data, we developed a target-mediated drug disposition (TMDD) model that includes the systemic target engagement of membrane BCMA (mBCMA), sBCMA, membrane CD16 (mCD16), and sCD16. We then used this model to simulate the PK/PD relationship of RO7297089 in MM patients by translating relevant PK parameters and target levels, based on the literature and newly generated data such as baseline sCD16A levels. Our model suggested that the impact of TMDD on RO7297089 exposure may be more significant in MM patients due to significantly higher expression levels of both mBCMA and sBCMA compared to healthy cynomolgus monkeys. Based on model simulations, we propose more frequent dosing of RO7297089 compared to regular monthly frequency in the clinic at the beginning of treatment to ensure sustained target engagement. This study demonstrates a translational research strategy for collecting relevant nonclinical data, establishing a TMDD model, and using simulations from this model to inform clinical dose regimens.
Collapse
Affiliation(s)
- Hao Cai
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Satoko Kakiuchi-Kiyota
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Robert Hendricks
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Shelly Zhong
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Luna Liu
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Adeyemi O Adedeji
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Pamela Chan
- Biochemical Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Melissa M Schutten
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Amrita V Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Meric A Ovacik
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
20
|
L'Imperio V, Morello G, Vegliante MC, Cancila V, Bertolazzi G, Mazzara S, Belmonte B, Mangogna A, Balzarini P, Corral L, Lopez G, Di Napoli A, Facchetti F, Pagni F, Tripodo C. Spatial transcriptome of a germinal center plasmablastic burst hints at MYD88/CD79B mutants-enriched diffuse large B-cell lymphomas. Eur J Immunol 2022; 52:1350-1361. [PMID: 35554927 PMCID: PMC9546146 DOI: 10.1002/eji.202149746] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022]
Abstract
The GC reaction results in the selection of B cells acquiring effector Ig secreting ability by progressing toward plasmablastic differentiation. This transition is associated with exclusion from the GC microenvironment. The aberrant expansion of plasmablastic elements within the GC fringes configures an atypical condition, the biological characteristics of which have not been defined yet. We investigated the in situ immunophenotypical and transcriptional characteristics of a nonclonal germinotropic expansion of plasmablastic elements (GEx) occurring in the tonsil of a young patient. Compared to neighboring GC and perifollicular regions, the GEx showed a distinctive signature featuring key regulators of plasmacytic differentiation, cytokine signaling, and cell metabolism. The GEx signature was tested in the setting of diffuse large B‐cell lymphoma (DLBCL) as a prototypical model of lymphomagenesis encompassing transformation at different stages of GC and post‐GC functional differentiation. The signature outlined DLBCL clusters with different immune microenvironment composition and enrichment in genetic subtypes. This report represents the first insight into the transcriptional features of a germinotropic plasmablastic burst, shedding light into the molecular hallmarks of B cells undergoing plasmablastic differentiation and aberrant expansion within the noncanonical setting of the GC microenvironment.
Collapse
Affiliation(s)
- Vincenzo L'Imperio
- Department of Medicine and Surgery, University of Milano-Bicocca, Pathology, San Gerardo Hospital, Via G.B. Pergolesi 33, Monza, Italy
| | - Gaia Morello
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | | | - Valeria Cancila
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Giorgio Bertolazzi
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Saveria Mazzara
- Division of Diagnostic Haematopathology, European Institute of Oncology, Milan, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, 34137, Italy
| | - Piera Balzarini
- Department of Molecular and Translational Medicine, University of Brescia, Piazzale Spedali Civili 1, Brescia, 25123, Italy
| | - Lilia Corral
- Centro Ricerca Tettamanti, Pediatric Clinic, University of Milan Bicocca, San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Gianluca Lopez
- Pathology Unit, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Arianna Di Napoli
- Pathology Unit, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | | | - Fabio Pagni
- Department of Medicine and Surgery, University of Milano-Bicocca, Pathology, San Gerardo Hospital, Via G.B. Pergolesi 33, Monza, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy.,the FIRC Institute of Molecular Oncology, Tumor and Microenvironment Histopathology Unit, IFOM, Milan, Italy
| |
Collapse
|
21
|
Rangel-Moreno J, Garcia-Hernandez MDLL, Owen T, Barnard J, Becerril-Villanueva E, Kashyap T, Argueta C, Gamboa-Dominguez A, Tamir S, Landesman Y, Goldman BI, Ritchlin CT, Anolik JH. Small molecule inhibitors of nuclear export ameliorate lupus by modulating plasma cell generation and survival. Arthritis Rheumatol 2022; 74:1363-1375. [PMID: 35333447 PMCID: PMC9339462 DOI: 10.1002/art.42128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/08/2022] [Accepted: 03/22/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To investigate the hypothesis that selective inhibitors of nuclear export (SINE), recently approved for the treatment of refractory plasma cell (PC) malignancy, may have potential in the treatment of lupus. METHODS NZB/NZW female mice were treated with SINE or vehicle control. Tissue was harvested and analyzed by flow cytometry using standard markers. Nephritis was monitored by evaluation for proteinuria and by histologic analysis of kidneys. Serum anti- double-stranded DNA (anti-dsDNA) levels were measured by enzyme-linked immunosorbent assay (ELISA) and total IgG and dsDNA antibody-secreting cells (ASC) by enzyme-linked immunospot assay. RESULTS SINE abrogated murine lupus nephritis at both early and late stages of the disease and rapidly impaired generation of autoreactive PC in germinal centers (GC). SINE inhibited the production of the NF-κB-driven homeostatic chemokines by stromal cells, altering splenic B and T cell strategic positioning and significantly reducing T follicular helper cells (TFH), GC B cells, and autoreactive PC. SINE also decreased cytokines/chemokines involved in PC survival and recruitment in the kidney of lupus-prone mice. Exportin 1, the SINE target, was detected in GC of human tonsils, splenic B cells of lupus patients, and multiple B cell subsets in the kidney of patients with lupus nephritis. CONCLUSION Our collective results support the therapeutic potential of SINE via targeting several molecular and cellular pathways critical in lupus pathogenesis, including autoantibody production by plasma cells.
Collapse
Affiliation(s)
- Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Maria de la Luz Garcia-Hernandez
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Teresa Owen
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Jennifer Barnard
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Enrique Becerril-Villanueva
- Departamento de Psicoinmunología, Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | | | | | - Armando Gamboa-Dominguez
- Departamento de Patologia, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Sharon Tamir
- Karyopharm Therapeutics, Newton, Massachusetts, 02459
| | | | - Bruce I Goldman
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642
| | - Christopher T Ritchlin
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Jennifer H Anolik
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14642.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642
| |
Collapse
|
22
|
Stage-Specific Non-Coding RNA Expression Patterns during In Vitro Human B Cell Differentiation into Antibody Secreting Plasma Cells. Noncoding RNA 2022; 8:ncrna8010015. [PMID: 35202088 PMCID: PMC8878715 DOI: 10.3390/ncrna8010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
The differentiation of B cells into antibody secreting plasma cells (PCs) is governed by a strict regulatory network that results in expression of specific transcriptomes along the activation continuum. In vitro models yielding significant numbers of PCs phenotypically identical to the in vivo state enable investigation of pathways, metabolomes, and non-coding (ncRNAs) not previously identified. The objective of our study was to characterize ncRNA expression during human B cell activation and differentiation. To achieve this, we used an in vitro system and performed RNA-seq on resting and activated B cells and PCs. Characterization of coding gene transcripts, including immunoglobulin (Ig), validated our system and also demonstrated that memory B cells preferentially differentiated into PCs. Importantly, we identified more than 980 ncRNA transcripts that are differentially expressed across the stages of activation and differentiation, some of which are known to target transcription, proliferation, cytoskeletal, autophagy and proteasome pathways. Interestingly, ncRNAs located within Ig loci may be targeting both Ig and non-Ig-related transcripts. ncRNAs associated with B cell malignancies were also identified. Taken together, this system provides a platform to study the role of specific ncRNAs in B cell differentiation and altered expression of those ncRNAs involved in B cell malignancies.
