1
|
Kim N, Lee J, Kim J, Kim Y, Cho KH. Canalizing kernel for cell fate determination. Brief Bioinform 2024; 25:bbae406. [PMID: 39171985 PMCID: PMC11339868 DOI: 10.1093/bib/bbae406] [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: 03/07/2024] [Revised: 07/14/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
The tendency for cell fate to be robust to most perturbations, yet sensitive to certain perturbations raises intriguing questions about the existence of a key path within the underlying molecular network that critically determines distinct cell fates. Reprogramming and trans-differentiation clearly show examples of cell fate change by regulating only a few or even a single molecular switch. However, it is still unknown how to identify such a switch, called a master regulator, and how cell fate is determined by its regulation. Here, we present CAESAR, a computational framework that can systematically identify master regulators and unravel the resulting canalizing kernel, a key substructure of interconnected feedbacks that is critical for cell fate determination. We demonstrate that CAESAR can successfully predict reprogramming factors for de-differentiation into mouse embryonic stem cells and trans-differentiation of hematopoietic stem cells, while unveiling the underlying essential mechanism through the canalizing kernel. CAESAR provides a system-level understanding of how complex molecular networks determine cell fates.
Collapse
Affiliation(s)
- Namhee Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jonghoon Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jongwan Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yunseong Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
2
|
Liu S, Liu ZC, Zhang MY, Wang SJ, Pan M, Ji P, Zhu C, Lin P, Wang Y. ICAM-1 mediated cell-cell adhesion exerts dual roles on human B cell differentiation and IgG production. iScience 2023; 26:108505. [PMID: 38162034 PMCID: PMC10755720 DOI: 10.1016/j.isci.2023.108505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 01/28/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1) plays prominent roles in mediating cell-cell adhesion which also facilitates B cell activation and differentiation with the help from CD4+ T cells. Here, we have reported a unique phenomenon that increased ICAM-1 on purified human CD4+ T cells upon anti-CD3/CD28 stimulation enhanced CD4+ T-B cell adhesion whereas induced less B cell differentiation and IgG production. This was largely due to increased PD-1 expression on CD19hi B cells after coculturing with hyperactivated CD4+ T cells. Consequently, ICAM-1 blockade during CD4+ T cell-B cell coculture promoted IgG production with the activation of ERK1/2 and Blimp-1/IRF4 upregulation. Consistently, CD4+ T cells from moderate-to-severe SLE patients with high ICAM-1 expression mediated less IgG production after T-B coculture. Therefore, ICAM-1-mediated human CD4+ T-B cell adhesion provides dual roles on B cell differentiation and IgG production partially depending on expression levels of PD-1 on B cells, supporting cell adhesion and subsequent PD-1 induction as an alternative intrinsic checkpoint for B cell differentiation.
Collapse
Affiliation(s)
- Shuai Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Diagnostic Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhi-cui Liu
- Department of Dermatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Mei-yu Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shu-jun Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Meng Pan
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ping Ji
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ping Lin
- Department of Diagnostic Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Institute of Virology, Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| |
Collapse
|
3
|
Zimmerman JA, Fang M, Pufall MA. PI3Kδ inhibition potentiates glucocorticoids in B-lymphoblastic leukemia by decreasing receptor phosphorylation and enhancing gene regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.527869. [PMID: 36798391 PMCID: PMC9934697 DOI: 10.1101/2023.02.10.527869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
Glucocorticoids, including dexamethasone and prednisone, are the cornerstone of B-lymphoblastic leukemia (B-ALL) therapy. Because response to glucocorticoids alone predicts overall outcomes for B-ALL, enhancing glucocorticoid potency is a route to improving outcomes. However, systematic toxicities prevent the use of higher dose and more potent glucocorticoids. We therefore took a functional genomic approach to identify targets to enhance glucocorticoid activity specifically in B-ALL cells. Here we show that inhibition of the lymphoid-restricted PI3Kδ, signaling through the RAS/MAPK pathway, enhances both prednisone and dexamethasone activity in almost all ex vivo B-ALL specimens tested. This potentiation is most synergistic at sub-saturating doses of glucocorticoids, approaching the EC50. Potentiation correlates with global enhancement of glucocorticoid-induced gene regulation, including regulation of effector genes that drive B-ALL cell death. Idelalisib reduces phosphorylation of the glucocorticoid receptor (GR) at MAPK1/ERK2 targets S203 and S226, and ablation of these phospho-acceptor sites enhances glucocorticoid potency. We further show that phosphorylation of S226 reduces the affinity of GR for DNA in vitro, which impairs DNA binding. We therefore propose that PI3Kδ inhibition improves glucocorticoid efficacy in B-ALL in part by decreasing GR phosphorylation, increasing DNA binding affinity, and enhancing downstream gene regulation. The overall enhancement of GR function suggests that idelalisib will provide benefit to most patients with B-ALL by improving outcomes for patients whose disease is less responsive to glucocorticoid-based therapy, including high-risk disease, and allowing less toxic glucocorticoid-sparing strategies for patients with standard-risk disease.