Collapse
|
23
|
RNA demethylase ALKBH5 promotes tumorigenesis in multiple myeloma via TRAF1-mediated activation of NF-κB and MAPK signaling pathways. Oncogene 2022; 41:400-413. [PMID: 34759347 PMCID: PMC8755544 DOI: 10.1038/s41388-021-02095-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023]
Abstract
N6-methyladenosine (m6A), an internal modification in mRNA, plays a critical role in regulating gene expression. Dysregulation of m6A modifiers promotes oncogenesis through enzymatic functions that disrupt the balance between the deposition and removal of m6A modification on critical transcripts. However, the roles of mRNA m6A in multiple myeloma (MM) are poorly understood. The present study showed that RNA demethylase ALKBH5 was overexpressed in MM and associated with a poor prognosis in MM patients. Knocking down ALKBH5 induced apoptosis and inhibited the growth of MM cells in vitro. Xenograft models and gene set enrichment analysis with patient transcriptome datasets also supported the oncogenic role of ALKBH5 in MM. Mechanistic studies showed that ALKBH5 exerted tumorigenic effects in myeloma in an m6A-dependent manner, and TNF receptor-associated factor 1 (TRAF1) was a critical target of ALKBH5. Specifically, ALKBH5 regulated TRAF1 expression via decreasing m6A abundance in the 3'-untranslated region (3'-UTR) of TRAF1 transcripts and enhancing TRAF1 mRNA stability. As a result, ALKBH5 promoted MM cell growth and survival through TRAF1-mediated activation of NF-κB and MAPK signaling pathways. Collectively, our data demonstrated that ALKBH5 played a critical role in MM tumorigenesis and suggested that ALKBH5 could be a novel therapeutic target in MM.
Collapse
|
24
|
Di Pietro A, Polmear J, Cooper L, Damelang T, Hussain T, Hailes L, O'Donnell K, Udupa V, Mi T, Preston S, Shtewe A, Hershberg U, Turner SJ, La Gruta NL, Chung AW, Tarlinton DM, Scharer CD, Good-Jacobson KL. Targeting BMI-1 in B cells restores effective humoral immune responses and controls chronic viral infection. Nat Immunol 2022; 23:86-98. [PMID: 34845392 DOI: 10.1038/s41590-021-01077-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/20/2021] [Indexed: 01/12/2023]
Abstract
Ineffective antibody-mediated responses are a key characteristic of chronic viral infection. However, our understanding of the intrinsic mechanisms that drive this dysregulation are unclear. Here, we identify that targeting the epigenetic modifier BMI-1 in mice improves humoral responses to chronic lymphocytic choriomeningitis virus. BMI-1 was upregulated by germinal center B cells in chronic viral infection, correlating with changes to the accessible chromatin landscape, compared to acute infection. B cell-intrinsic deletion of Bmi1 accelerated viral clearance, reduced splenomegaly and restored splenic architecture. Deletion of Bmi1 restored c-Myc expression in B cells, concomitant with improved quality of antibody and coupled with reduced antibody-secreting cell numbers. Specifically, BMI-1-deficiency induced antibody with increased neutralizing capacity and enhanced antibody-dependent effector function. Using a small molecule inhibitor to murine BMI-1, we could deplete antibody-secreting cells and prohibit detrimental immune complex formation in vivo. This study defines BMI-1 as a crucial immune modifier that controls antibody-mediated responses in chronic infection.
Collapse
Affiliation(s)
- Andrea Di Pietro
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jack Polmear
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Lucy Cooper
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Timon Damelang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Tabinda Hussain
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Lauren Hailes
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kristy O'Donnell
- Department of Immunology & Pathology, Alfred Research Alliance, Monash University, Melbourne, Victoria, Australia
| | - Vibha Udupa
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia.,Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Tian Mi
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Simon Preston
- Divisions of Immunology and Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Areen Shtewe
- Department of Human Biology, Faculty of Science, University of Haifa, Haifa, Israel
| | - Uri Hershberg
- Department of Human Biology, Faculty of Science, University of Haifa, Haifa, Israel
| | - Stephen J Turner
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Nicole L La Gruta
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - David M Tarlinton
- Department of Immunology & Pathology, Alfred Research Alliance, Monash University, Melbourne, Victoria, Australia
| | - Christopher D Scharer
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kim L Good-Jacobson
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia. .,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
25
|
Aggarwal C, Saini K, Reddy ES, Singla M, Nayak K, Chawla YM, Maheshwari D, Singh P, Sharma P, Bhatnagar P, Kumar S, Gottimukkala K, Panda H, Gunisetty S, Davis CW, Kissick HT, Kabra SK, Lodha R, Medigeshi GR, Ahmed R, Murali-Krishna K, Chandele A. Immunophenotyping and Transcriptional Profiling of Human Plasmablasts in Dengue. J Virol 2021; 95:e0061021. [PMID: 34523972 PMCID: PMC8577383 DOI: 10.1128/jvi.00610-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/11/2021] [Indexed: 12/07/2022] Open
Abstract
Plasmablasts represent a specialized class of antibody-secreting effector B cells that transiently appear in blood circulation following infection or vaccination. The expansion of these cells generally tends to be massive in patients with systemic infections such as dengue or Ebola that cause hemorrhagic fever. To gain a detailed understanding of human plasmablast responses beyond antibody expression, here, we performed immunophenotyping and RNA sequencing (RNA-seq) analysis of the plasmablasts from dengue febrile children in India. We found that plasmablasts expressed several adhesion molecules and chemokines or chemokine receptors that are involved in endothelial interactions or homing to inflamed tissues, including skin, mucosa, and intestine, and upregulated the expression of several cytokine genes that are involved in leukocyte extravasation and angiogenesis. These plasmablasts also upregulated the expression of receptors for several B-cell prosurvival cytokines that are known to be induced robustly in systemic viral infections such as dengue, some of which generally tend to be relatively higher in patients manifesting hemorrhage and/or shock than in patients with mild febrile infection. These findings improve our understanding of human plasmablast responses during the acute febrile phase of systemic dengue infection. IMPORTANCE Dengue is globally spreading, with over 100 million clinical cases annually, with symptoms ranging from mild self-limiting febrile illness to more severe and sometimes life-threatening dengue hemorrhagic fever or shock, especially among children. The pathophysiology of dengue is complex and remains poorly understood despite many advances indicating a key role for antibody-dependent enhancement of infection. While serum antibodies have been extensively studied, the characteristics of the early cellular factories responsible for antibody production, i.e., plasmablasts, are only beginning to emerge. This study provides a comprehensive understanding of the transcriptional profiles of human plasmablasts from dengue patients.
Collapse
Affiliation(s)
- Charu Aggarwal
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Keshav Saini
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Elluri Seetharami Reddy
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| | - Mohit Singla
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Kaustuv Nayak
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Yadya M. Chawla
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Deepti Maheshwari
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Prabhat Singh
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Pragati Sharma
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Biotechnology, School of Chemical and Life Sciences, New Delhi, India
| | - Priya Bhatnagar
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- TERI School of Advanced Studies, New Delhi, India
| | - Sanjeev Kumar
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Kamalvishnu Gottimukkala
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Harekrushna Panda
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sivaram Gunisetty
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Carl W. Davis
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Haydn Thomas Kissick
- Department of Microbiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sushil Kumar Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rakesh Lodha
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Microbiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kaja Murali-Krishna
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anmol Chandele
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
26
|
Mukund K, Nayak P, Ashokkumar C, Rao S, Almeda J, Betancourt-Garcia MM, Sindhi R, Subramaniam S. Immune Response in Severe and Non-Severe Coronavirus Disease 2019 (COVID-19) Infection: A Mechanistic Landscape. Front Immunol 2021; 12:738073. [PMID: 34721400 PMCID: PMC8548832 DOI: 10.3389/fimmu.2021.738073] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
The mechanisms underlying the immune remodeling and severity response in coronavirus disease 2019 (COVID-19) are yet to be fully elucidated. Our comprehensive integrative analyses of single-cell RNA sequencing (scRNAseq) data from four published studies, in patients with mild/moderate and severe infections, indicate a robust expansion and mobilization of the innate immune response and highlight mechanisms by which low-density neutrophils and megakaryocytes play a crucial role in the cross talk between lymphoid and myeloid lineages. We also document a marked reduction of several lymphoid cell types, particularly natural killer cells, mucosal-associated invariant T (MAIT) cells, and gamma-delta T (γδT) cells, and a robust expansion and extensive heterogeneity within plasmablasts, especially in severe COVID-19 patients. We confirm the changes in cellular abundances for certain immune cell types within a new patient cohort. While the cellular heterogeneity in COVID-19 extends across cells in both lineages, we consistently observe certain subsets respond more potently to interferon type I (IFN-I) and display increased cellular abundances across the spectrum of severity, as compared with healthy subjects. However, we identify these expanded subsets to have a more muted response to IFN-I within severe disease compared to non-severe disease. Our analyses further highlight an increased aggregation potential of the myeloid subsets, particularly monocytes, in COVID-19. Finally, we provide detailed mechanistic insights into the interaction between lymphoid and myeloid lineages, which contributes to the multisystemic phenotype of COVID-19, distinguishing severe from non-severe responses.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Priya Nayak
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Chethan Ashokkumar
- Plexision Inc., Pittsburgh, PA, United States
- Hillman Center for Pediatric Transplantation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sohail Rao
- DHR Health and DHR Health Institute for Research and Development, Edinburg, TX, United States
| | - Jose Almeda
- DHR Health and DHR Health Institute for Research and Development, Edinburg, TX, United States
| | | | - Rakesh Sindhi
- Plexision Inc., Pittsburgh, PA, United States
- Hillman Center for Pediatric Transplantation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
27
|
Gupta VA, Barwick BG, Matulis SM, Shirasaki R, Jaye DL, Keats JJ, Oberlton B, Joseph NS, Hofmeister CC, Heffner LT, Dhodapkar MV, Nooka AK, Lonial S, Mitsiades CS, Kaufman JL, Boise LH. Venetoclax sensitivity in multiple myeloma is associated with B-cell gene expression. Blood 2021; 137:3604-3615. [PMID: 33649772 PMCID: PMC8462405 DOI: 10.1182/blood.2020007899] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
Venetoclax is a highly potent, selective BCL2 inhibitor capable of inducing apoptosis in cells dependent on BCL2 for survival. Most myeloma is MCL1-dependent; however, a subset of myeloma enriched for translocation t(11;14) is codependent on BCL2 and thus sensitive to venetoclax. The biology underlying this heterogeneity remains poorly understood. We show that knockdown of cyclin D1 does not induce resistance to venetoclax, arguing against a direct role for cyclin D1 in venetoclax sensitivity. To identify other factors contributing to venetoclax response, we studied a panel of 31 myeloma cell lines and 25 patient samples tested for venetoclax sensitivity. In cell lines, we corroborated our previous observation that BIM binding to BCL2 correlates with venetoclax response and further showed that knockout of BIM results in decreased venetoclax sensitivity. RNA-sequencing analysis identified expression of B-cell genes as enriched in venetoclax-sensitive myeloma, although no single gene consistently delineated sensitive and resistant cells. However, a panel of cell surface makers correlated well with ex vivo prediction of venetoclax response in 21 patient samples and may serve as a biomarker independent of t(11;14). Assay for transposase-accessible chromatin sequencing of myeloma cell lines also identified an epigenetic program in venetoclax-sensitive cells that was more similar to B cells than that of venetoclax-resistant cells, as well as enrichment for basic leucine zipper domain-binding motifs such as BATF. Together, these data indicate that remnants of B-cell biology are associated with BCL2 dependency and point to novel biomarkers of venetoclax-sensitive myeloma independent of t(11;14).
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Tumor
- Chromosomes, Human, Pair 11/genetics
- Chromosomes, Human, Pair 11/metabolism
- Chromosomes, Human, Pair 14/genetics
- Chromosomes, Human, Pair 14/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Knockdown Techniques
- Humans
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Sulfonamides/pharmacology
- Translocation, Genetic/drug effects
Collapse
Affiliation(s)
- Vikas A Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Shannon M Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Ryosuke Shirasaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - David L Jaye
- Department of Pathology and Laboratory Medicine, Winship Cancer Institute of Emory University, Atlanta, GA; and
| | - Jonathan J Keats
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Benjamin Oberlton
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Nisha S Joseph
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Leonard T Heffner
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | | | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| |
Collapse
|
28
|
Su MT, Inui M, Wong YL, Takahashi M, Sugahara-Tobinai A, Ono K, Miyamoto S, Murakami K, Itoh-Nakadai A, Kezuka D, Itoi S, Endo S, Hirayasu K, Arase H, Takai T. Blockade of checkpoint ILT3/LILRB4/gp49B binding to fibronectin ameliorates autoimmune disease in BXSB/Yaa mice. Int Immunol 2021; 33:447-458. [PMID: 34089617 DOI: 10.1093/intimm/dxab028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 06/02/2021] [Indexed: 01/01/2023] Open
Abstract
The extracellular matrix (ECM) is the basis for virtually all cellular processes and is also related to tumor metastasis. Fibronectin (FN), a major ECM macromolecule expressed by different cell types and also present in plasma, consists of multiple functional modules that bind to ECM-associated, plasma, and cell-surface proteins such as integrins and FN itself, thus ensuring its cell-adhesive and modulatory role. Here we show that FN constitutes an immune checkpoint. Thus, FN was identified as a physiological ligand for a tumor/leukemia/lymphoma- as well as autoimmune-associated checkpoint, ILT3/LILRB4 (B4, CD85k). Human B4 and the murine ortholog, gp49B, bound FN with sub-micromolar affinities as assessed by bio-layer interferometry. The major B4-binding site in FN was located at the N-terminal 30-kDa module (FN30), which is apart from the major integrin-binding site present at the middle of the molecule. Blockade of B4-FN binding such as with B4 antibodies or a recombinant FN30-Fc fusion protein paradoxically ameliorated autoimmune disease in lupus-prone BXSB/Yaa mice. The unexpected nature of the B4-FN checkpoint in autoimmunity is discussed, referring to its potential role in tumor immunity.
Collapse
Affiliation(s)
- Mei-Tzu Su
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Masanori Inui
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Yi Li Wong
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Maika Takahashi
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Akiko Sugahara-Tobinai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Karin Ono
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Shotaro Miyamoto
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Keiichi Murakami
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Ari Itoh-Nakadai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Dai Kezuka
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - So Itoi
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Shota Endo
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kouyuki Hirayasu
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa 920-8640, Japan.,Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Hisashi Arase
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
29
|
Demers-Mathieu V, Mathijssen GB, DaPra C, Medo E. The effects of probiotic supplementation on the gene expressions of immune cell surface markers and levels of antibodies and pro-inflammatory cytokines in human milk. J Perinatol 2021; 41:1083-1091. [PMID: 33208844 DOI: 10.1038/s41372-020-00875-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/07/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study investigated the impact of probiotic supplementation on the gene expressions of cluster of differentiation (CD) as cell markers and the concentrations of antibodies and cytokines in human milk. STUDY DESIGN Gene expressions of CD28, CD19, and CD38 were determined in milk from 15 women ingesting daily probiotics (from Greek yogurt) and 12 women who do not consume probiotics. Concentrations of antibodies and cytokines were measured using ELISA. RESULTS Gene expression of CD28 tended to be higher in milk from mothers ingesting daily probiotics than mothers who did not take probiotics. Interleukin-6 (IL-6) concentration in milk was higher in mothers ingesting probiotics than those who do not consume probiotics. The increase of IL-6 level in human milk was positively correlated with total IgA and IgG concentrations. CONCLUSIONS Probiotic supplementation could enhance the secretion of IL-6 in human milk. Human milk IL-6 may improve neonatal immunity due to its stimulation of total IgA and IgG.
Collapse
Affiliation(s)
- Veronique Demers-Mathieu
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA.
| | - Gabrielle B Mathijssen
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Ciera DaPra
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Elena Medo
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| |
Collapse
|
30
|
Kassambara A, Herviou L, Ovejero S, Jourdan M, Thibaut C, Vikova V, Pasero P, Elemento O, Moreaux J. RNA-sequencing data-driven dissection of human plasma cell differentiation reveals new potential transcription regulators. Leukemia 2021; 35:1451-1462. [PMID: 33824465 PMCID: PMC8102200 DOI: 10.1038/s41375-021-01234-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Plasma cells (PCs) play an important role in the adaptive immune system through a continuous production of antibodies. We have demonstrated that PC differentiation can be modeled in vitro using complex multistep culture systems reproducing sequential differentiation process occurring in vivo. Here we present a comprehensive, temporal program of gene expression data encompassing human PC differentiation (PCD) using RNA sequencing (RNA-seq). Our results reveal 6374 differentially expressed genes classified into four temporal gene expression patterns. A stringent pathway enrichment analysis of these gene clusters highlights known pathways but also pathways largely unknown in PCD, including the heme biosynthesis and the glutathione conjugation pathways. Additionally, our analysis revealed numerous novel transcriptional networks with significant stage-specific overexpression and potential importance in PCD, including BATF2, BHLHA15/MIST1, EZH2, WHSC1/MMSET, and BLM. We have experimentally validated a potent role for BLM in regulating cell survival and proliferation during human PCD. Taken together, this RNA-seq analysis of PCD temporal stages helped identify coexpressed gene modules with associated up/downregulated transcription regulator genes that could represent major regulatory nodes for human PC maturation. These data constitute a unique resource of human PCD gene expression programs in support of future studies for understanding the underlying mechanisms that control PCD.