Collapse
Affiliation(s)
- Jessica A.O. Zimmerman
- Division of Pediatric Hematology/Oncology, Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Mimi Fang
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Miles A. Pufall
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
4
|
Kojima Y, Kawashima F, Yasuda T, Odaira K, Inagaki Y, Yamada C, Muraki A, Noura M, Okamoto S, Tamura S, Iwamoto E, Sanada M, Matsumura I, Miyazaki Y, Kojima T, Kiyoi H, Tsuzuki S, Hayakawa F. EBF1-JAK2 inhibits the PAX5 function through physical interaction with PAX5 and kinase activity. Int J Hematol 2023:10.1007/s12185-023-03585-z. [PMID: 37149540 DOI: 10.1007/s12185-023-03585-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 05/08/2023]
Abstract
Gene aberrations of B-cell regulators and growth signal components such as the JAK-STAT pathway are frequently found in B-cell acute lymphoblastic leukemia (B-ALL). EBF1 is a B-cell regulator that regulates the expression of PAX5 and co-operates with PAX5 to regulate B-cell differentiation. Here, we analyzed the function of the fusion protein of EBF1 and JAK2, EBF1-JAK2 (E-J). E-J caused constitutive activation of JAK-STAT and MAPK pathways and induced autonomous cell growth in a cytokine-dependent cell line. E-J did not affect the transcriptional activity of EBF1 but inhibited that of PAX5. Both the physical interaction of E-J with PAX5 and kinase activity of E-J were required for E-J to inhibit PAX5 function, although the detailed mechanism of inhibition remains unclear. Importantly, gene set enrichment analysis using the results of our previous RNA-seq data of 323 primary BCR-ABL1-negative ALL samples demonstrated repression of the transcriptional target genes of PAX5 in E-J-positive ALL cells, which suggests that E-J also inhibited PAX5 function in ALL cells. Our results shed new light on the mechanisms of differentiation block by kinase fusion proteins.
Collapse
Affiliation(s)
- Yukino Kojima
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Fumika Kawashima
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Takahiko Yasuda
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Koya Odaira
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Yuichiro Inagaki
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo, Japan
| | - Chiharu Yamada
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Ami Muraki
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Mina Noura
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Shuichi Okamoto
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Shogo Tamura
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
| | - Eisuke Iwamoto
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Masashi Sanada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tetsuhito Kojima
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan
- Aichi Health Promotion Foundation, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Fumihiko Hayakawa
- Division of Cellular and Genetic Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, 461-0047, Japan.