Collapse
Affiliation(s)
- Alboukadel Kassambara
- Department of Biological Hematology, CHU Montpellier, Montpellier, France.,IGH, CNRS, University of Montpellier, Montpellier, France
| | - Laurie Herviou
- IGH, CNRS, University of Montpellier, Montpellier, France
| | - Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, Montpellier, France.,IGH, CNRS, University of Montpellier, Montpellier, France
| | - Michel Jourdan
- IGH, CNRS, University of Montpellier, Montpellier, France
| | | | | | | | - Olivier Elemento
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France. .,IGH, CNRS, University of Montpellier, Montpellier, France. .,University of Montpellier, UFR Medicine, Montpellier, France. .,Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
31
|
Hosoya H, Sidana S. Antibody-Based Treatment Approaches in Multiple Myeloma. Curr Hematol Malig Rep 2021; 16:183-191. [PMID: 33730360 PMCID: PMC8715952 DOI: 10.1007/s11899-021-00624-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 01/25/2023]
Abstract
PURPOSE OF REVIEW The field of multiple myeloma treatment has entered a new era with antibody-based approaches in clinical practice. In this review, we focus on the clinical approaches of utilizing antibody-based modality, specifically monoclonal antibodies, antibody-drug conjugates, and bispecific T-cell antibodies in the treatment of multiple myeloma. RECENT FINDINGS Three monoclonal antibodies (daratumumab, isatuximab, elotuzumab) and one anti-BCMA (B-cell maturation antigen) antibody-drug conjugate (belantamab mafodotin) have been approved by the FDA in the last 5 years for the treatment of multiple myeloma. There are many ongoing clinical trials using novel targets and constructs, including bispecific antibodies against BCMA, GPRC5D, and FCRH5. In addition to exploring efficacy, there are ongoing efforts to overcome the resistance to therapy. Antibody-based therapy has improved the outcomes of patients with multiple myeloma and has been incorporated in the standard of care. We expect to see novel targets and constructs that can achieve a deeper and more durable response while minimizing toxicity, as well as better strategies for toxicity management for existing agents. We also expect that antibody-based strategies will be used in earlier lines of therapy in the future.
Collapse
Affiliation(s)
- Hitomi Hosoya
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford, CA, USA
| | - Surbhi Sidana
- Stanford Cancer Institute, Stanford, CA, USA.
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, 300 Pasteur Drive, Room H0101c, Stanford, CA, USA.
| |
Collapse
|
32
|
Targeting Multiple Myeloma through the Biology of Long-Lived Plasma Cells. Cancers (Basel) 2020; 12:cancers12082117. [PMID: 32751699 PMCID: PMC7466116 DOI: 10.3390/cancers12082117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of terminally differentiated bone marrow (BM) resident B lymphocytes known as plasma cells (PC). PC that reside in the bone marrow include a distinct population of long-lived plasma cells (LLPC) that have the capacity to live for very long periods of time (decades in the human population). LLPC biology is critical for understanding MM disease induction and progression because MM shares many of the same extrinsic and intrinsic survival programs as LLPC. Extrinsic survival signals required for LLPC survival include soluble factors and cellular partners in the bone marrow microenvironment. Intrinsic programs that enhance cellular fidelity are also required for LLPC survival including increased autophagy, metabolic fitness, the unfolded protein response (UPR), and enhanced responsiveness to endoplasmic reticulum (ER) stress. Targeting LLPC cell survival mechanisms have led to standard of care treatments for MM including proteasome inhibition (Bortezomib), steroids (Dexamethasone), and immunomodulatory drugs (Lenalidomide). MM patients that relapse often do so by circumventing LLPC survival pathways targeted by treatment. Understanding the mechanisms by which LLPC are able to survive can allow us insight into the treatment of MM, which allows for the enhancement of therapeutic strategies in MM both at diagnosis and upon patient relapse.
Collapse
|
33
|
Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020; 577:561-565. [DOI: 10.1038/s41586-019-1914-8] [Citation(s) in RCA: 698] [Impact Index Per Article: 174.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023]
|
34
|
KSHV/HHV8-positive large B-cell lymphomas and associated diseases: a heterogeneous group of lymphoproliferative processes with significant clinicopathological overlap. Mod Pathol 2020; 33:18-28. [PMID: 31527708 DOI: 10.1038/s41379-019-0365-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 02/08/2023]
Abstract
In this review, we focus on the current understanding of the diagnosis of human herpesvirus 8 (HHV8)-associated lymphoproliferative disorders-a group of entities that range from hyperplastic proliferations to frank lymphomas. These diseases tend to occur in immunodeficient patients, but may occur in immunocompetent individuals as well. In recent years, we have learned of occasional cases with overlapping features among HHV8 entities, such as lesions intermediate between primary effusion lymphoma and HHV8-positive diffuse large B-cell lymphoma, not otherwise specified or cases sharing features of multicentric Castleman disease and germinotropic lymphoproliferative disorder. There is also a significant clinical overlap between these entities. It is important to have a better understanding of the biology of these lesions and to refine diagnostic criteria of these lesions, as the use of immunosuppressive agents to treat a variety of diseases, the expanded use of transplant as a therapeutic modality for a variety of cancers and organ failure patients, and the extended longevity of HIV-positive patients will likely result in an increased incidence of these lymphoproliferative processes in the future.
Collapse
|
35
|
Wu L, Yang Y, Kong L, Bian X, Guo Z, Fu S, Liang F, Li B, Ye J. Comparative transcriptome analysis of the transcriptional heterogeneity in different IgM + cell subsets from peripheral blood of Nile tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2019; 93:612-622. [PMID: 31408730 DOI: 10.1016/j.fsi.2019.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
In teleost fish, IgM+ B cells play important roles in innate and adaptive immunity. Different IgM+ B cells are detected in teleost, named IgMlo and IgMhi B cell subsets, according to the distinct expression levels of membrane IgM (mIgM). However, the study on the heterogeneity in IgM+ B cell subsets remains poorly understood. In this study, the comparative transcriptomic profiles of IgM-, IgMlo and IgMhi from peripheral blood of Nile tilapia (Oreochromis niloticus) were carried out by using RNA-sequencing technique. A total of 6045 and 5470 differentially expressed genes (DEGs) were detected in IgMlo and IgMhi cells, respectively, as compared with IgM- lymphocytes, whereas 3835 genes were differentially expressed when IgMlo compared to IgMhi cells. Analysis of the KEGG database indicated that the DEGs were enriched in immune system categories and signaling transduction and interaction in IgM- vs IgMhi, IgM- vs IgMlo and IgMlo vs IgMhi. Comparatively, in IgMlo vs IgMhi, GO enrichment analysis indicated that the DEGs enriched in nucleic acid binding transcription factor activity. Analysis of crucial transcription factors for B cell differentiation indicated that IgMlo and IgMhi cell clusters belonged to the different B cell subsets. The data generated in this study may provide insights into understanding the heterogeneity of IgM+ cells in teleost, and suggest that IgM+ B cells play a crucial role in innate immunity.
Collapse
Affiliation(s)
- Liting Wu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Yanjian Yang
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Linghe Kong
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Xia Bian
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Zheng Guo
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Shengli Fu
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Fang Liang
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Bingxi Li
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China
| | - Jianmin Ye
- Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research, Center for Environmentally-Friendly Aquaculture, Guangzhou, 510631, PR China.
| |
Collapse
|
36
|
CD19-positive antibody-secreting cells provide immune memory. Blood Adv 2019; 2:3163-3176. [PMID: 30478153 DOI: 10.1182/bloodadvances.2017015172] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
Long-lived antibody-secreting cells (ASCs) are critical for the maintenance of humoral immunity through the continued production of antibodies specific for previously encountered pathogen or vaccine antigens. Recent reports describing humoral immune memory have suggested the importance of long-lived CD19- bone marrow (BM) ASCs, which secrete antibodies recognizing previously encountered vaccine antigens. However, these reports do not agree upon the unique contribution of the CD19+ BM ASC subset toward humoral immunity. Here, we found both CD19+ and negative ASCs from human BM were similar in functional capacity to react to a number of vaccine antigens via ELISpot assays. The CD19+ cells were the predominant ASC population found in lymphoid tissues, and unlike the CD19- ASCs, which were found only in spleen and BM, the CD19+ ASCs were found in tonsil and blood. CD19+ ASCs from the BM, spleen, and tonsil were capable of recognizing polio vaccine antigens, indicating the CD19+ ASC cells play a novel role in long-lasting immune defense. Comparative gene expression analysis indicated CD19+ and negative BM ASCs differed significantly by only 14 distinct messenger RNAs and exhibited similar gene expression for cell cycle, autophagy, and apoptosis control necessary for long life. In addition, we show identical CDR-H3 sequences found on both BM ASC subsets, indicating a shared developmental path. Together, these results provide novel insight for the distribution, function, genetic regulation, and development of long-lived ASCs and may not only impact improved cell therapies but also enhance strategies for vaccine development.
Collapse
|
37
|
Wu L, Kong L, Yang Y, Bian X, Wu S, Li B, Yin X, Mu L, Li J, Ye J. Effects of Cell Differentiation on the Phagocytic Activities of IgM + B Cells in a Teleost Fish. Front Immunol 2019; 10:2225. [PMID: 31608055 PMCID: PMC6761302 DOI: 10.3389/fimmu.2019.02225] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 09/02/2019] [Indexed: 11/29/2022] Open
Abstract
Teleost B cells have phagocytic activities for ingesting particulate antigens, such as bacteria, in addition to the functional secretion of immunoglobulins (Igs). In the present study, the phagocytic activities of IgM+ B cells under various differentiational conditions residing in peripheral blood leukocytes were investigated in a teleost fish Nile tilapia (Oreochromis niloticus). The IgM+ B cells were recognized as IgMlo or IgMhi subsets based on their membrane IgM (mIgM) levels. The mIgM, secreted IgM (sIgM), major histocompatibility complex class II and reactive oxygen species were detected. Expressions of transcription factors (Pax5 and Blimp-1) and B cell signaling molecules (CD79a, CD79b, BLNK, and LYN) suggested that IgMlo B cells were resembling as plasma-like cells and IgMhi resembling as naïve/mature B cells, respectively. Analysis of phagocytic activities demonstrated that both IgMlo and IgMhi B cells have a similar phagocytic ability (phagocytosis percentage); however, the phagocytic capacity [phagocytic index and the mean fluorescence intensity (MFI)] of IgMhi B cells was significantly higher than that of IgMlo B cells. Taken together, the results indicated that B cell differentiation may cause the decrease of phagocytic capacity but not phagocytic ability of phagocytic IgM+ B cells in teleost. The finding may provide an evolutionary evidence for understanding the greater specialization of the B cell in more sophisticated adaptive humoral immunity, by decreasing phagocytic activity in order to contribute its function more specifically into antibody-secreting.
Collapse
Affiliation(s)
- Liting Wu
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Linghe Kong
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yanjian Yang
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xia Bian
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Siwei Wu
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Bingxi Li
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiaoxue Yin
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Liangliang Mu
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jun Li
- School of Biological Sciences, Lake Superior State University, Sault Ste. Marie, MI, United States
| | - Jianmin Ye
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
38
|
The Unsolved Puzzle of c-Rel in B Cell Lymphoma. Cancers (Basel) 2019; 11:cancers11070941. [PMID: 31277480 PMCID: PMC6678315 DOI: 10.3390/cancers11070941] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 01/04/2023] Open
Abstract
Aberrant constitutive activation of Rel/NF-κB transcription factors is a hallmark of numerous cancers. Of the five Rel family members, c-Rel has the strongest direct links to tumorigenesis. c-Rel is the only member that can malignantly transform lymphoid cells in vitro. Furthermore, c-Rel is implicated in human B cell lymphoma through the frequent occurrence of REL gene locus gains and amplifications. In normal physiology, high c-Rel expression predominates in the hematopoietic lineage and a diverse range of stimuli can trigger enhanced expression and activation of c-Rel. Both expression and activation of c-Rel are tightly regulated on multiple levels, indicating the necessity to keep its functions under control. In this review we meta-analyze and integrate studies reporting gene locus aberrations to provide an overview on the frequency of REL gains in human B cell lymphoma subtypes, namely follicular lymphoma, diffuse large B cell lymphoma, primary mediastinal B cell lymphoma, and classical Hodgkin lymphoma. We also summarize current knowledge on c-Rel expression and protein localization in these human B cell lymphomas and discuss the co-amplification of BCL11A with REL. In addition, we highlight and illustrate key pathways of c-Rel activation and regulation with a specific focus on B cell biology.
Collapse
|
39
|
Zumaquero E, Stone SL, Scharer CD, Jenks SA, Nellore A, Mousseau B, Rosal-Vela A, Botta D, Bradley JE, Wojciechowski W, Ptacek T, Danila MI, Edberg JC, Bridges SL, Kimberly RP, Chatham WW, Schoeb TR, Rosenberg AF, Boss JM, Sanz I, Lund FE. IFNγ induces epigenetic programming of human T-bet hi B cells and promotes TLR7/8 and IL-21 induced differentiation. eLife 2019; 8:e41641. [PMID: 31090539 PMCID: PMC6544433 DOI: 10.7554/elife.41641] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 05/10/2019] [Indexed: 12/24/2022] Open
Abstract
Although B cells expressing the IFNγR or the IFNγ-inducible transcription factor T-bet promote autoimmunity in Systemic Lupus Erythematosus (SLE)-prone mouse models, the role for IFNγ signaling in human antibody responses is unknown. We show that elevated levels of IFNγ in SLE patients correlate with expansion of the T-bet expressing IgDnegCD27negCD11c+CXCR5neg (DN2) pre-antibody secreting cell (pre-ASC) subset. We demonstrate that naïve B cells form T-bethi pre-ASCs following stimulation with either Th1 cells or with IFNγ, IL-2, anti-Ig and TLR7/8 ligand and that IL-21 dependent ASC formation is significantly enhanced by IFNγ or IFNγ-producing T cells. IFNγ promotes ASC development by synergizing with IL-2 and TLR7/8 ligands to induce genome-wide epigenetic reprogramming of B cells, which results in increased chromatin accessibility surrounding IRF4 and BLIMP1 binding motifs and epigenetic remodeling of IL21R and PRDM1 loci. Finally, we show that IFNγ signals poise B cells to differentiate by increasing their responsiveness to IL-21.
Collapse
Affiliation(s)
- Esther Zumaquero
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Sara L Stone
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Division of RheumatologyEmory UniversityAtlantaUnited States
| | - Scott A Jenks
- Department of Medicine, Division of RheumatologyEmory UniversityAtlantaUnited States
| | - Anoma Nellore
- Department of Medicine, Division of Infectious DiseaseThe University of Alabama at BirminghamBirminghamUnited States
| | - Betty Mousseau
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Antonio Rosal-Vela
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Davide Botta
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
| | - John E Bradley
- Department of Medicine, Division of Clinical Immunology and RheumatologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Wojciech Wojciechowski
- Center for Pediatric Biomedical Research, Flow Cytometry Shared Resource LaboratoryUniversity of Rochester School of Medicine and DentistryRochesterUnited States
| | - Travis Ptacek
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
- Informatics Group, Center for Clinical and Translational ScienceThe University of Alabama at BirminghamBirminghamUnited States
| | - Maria I Danila
- Department of Medicine, Division of Clinical Immunology and RheumatologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Jeffrey C Edberg
- Department of Medicine, Division of Clinical Immunology and RheumatologyThe University of Alabama at BirminghamBirminghamUnited States
| | - S Louis Bridges
- Department of Medicine, Division of Clinical Immunology and RheumatologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Robert P Kimberly
- Department of Medicine, Division of Clinical Immunology and RheumatologyThe University of Alabama at BirminghamBirminghamUnited States
| | - W Winn Chatham
- Department of Medicine, Division of Clinical Immunology and RheumatologyThe University of Alabama at BirminghamBirminghamUnited States
| | - Trenton R Schoeb
- Department of Genetics, Animal Resources ProgramThe University of Alabama at BirminghamBirminghamUnited States
| | - Alexander F Rosenberg
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
- The Informatics InstituteThe University of Alabama at BirminghamBirminghamUnited States
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Division of RheumatologyEmory UniversityAtlantaUnited States
| | - Ignacio Sanz
- Department of Medicine, Division of RheumatologyEmory UniversityAtlantaUnited States
| | - Frances E Lund
- Department of MicrobiologyThe University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
40
|
Lightman SM, Utley A, Lee KP. Survival of Long-Lived Plasma Cells (LLPC): Piecing Together the Puzzle. Front Immunol 2019; 10:965. [PMID: 31130955 PMCID: PMC6510054 DOI: 10.3389/fimmu.2019.00965] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022] Open
Abstract
Durable humoral immunity is dependent upon the generation of antigen-specific antibody titers, produced by non-proliferating bone marrow resident long-lived plasma cells (LLPC). Longevity is the hallmark of LLPC, but why and how they survive and function for years after antigen exposure is only beginning to be understood. LLPC are not intrinsically long-lived; they require continuous signals from the LLPC niche to survive. Signals unique to LLPC survival (vs. PC survival in general) most notably include those that upregulate the anti-apoptotic factor Mcl-1 and activation of the CD28 receptor expressed on LLPC. Other potential factors include expression of BCMA, upregulation of the transcription factor ZBTB20, and upregulation of the enzyme ENPP1. Metabolic fitness is another key component of LLPC longevity, facilitating the diversion of glucose to generate pyruvate during times of stress to facilitate long term survival. A third major component of LLPC survival is the microenvironment/LLPC niche itself. Cellular partners such as stromal cells, dendritic cells, and T regulatory cells establish a niche for LLPC and drive survival signaling by expressing ligands such as CD80/CD86 for CD28 and producing soluble and stromal factors that contribute to LLPC longevity. These findings have led to the current paradigm wherein both intrinsic and extrinsic mechanisms are required for the survival of LLPC. Here we outline this diverse network of signals and highlight the mechanisms thought to regulate and promote the survival of LLPC. Understanding this network of signals has direct implications in increasing our basic understanding of plasma cell biology, but also in vaccine and therapeutic drug development to address the pathologies that can arise from this subset.