| |
Collapse
|
5
|
Ahmad SM, Bhat SS, Shafi S, Dar MA, Saleem A, Haq Z, Farooq N, Nazir J, Bhat B. Identification of key transcription factors and their functional role involved in Salmonella typhimurium infection in chicken using integrated transcriptome analysis and bioinformatics approach. BMC Genomics 2023; 24:214. [PMID: 37098463 PMCID: PMC10127038 DOI: 10.1186/s12864-023-09315-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/16/2023] [Indexed: 04/27/2023] Open
Abstract
Salmonella enterica serovar typhimurium is the cause of significant morbidity and mortality worldwide that causes economic losses to poultry and is able to cause infection in humans. Indigenous chicken breeds are a potential source of animal protein and have the added advantage of being disease resistant. An indigenous chicken, Kashmir favorella and commercial broiler were selected for understanding the mechanism of disease resistance. Following infection in Kashmir favorella, three differentially expressed genes Nuclear Factor Kappa B (NF-κB1), Forkhead Box Protein O3 (FOXO3) and Paired box 5 (Pax5) were identified. FOXO3, a transcriptional activator, is the potential marker of host resistance in Salmonella infection. NF-κB1 is an inducible transcription factor which lays the foundation for studying gene network of the innate immune response of Salmonella infection in chicken. Pax5 is essential for differentiation of pre-B cells into mature B cell. The real time PCR analysis showed that in response to Salmonella Typhimurium infection a remarkable increase of NF-κB1 (P˂0.01), FOXO3 (P˂0.01) gene expression in liver and Pax5 (P˂0.01) gene expression in spleen of Kashmir favorella was observed. The protein-protein interaction (PPI) and protein-TF interaction network by STRINGDB analysis suggests that FOXO3 is a hub gene in the network and is closely related to Salmonella infection along with NF-κB1. All the three differentially expressed genes (NF-κB1, FOXO3 and PaX5) showed their influence on 12 interacting proteins and 16 TFs, where cyclic adenosine monophosphate Response Element Binding protein (CREBBP), erythroblast transformation-specific (ETSI), Tumour-protein 53(TP53I), IKKBK, lymphoid enhancer-binding factor-1 (LEF1), and interferon regulatory factor-4 (IRF4) play role in immune responses. This study shall pave the way for newer strategies for treatment and prevention of Salmonella infection and may help in increasing the innate disease resistance.
Collapse
Affiliation(s)
- Syed Mudasir Ahmad
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India.
| | - Sahar Saleem Bhat
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Shaista Shafi
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | - Mashooq Ahmad Dar
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Afnan Saleem
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Zulfqarul Haq
- Division of Livestock Production and Management, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Nida Farooq
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Junaid Nazir
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Basharat Bhat
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India.
| |
Collapse
|
6
|
Di Zazzo E, Rienzo M, Casamassimi A, De Rosa C, Medici N, Gazzerro P, Bifulco M, Abbondanza C. Exploring the putative role of PRDM1 and PRDM2 transcripts as mediators of T lymphocyte activation. J Transl Med 2023; 21:217. [PMID: 36964555 PMCID: PMC10039509 DOI: 10.1186/s12967-023-04066-x] [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: 02/15/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND T cell activation and programming from their naïve/resting state, characterized by widespread modifications in chromatin accessibility triggering extensive changes in transcriptional programs, is orchestrated by several cytokines and transcription regulators. PRDM1 and PRDM2 encode for proteins with PR/SET and zinc finger domains that control several biological processes, including cell differentiation, through epigenetic regulation of gene expression. Different transcripts leading to main protein isoforms with (PR +) or without (PR-) the PR/SET domain have been described. Although many studies have established the critical PRDM1 role in hematopoietic cell differentiation, maintenance and/or function, the single transcript contribution has not been investigated before. Otherwise, very few evidence is currently available on PRDM2. Here, we aimed to analyze the role of PRDM1 and PRDM2 different transcripts as mediators of T lymphocyte activation. METHODS We analyzed the transcription signature of the main variants from PRDM1 (BLIMP1a and BLIMP1b) and PRDM2 (RIZ1 and RIZ2) genes, in human T lymphocytes and Jurkat cells overexpressing PRDM2 cDNAs following activation through different signals. RESULTS T lymphocyte activation induced an early increase of RIZ2 and RIZ1 followed by BLIMP1b increase and finally by BLIMP1a increase. The "first" and the "second" signals shifted the balance towards the PR- forms for both genes. Interestingly, the PI3K signaling pathway modulated the RIZ1/RIZ2 ratio in favor of RIZ1 while the balance versus RIZ2 was promoted by MAPK pathway. Cytokines mediating different Jak/Stat signaling pathways (third signal) early modulated the expression of PRDM1 and PRDM2 and the relationship of their different transcripts confirming the early increase of the PR- transcripts. Different responses of T cell subpopulations were also observed. Jurkat cells showed that the acute transient RIZ2 increase promoted the balancing of PRDM1 forms towards BLIMP1b. The stable forced expression of RIZ1 or RIZ2 induced a significant variation in the expression of key transcription factors involved in T lymphocyte differentiation. The BLIMP1a/b balance shifted in favor of BLIMP1a in RIZ1-overexpressing cells and of BLIMP1b in RIZ2-overexpressing cells. CONCLUSIONS This study provides the first characterization of PRDM2 in T-lymphocyte activation/differentiation and novel insights on PRDM1 and PRDM2 transcription regulation during initial activation phases.