Collapse
Affiliation(s)
- Shivana M Lightman
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Adam Utley
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
41
|
Loeffler-Wirth H, Kreuz M, Hopp L, Arakelyan A, Haake A, Cogliatti SB, Feller AC, Hansmann ML, Lenze D, Möller P, Müller-Hermelink HK, Fortenbacher E, Willscher E, Ott G, Rosenwald A, Pott C, Schwaenen C, Trautmann H, Wessendorf S, Stein H, Szczepanowski M, Trümper L, Hummel M, Klapper W, Siebert R, Loeffler M, Binder H. A modular transcriptome map of mature B cell lymphomas. Genome Med 2019; 11:27. [PMID: 31039827 PMCID: PMC6492344 DOI: 10.1186/s13073-019-0637-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/04/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Germinal center-derived B cell lymphomas are tumors of the lymphoid tissues representing one of the most heterogeneous malignancies. Here we characterize the variety of transcriptomic phenotypes of this disease based on 873 biopsy specimens collected in the German Cancer Aid MMML (Molecular Mechanisms in Malignant Lymphoma) consortium. They include diffuse large B cell lymphoma (DLBCL), follicular lymphoma (FL), Burkitt's lymphoma, mixed FL/DLBCL lymphomas, primary mediastinal large B cell lymphoma, multiple myeloma, IRF4-rearranged large cell lymphoma, MYC-negative Burkitt-like lymphoma with chr. 11q aberration and mantle cell lymphoma. METHODS We apply self-organizing map (SOM) machine learning to microarray-derived expression data to generate a holistic view on the transcriptome landscape of lymphomas, to describe the multidimensional nature of gene regulation and to pursue a modular view on co-expression. Expression data were complemented by pathological, genetic and clinical characteristics. RESULTS We present a transcriptome map of B cell lymphomas that allows visual comparison between the SOM portraits of different lymphoma strata and individual cases. It decomposes into one dozen modules of co-expressed genes related to different functional categories, to genetic defects and to the pathogenesis of lymphomas. On a molecular level, this disease rather forms a continuum of expression states than clearly separated phenotypes. We introduced the concept of combinatorial pattern types (PATs) that stratifies the lymphomas into nine PAT groups and, on a coarser level, into five prominent cancer hallmark types with proliferation, inflammation and stroma signatures. Inflammation signatures in combination with healthy B cell and tonsil characteristics associate with better overall survival rates, while proliferation in combination with inflammation and plasma cell characteristics worsens it. A phenotypic similarity tree is presented that reveals possible progression paths along the transcriptional dimensions. Our analysis provided a novel look on the transition range between FL and DLBCL, on DLBCL with poor prognosis showing expression patterns resembling that of Burkitt's lymphoma and particularly on 'double-hit' MYC and BCL2 transformed lymphomas. CONCLUSIONS The transcriptome map provides a tool that aggregates, refines and visualizes the data collected in the MMML study and interprets them in the light of previous knowledge to provide orientation and support in current and future studies on lymphomas and on other cancer entities.
Collapse
Affiliation(s)
- Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - Markus Kreuz
- Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - Lydia Hopp
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - Arsen Arakelyan
- Group of Bioinformatics, Institute of Molecular Biology, National Academy of Sciences, 7 Hasratyan str, 0014 Yerevan, Armenia
| | - Andrea Haake
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
| | - Sergio B. Cogliatti
- Institute of Pathology, Kantonal Hospital St. Gallen, Rorschacher Str. 95, 9007 St. Gallen, Switzerland
| | - Alfred C. Feller
- Hematopathology Lübeck, Maria-Goeppert-Str. 9a, 23562 Lübeck, Germany
| | - Martin-Leo Hansmann
- Institute of Pathology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Dido Lenze
- AstraZeneca, Tinsdaler Weg 183, 22880 Wedel, Germany
| | - Peter Möller
- Institute of Pathology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | | | - Erik Fortenbacher
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - Edith Willscher
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - German Ott
- Department of Pathology, Robert-Bosch-Hospital, Auerbachstr. 110, 70376 Stuttgart, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
| | - Carsten Schwaenen
- Ortenau Hospital Offenburg-Gengenbach, Ebertpl. 12, 77654 Offenburg, Germany
- Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Heiko Trautmann
- Second Medical Department, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
| | - Swen Wessendorf
- Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Hospital Esslingen, Hirschlandstr. 97, 73730 Esslingen a. N, Germany
| | - Harald Stein
- Pathodiagnostik, Komturstr. 58-62, 12099 Berlin, Germany
| | - Monika Szczepanowski
- Second Medical Department, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
| | - Lorenz Trümper
- Department of Hematology and Oncology, Georg-August University, Robert-Koch-Str. 42, 37077 Göttingen, Germany
| | - Michael Hummel
- Institute of Pathology, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Wolfram Klapper
- Hematopathology Section, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
- Institute of Human Genetics, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Markus Loeffler
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
| | - for the German Cancer Aid consortium Molecular Mechanisms for Malignant Lymphoma
- Interdisciplinary Centre for Bioinformatics, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, Härtelstr. 16–18, 04107 Leipzig, Germany
- Group of Bioinformatics, Institute of Molecular Biology, National Academy of Sciences, 7 Hasratyan str, 0014 Yerevan, Armenia
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
- Institute of Pathology, Kantonal Hospital St. Gallen, Rorschacher Str. 95, 9007 St. Gallen, Switzerland
- Hematopathology Lübeck, Maria-Goeppert-Str. 9a, 23562 Lübeck, Germany
- Institute of Pathology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- AstraZeneca, Tinsdaler Weg 183, 22880 Wedel, Germany
- Institute of Pathology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Institute of Pathology, University Hospital Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Department of Pathology, Robert-Bosch-Hospital, Auerbachstr. 110, 70376 Stuttgart, Germany
- Second Medical Department, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
- Ortenau Hospital Offenburg-Gengenbach, Ebertpl. 12, 77654 Offenburg, Germany
- Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Hospital Esslingen, Hirschlandstr. 97, 73730 Esslingen a. N, Germany
- Pathodiagnostik, Komturstr. 58-62, 12099 Berlin, Germany
- Department of Hematology and Oncology, Georg-August University, Robert-Koch-Str. 42, 37077 Göttingen, Germany
- Institute of Pathology, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
- Hematopathology Section, University Hospital Schleswig-Holstein, Arnold-Heller Str. 3, 24105 Kiel, Germany
- Institute of Human Genetics, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| |
Collapse
|
42
|
Mahendra A, Yang X, Abnouf S, Adolacion JRT, Park D, Soomro S, Roszik J, Coarfa C, Romain G, Wanzeck K, Bridges SL, Aggarwal A, Qiu P, Agarwal SK, Mohan C, Varadarajan N. Beyond Autoantibodies: Biologic Roles of Human Autoreactive B Cells in Rheumatoid Arthritis Revealed by RNA-Sequencing. Arthritis Rheumatol 2019; 71:529-541. [PMID: 30407753 PMCID: PMC6741783 DOI: 10.1002/art.40772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 11/01/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To obtain the comprehensive transcriptome profile of human citrulline-specific B cells from patients with rheumatoid arthritis (RA). METHODS Citrulline- and hemagglutinin-specific B cells were sorted by flow cytometry using peptide-streptavidin conjugates from the peripheral blood of RA patients and healthy individuals. The transcriptome profile of the sorted cells was obtained by RNA-sequencing, and expression of key protein molecules was evaluated by aptamer-based SOMAscan assay and flow cytometry. The ability of these proteins to effect differentiation of osteoclasts and proliferation and migration of synoviocytes was examined by in vitro functional assays. RESULTS Citrulline-specific B cells, in comparison to citrulline-negative B cells, from patients with RA differentially expressed the interleukin-15 receptor α (IL-15Rα) gene as well as genes related to protein citrullination and cyclic AMP signaling. In analyses of an independent cohort of cyclic citrullinated peptide-seropositive RA patients, the expression of IL-15Rα protein was enriched in citrulline-specific B cells from the patients' peripheral blood, and surprisingly, all B cells from RA patients were capable of producing the epidermal growth factor ligand amphiregulin (AREG). Production of AREG directly led to increased migration and proliferation of fibroblast-like synoviocytes, and, in combination with anti-citrullinated protein antibodies, led to the increased differentiation of osteoclasts. CONCLUSION To the best of our knowledge, this is the first study to document the whole transcriptome profile of autoreactive B cells in any autoimmune disease. These data identify several genes and pathways that may be targeted by repurposing several US Food and Drug Administration-approved drugs, and could serve as the foundation for the comparative assessment of B cell profiles in other autoimmune diseases.
Collapse
Affiliation(s)
- Ankit Mahendra
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Xingyu Yang
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Shaza Abnouf
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Jay R T Adolacion
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Daechan Park
- Department of Biological Sciences, College of Natural Sciences, Ajou University, Republic of Korea
| | - Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, TX
| | - Jason Roszik
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Cristian Coarfa
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX
| | - Gabrielle Romain
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| | - Keith Wanzeck
- Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - S. Louis Bridges
- Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - Amita Aggarwal
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Immunology, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX
| | - Navin Varadarajan
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX
| |
Collapse
|
43
|
Leibler C, Thiolat A, Elsner RA, El Karoui K, Samson C, Grimbert P. Costimulatory blockade molecules and B-cell-mediated immune response: current knowledge and perspectives. Kidney Int 2019; 95:774-786. [PMID: 30711200 DOI: 10.1016/j.kint.2018.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
There is an urgent need for therapeutic agents that target humoral alloimmunity in solid organ transplantation. This includes sensitized patients with preformed donor-specific human leukocyte antigen antibodies and patients who develop de novo donor-specific antibodies, both of which are associated with acute and chronic antibody-mediated rejection and allograft loss. In the last decade, both experimental and clinical studies highlighted the major impact of costimulation molecules in the control of immune responses both in the field of transplantation and autoimmune disease. Although these molecules have been initially developed to control the early steps of T-cell activation, recent evidence also supports their influence at several steps of the humoral response. In this review, we aim to provide an overview of the current knowledge of the effects of costimulatory blockade agents on humoral responses in both autoimmune and allogeneic contexts. We first present the effects of costimulatory molecules on the different steps of alloantibody production. We then summarize mechanisms and clinical results observed using cytotoxic T lymphocyte antigen-4 (CTLA4)-Ig molecules both in transplantation and autoimmunity. Finally, we present the potential interest and implications of other costimulatory family members as therapeutic targets, with emphasis on combinatorial approaches, for the optimal control of the alloantigen-specific humoral response.
Collapse
Affiliation(s)
- Claire Leibler
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Allan Thiolat
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Rebecca A Elsner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Khalil El Karoui
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Chloe Samson
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Philippe Grimbert
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France.
| |
Collapse
|
44
|
Wu L, Fu S, Yin X, Leng W, Guo Z, Wang A, Ye J. Affinity maturation occurs in channel catfish (Ictalurus punctaus) following immunization with a T-cell dependent antigen. FISH & SHELLFISH IMMUNOLOGY 2019; 84:781-786. [PMID: 30393175 DOI: 10.1016/j.fsi.2018.10.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/13/2018] [Accepted: 10/22/2018] [Indexed: 06/08/2023]
Abstract
Affinity maturation of the antibody response, a process of antibody affinity increasing over response, is one of the key features of the mammalian immune system. However, the process is incompletely understood in teleost, including channel catfish (Ictalurus punctaus). In this study, IgM affinity maturation in channel catfish was investigated by estimating the kinetics of antibody affinity using ELISA and ELISPOT assays. Fish were immunized with a T-cell dependent antigen (TNP-KLH), and individual serum IgM antibody titers and affinities, and IgM+ antibody-secreting cells (ASCs) in peripheral blood were analyzed over a period of 14 weeks. A detectable serum anti-TNP response developed by 2-weeks post-immunization, and the maximal antibody production was observed by 6-weeks post-immunization. The average affinity of anti-TNP serum antibody increased consistently and reached the maximum by 10-weeks post-immunization. The increase of antibody affinity beyond the point of optimal antibody titer revealed that the affinity maturation of IgM antibody response occurred in channel catfish. Dissection of dynamics of individual affinity subpopulations indicated that a significant proportion of low affinity subpopulations appeared at early response, and high affinity subpopulations appeared predominantly at later, resulting in a 100-fold increase in affinity over response. Additional, TNP+ IgM+ ASCs was detected by 2-weeks post-immunization and achieved the maximal number by 6-weeks post-immunization. Using an inhibition ELISPOT assay, the findings of a consistent increase in the average affinity of secreted IgM antibody by peripheral blood ASCs, as the immune response progressed, confirmed the occurrence of the affinity maturation. Taken together, the results of this study indicated that affinity maturation occurred in channel catfish following immunization with a TD antigen TNP-KLH.
Collapse
Affiliation(s)
- Liting Wu
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China
| | - Shengli Fu
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China
| | - Xiaoxue Yin
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China
| | - Wenna Leng
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China
| | - Zheng Guo
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China
| | - Anli Wang
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China
| | - Jianmin Ye
- School of Life Sciences, South China Normal University, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou, 510631, PR China.
| |
Collapse
|
45
|
Wei X, Li B, Wu L, Yin X, Zhong X, Li Y, Wang Y, Guo Z, Ye J. Interleukin-6 gets involved in response to bacterial infection and promotes antibody production in Nile tilapia (Oreochromis niloticus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 89:141-151. [PMID: 30142358 DOI: 10.1016/j.dci.2018.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/16/2018] [Accepted: 08/16/2018] [Indexed: 06/08/2023]
Abstract
Interleukin 6 (IL-6), a pleiotropic cytokine, plays an important role in humoral immune response, not only inducing the differentiation of B cells into plasma cells, but also promoting antibody-secreting cells (ASCs) to produce antibodies. In this study, Nile tilapia (Oreochromis niloticus) IL-6 (OnIL-6) was identified and characterized at expression level in response to bacterial infection and promotion of antibody production. The open reading frame of OnIL-6 ORF is consisted of 663 bp encoding a polypeptide of 220 amino acids. The deduced OnIL-6 protein contained an IL-6/G-CSF family signature, two conserved cysteine, and four α-helix bundles, which was highly homologous to other species. Spatial mRNA expression analysis revealed that the highest expression of OnIL-6 was observed in the thymus. After in vivo challenges of lipopolysaccharide (LPS) and Streptococcus agalactia (S. agalactiae), OnIL-6 expressions were significantly up-regulated in head kidney and spleen. The similar up-regulation of OnIL-6 was observed in the head kidney and spleen leukocytes in vitro stimulation with LPS and S. agalactiae. In addition, inducement with the recombinant OnIL-6 ((r)OnIL-6) in vitro caused significant increases in expressions of both sIgM and mIgM. Moreover, the (r)OnIL-6 stimulation enhanced the secretion of sIgM (more especially in P50 plasma-like B cells) and the production of mIgM in P60 and P70 B cell subsets (resting B cells, activated B cells and plasmablast-like B cells) in vitro. Taken together, this study indicated that OnIL-6 might be involved in host defense against bacterial infection and promote the production of antibody in Nile tilapia.