Collapse
Affiliation(s)
- Erika Di Zazzo
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, 86100, Campobasso, Italy
| | - Monica Rienzo
- Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", 81100, Caserta, Italy
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Caterina De Rosa
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Nicola Medici
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084, Salerno, Fisciano (SA), Italy
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131, Naples, Italy
| | - Ciro Abbondanza
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.
| |
Collapse
|
7
|
Zhao Y, Song J, Dong W, Liu X, Yang C, Wang D, Xue Y, Ruan X, Liu L, Wang P, Zhang M, Liu Y. The MBNL1/circNTRK2/PAX5 pathway regulates aerobic glycolysis in glioblastoma cells by encoding a novel protein NTRK2-243aa. Cell Death Dis 2022; 13:767. [PMID: 36064939 PMCID: PMC9445070 DOI: 10.1038/s41419-022-05219-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 01/21/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common tumor of the human central nervous system. Aerobic glycolysis has been strongly related to tumor development and malignant behavior. In this study, we found that MBNL1, circNTRK2, and NTRK2-243aa were markedly downregulated and inhibited glycolysis in GBM, whereas PAX5 was upregulated and promoted glycolysis. Functionally, MBNL1 promoted the expression of circNTRK2 by binding to NTRK2 pre-mRNA, as validated using RNA pull-down and nascent RNA immunoprecipitation assays. Mass spectrometry, western blotting, and immunofluorescence staining methods were used to detect the expression of NTRK2-243aa. NTRK2-243aa-encoded by circNTRK2-phosphorylated PAX5 at Y102, leading to the attenuation of the half-life of PAX5, as validated by in vitro kinase and MG132 rescue assays. Besides, PAX5 transcriptionally facilitated the expression of PKM2 and HK2 by binding to their promoter regions, as verified by luciferase reporter and chromatin immunoprecipitation assays. Finally, overexpression of MBNL1 and circNTRK2 combined with PAX5 knockdown effectively inhibited the formation of GBM xenograft tumors and significantly prolonged the survival of orthotopic nude mice. We have delineated that the MBNL1/circNTRK2/PAX5 pathway plays a crucial role in regulating GBM glycolysis and could provide potential targets and alternative strategies for the treatment of GBM.
Collapse
Affiliation(s)
- Yubo Zhao
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Jian Song
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Weiwei Dong
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Xiaobai Liu
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Chunqing Yang
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Di Wang
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| | - Yixue Xue
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Xuelei Ruan
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Libo Liu
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Ping Wang
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Mengyang Zhang
- grid.412449.e0000 0000 9678 1884Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122 China
| | - Yunhui Liu
- grid.412467.20000 0004 1806 3501Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004 China ,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004 China ,Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004 China
| |
Collapse
|
8
|
The Pleiotropy of PAX5 Gene Products and Function. Int J Mol Sci 2022; 23:ijms231710095. [PMID: 36077495 PMCID: PMC9456430 DOI: 10.3390/ijms231710095] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
PAX5, a member of the Paired Box (PAX) transcription factor family, is an essential factor for B-lineage identity during lymphoid differentiation. Mechanistically, PAX5 controls gene expression profiles, which are pivotal to cellular processes such as viability, proliferation, and differentiation. Given its crucial function in B-cell development, PAX5 aberrant expression also correlates with hallmark cancer processes leading to hematological and other types of cancer lesions. Despite the well-established association of PAX5 in the development, maintenance, and progression of cancer disease, the use of PAX5 as a cancer biomarker or therapeutic target has yet to be implemented. This may be partly due to the assortment of PAX5 expressed products, which layers the complexity of their function and role in various regulatory networks and biological processes. In this review, we provide an overview of the reported data describing PAX5 products, their regulation, and function in cellular processes, cellular biology, and neoplasm.