Collapse
Affiliation(s)
- Xiufang Wei
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Bingxi Li
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Liting Wu
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Xiaoxue Yin
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Xiaofang Zhong
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Yuan Li
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Yuhong Wang
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Zheng Guo
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China
| | - Jianmin Ye
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, College of Life Sciences, South China Normal University, Guangdong, 510631, PR China.
| |
Collapse
|
46
|
Raeven RHM, van Riet E, Meiring HD, Metz B, Kersten GFA. Systems vaccinology and big data in the vaccine development chain. Immunology 2018; 156:33-46. [PMID: 30317555 PMCID: PMC6283655 DOI: 10.1111/imm.13012] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023] Open
Abstract
Systems vaccinology has proven a fascinating development in the last decade. Where traditionally vaccine development has been dominated by trial and error, systems vaccinology is a tool that provides novel and comprehensive understanding if properly used. Data sets retrieved from systems‐based studies endorse rational design and effective development of safe and efficacious vaccines. In this review we first describe different omics‐techniques that form the pillars of systems vaccinology. In the second part, the application of systems vaccinology in the different stages of vaccine development is described. Overall, this review shows that systems vaccinology has become an important tool anywhere in the vaccine development chain.
Collapse
Affiliation(s)
- René H M Raeven
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Elly van Riet
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Hugo D Meiring
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Bernard Metz
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands
| | - Gideon F A Kersten
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands.,Leiden Academic Center for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| |
Collapse
|
47
|
Aberrant Activation of NF-κB Signalling in Aggressive Lymphoid Malignancies. Cells 2018; 7:cells7110189. [PMID: 30380749 PMCID: PMC6262606 DOI: 10.3390/cells7110189] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/28/2022] Open
Abstract
Lymphoid malignancies frequently harbor genetic mutations leading to aberrant activation of nuclear factor-κB (NF-κB) signaling; in normal cells, this pathway has important roles in the control of cell growth, survival, stress responses, and inflammation. Malignancies with mutations in NF-κB pathway components can derive from all cell stages of mature B-cell development; however, aberrant NF-κB activity is particularly prevalent in aggressive subtypes of non-Hodgkin lymphoma and myeloma. NF-κB activation is mediated by two separate pathways, the canonical and alternative pathway, and five downstream transcription factor subunits. Recent findings implicate a predominant role for distinct NF-κB pathways and subunits in certain lymphoma subtypes and myeloma; findings which are complemented by the realization that individual NF-κB subunits can have unique, non-redundant biological roles in the putative tumor precursor cells, including activated B cells, germinal center B cells and plasma cells. The knowledge gained from these studies may be exploited for the development of therapeutic strategies to inhibit aberrant NF-κB activity at the level of the transcription-factor subunits and their target genes, as global inhibition of the pathway is toxic. Here, we provide an overview on the role of aberrant NF-κB activation in aggressive lymphoid malignancies and discuss the potential importance of individual NF-κB subunits in the pathogenesis of tumor subtypes.
Collapse
|
48
|
Wong KY, Baron R, Seldon TA, Jones ML, Rice AM, Munster DJ. CD83 Antibody Inhibits Human B Cell Responses to Antigen as well as Dendritic Cell-Mediated CD4 T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:3383-3396. [PMID: 29643191 DOI: 10.4049/jimmunol.1700064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/20/2018] [Indexed: 01/11/2023]
Abstract
Anti-CD83 Ab capable of Ab-dependent cellular cytotoxicity can deplete activated CD83+ human dendritic cells, thereby inhibiting CD4 T cell-mediated acute graft-versus-host disease. As CD83 is also expressed on the surface of activated B lymphocytes, we hypothesized that anti-CD83 would also inhibit B cell responses to stimulation. We found that anti-CD83 inhibited total IgM and IgG production in vitro by allostimulated human PBMC. Also, Ag-specific Ab responses to immunization of SCID mice xenografted with human PBMC were inhibited by anti-CD83 treatment. This inhibition occurred without depletion of all human B cells because anti-CD83 lysed activated CD83+ B cells by Ab-dependent cellular cytotoxicity and spared resting (CD83-) B cells. In cultured human PBMC, anti-CD83 inhibited tetanus toxoid-stimulated B cell proliferation and concomitant dendritic cell-mediated CD4 T cell proliferation and expression of IFN-γ and IL-17A, with minimal losses of B cells (<20%). In contrast, the anti-CD20 mAb rituximab depleted >80% of B cells but had no effect on CD4 T cell proliferation and cytokine expression. By virtue of the ability of anti-CD83 to selectively deplete activated, but not resting, B cells and dendritic cells, with the latter reducing CD4 T cell responses, anti-CD83 may be clinically useful in autoimmunity and transplantation. Advantages might include inhibited expansion of autoantigen- or alloantigen-specific B cells and CD4 T cells, thus preventing further production of pathogenic Abs and inflammatory cytokines while preserving protective memory and regulatory cells.
Collapse
Affiliation(s)
- Kuan Y Wong
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - Rebecca Baron
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - Therese A Seldon
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alison M Rice
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - David J Munster
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| |
Collapse
|
49
|
Malkiel S, Barlev AN, Atisha-Fregoso Y, Suurmond J, Diamond B. Plasma Cell Differentiation Pathways in Systemic Lupus Erythematosus. Front Immunol 2018; 9:427. [PMID: 29556239 PMCID: PMC5845388 DOI: 10.3389/fimmu.2018.00427] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/16/2018] [Indexed: 01/20/2023] Open
Abstract
Plasma cells (PCs) are responsible for the production of protective antibodies against infectious agents but they also produce pathogenic antibodies in autoimmune diseases, such as systemic lupus erythematosus (SLE). Traditionally, high affinity IgG autoantibodies are thought to arise through germinal center (GC) responses. However, class switching and somatic hypermutation can occur in extrafollicular (EF) locations, and this pathway has also been implicated in SLE. The pathway from which PCs originate may determine several characteristics, such as PC lifespan and sensitivity to therapeutics. Although both GC and EF responses have been implicated in SLE, we hypothesize that one of these pathways dominates in each individual patient and genetic risk factors may drive this predominance. While it will be important to distinguish polymorphisms that contribute to a GC-driven or EF B cell response to develop targeted treatments, the challenge will be not only to identify the differentiation pathway but the molecular mechanisms involved. In B cells, this task is complicated by the cross-talk between the B cell receptor, toll-like receptors (TLR), and cytokine signaling molecules, which contribute to both GC and EF responses. While risk variants that affect the function of dendritic cells and T follicular helper cells are likely to primarily influence GC responses, it will be important to discover whether some risk variants in the interferon and TLR pathways preferentially influence EF responses. Identifying the pathways of autoreactive PC differentiation in SLE may help us to understand patient heterogeneity and thereby guide precision therapy.
Collapse
Affiliation(s)
- Susan Malkiel
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Ashley N Barlev
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Yemil Atisha-Fregoso
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States.,Tecnologico de Monterrey, Monterrey, Mexico
| | - Jolien Suurmond
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Betty Diamond
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| |
Collapse
|
50
|
Abstract
The two ligands B cell-activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL) and the three receptors BAFF receptor (BAFF-R), transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI), and B cell maturation antigen (BCMA) are members of the "BAFF system molecules." BAFF system molecules are primarily involved in B cell homeostasis. The relevance of BAFF system molecules in host responses to microbial assaults has been investigated in clinical studies and in mice deficient for each of these molecules. Many microbial products modulate the expression of these molecules. Data from clinical studies suggest a correlation between increased expression levels of BAFF system molecules and elevated B cell responses. Depending on the pathogen, heightened B cell responses may strengthen the host response or promote susceptibility. Whereas pathogen-mediated increases in the expression levels of the ligands and/or the receptors appear to promote microbial clearance, certain pathogens have evolved to ablate B cell responses by suppressing the expression of TACI and/or BAFF-R on B cells. Other than its well-established role in B cell responses, the TACI-mediated activation of macrophages is also implicated in resistance to intracellular pathogens. An improved understanding of the role that BAFF system molecules play in infection may assist in devising novel strategies for vaccine development.
Collapse
Affiliation(s)
- Jiro Sakai
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|