Collapse
|
9
|
DeLuca JM, Murphy MK, Wang X, Wilson TJ. FCRL1 Regulates B Cell Receptor-Induced ERK Activation through GRB2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2688-2698. [PMID: 34697226 PMCID: PMC8629370 DOI: 10.4049/jimmunol.2100218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Regulation of BCR signaling has important consequences for generating effective Ab responses to pathogens and preventing production of autoreactive B cells during development. Currently defined functions of Fc receptor-like (FCRL) 1 include positive regulation of BCR-induced calcium flux, proliferation, and Ab production; however, the mechanistic basis of FCRL1 signaling and its contributions to B cell development remain undefined. Molecular characterization of FCRL1 signaling shows phosphotyrosine-dependent associations with GRB2, GRAP, SHIP-1, and SOS1, all of which can profoundly influence MAPK signaling. In contrast with previous characterizations of FCRL1 as a strictly activating receptor, we discover a role for FCRL1 in suppressing ERK activation under homeostatic and BCR-stimulated conditions in a GRB2-dependent manner. Our analysis of B cells in Fcrl1 -/- mice shows that ERK suppression by FCRL1 is associated with a restriction in the number of cells surviving splenic maturation in vivo. The capacity of FCRL1 to modulate ERK activation presents a potential for FCRL1 to be a regulator of peripheral B cell tolerance, homeostasis, and activation.
Collapse
Affiliation(s)
- Jenna M DeLuca
- Department of Microbiology, Miami University, Oxford, OH
| | | | - Xin Wang
- Department of Microbiology, Miami University, Oxford, OH
| | | |
Collapse
|
10
|
Verstegen NJM, Ubels V, Westerhoff HV, van Ham SM, Barberis M. System-Level Scenarios for the Elucidation of T Cell-Mediated Germinal Center B Cell Differentiation. Front Immunol 2021; 12:734282. [PMID: 34616402 PMCID: PMC8488341 DOI: 10.3389/fimmu.2021.734282] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Germinal center (GC) reactions are vital to the correct functioning of the adaptive immune system, through formation of high affinity, class switched antibodies. GCs are transient anatomical structures in secondary lymphoid organs where specific B cells, after recognition of antigen and with T cell help, undergo class switching. Subsequently, B cells cycle between zones of proliferation and somatic hypermutation and zones where renewed antigen acquisition and T cell help allows for selection of high affinity B cells (affinity maturation). Eventually GC B cells first differentiate into long-lived memory B cells (MBC) and finally into plasma cells (PC) that partially migrate to the bone marrow to encapsulate into long-lived survival niches. The regulation of GC reactions is a highly dynamically coordinated process that occurs between various cells and molecules that change in their signals. Here, we present a system-level perspective of T cell-mediated GC B cell differentiation, presenting and discussing the experimental and computational efforts on the regulation of the GCs. We aim to integrate Systems Biology with B cell biology, to advance elucidation of the regulation of high-affinity, class switched antibody formation, thus to shed light on the delicate functioning of the adaptive immune system. Specifically, we: i) review experimental findings of internal and external factors driving various GC dynamics, such as GC initiation, maturation and GCBC fate determination; ii) draw comparisons between experimental observations and mathematical modeling investigations; and iii) discuss and reflect on current strategies of modeling efforts, to elucidate B cell behavior during the GC tract. Finally, perspectives are specifically given on to the areas where a Systems Biology approach may be useful to predict novel GCBC-T cell interaction dynamics.
Collapse
Affiliation(s)
- Niels J M Verstegen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Victor Ubels
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford, United Kingdom
| | - Hans V Westerhoff
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands.,Department of Molecular Cell Physiology, VU University Amsterdam, Amsterdam, Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands.,Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
11
|
Szodoray P, Andersen TK, Heinzelbecker J, Imbery JF, Huszthy PC, Stanford SM, Bogen B, Landsverk OB, Bottini N, Tveita A, Munthe LA, Nakken B. Integration of T helper and BCR signals governs enhanced plasma cell differentiation of memory B cells by regulation of CD45 phosphatase activity. Cell Rep 2021; 36:109525. [PMID: 34380042 PMCID: PMC8435664 DOI: 10.1016/j.celrep.2021.109525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 06/11/2021] [Accepted: 07/22/2021] [Indexed: 12/29/2022] Open
Abstract
Humoral immunity relies on the efficient differentiation of memory B cells (MBCs) into antibody-secreting cells (ASCs). T helper (Th) signals upregulate B cell receptor (BCR) signaling by potentiating Src family kinases through increasing CD45 phosphatase activity (CD45 PA). In this study, we show that high CD45 PA in MBCs enhances BCR signaling and is essential for their effective ASC differentiation. Mechanistically, Th signals upregulate CD45 PA through intensifying the surface binding of a CD45 ligand, Galectin-1. CD45 PA works as a sensor of T cell help and defines high-affinity germinal center (GC) plasma cell (PC) precursors characterized by IRF4 expression in vivo. Increasing T cell help in vitro results in an incremental CD45 PA increase and enhances ASC differentiation by facilitating effective induction of the transcription factors IRF4 and BLIMP1. This study connects Th signals with BCR signaling through Galectin-1-dependent regulation of CD45 PA and provides a mechanism for efficient ASC differentiation of MBCs.
Collapse
Affiliation(s)
- Peter Szodoray
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Tor Kristian Andersen
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for Influenza Vaccine Research, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Julia Heinzelbecker
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - John F Imbery
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Peter C Huszthy
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway
| | - Stephanie M Stanford
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, San Diego, 9500 Gilman Drive MC #0656, La Jolla, CA 92093, USA
| | - Bjarne Bogen
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for Influenza Vaccine Research, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ole B Landsverk
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway
| | - Nunzio Bottini
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, San Diego, 9500 Gilman Drive MC #0656, La Jolla, CA 92093, USA
| | - Anders Tveita
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Britt Nakken
- Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Center for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
EBF1 drives hallmark B cell gene expression by enabling the interaction of PAX5 with the MLL H3K4 methyltransferase complex. Sci Rep 2021; 11:1537. [PMID: 33452395 PMCID: PMC7810865 DOI: 10.1038/s41598-021-81000-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/28/2020] [Indexed: 12/15/2022] Open
Abstract
PAX5 and EBF1 work synergistically to regulate genes that are involved in B lymphocyte differentiation. We used the KIS-1 diffuse large B cell lymphoma cell line, which is reported to have elevated levels of PAX5 expression, to investigate the mechanism of EBF1- and PAX5-regulated gene expression. We demonstrate the lack of expression of hallmark B cell genes, including CD19, CD79b, and EBF1, in the KIS-1 cell line. Upon restoration of EBF1 expression we observed activation of CD19, CD79b and other genes with critical roles in B cell differentiation. Mass spectrometry analyses of proteins co-immunoprecipitated with PAX5 in KIS-1 identified components of the MLL H3K4 methylation complex, which drives histone modifications associated with transcription activation. Immunoblotting showed a stronger association of this complex with PAX5 in the presence of EBF1. Silencing of KMT2A, the catalytic component of MLL, repressed the ability of exogenous EBF1 to activate transcription of both CD19 and CD79b in KIS-1 cells. We also find association of PAX5 with the MLL complex and decreased CD19 expression following silencing of KMT2A in other human B cell lines. These data support an important role for the MLL complex in PAX5-mediated transcription regulation.
Collapse
|
13
|
Davidzohn N, Biram A, Stoler-Barak L, Grenov A, Dassa B, Shulman Z. Syk degradation restrains plasma cell formation and promotes zonal transitions in germinal centers. J Exp Med 2020; 217:133542. [PMID: 31873727 PMCID: PMC7062533 DOI: 10.1084/jem.20191043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/08/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
In germinal centers, B cells interact with antigen in the light zone and clonally expand in the dark zone. Davidzohn et al. show that BCR-induced Syk degradation in the light zone attenuates signal transduction, impedes plasma cell formation, and promotes B cell transition to the dark zone. Germinal centers (GCs) are sites at which B cells proliferate and mutate their antibody-encoding genes in the dark zone (DZ), followed by affinity-based selection in the light zone (LZ). B cell antigen receptor (BCR) signals induce Syk activation followed by rapid phosphatase-mediated desensitization; however, how degradation events regulate BCR functions in GCs is unclear. Here, we found that Syk degradation restrains plasma cell (PC) formation in GCs and promotes B cell LZ to DZ transition. Using a mouse model defective in Cbl-mediated Syk degradation, we demonstrate that this machinery attenuates BCR signaling intensity by mitigating the Kras/Erk and PI3K/Foxo1 pathways, and restricting the expression of PC transcription factors in GC B cells. Inhibition of Syk degradation perturbed gene expression, specifically in the LZ, and enhanced the generation of PCs without affecting B cell proliferation. These findings reveal how long-lasting attenuation of signal transduction by degradation events regulates cell fate within specialized microanatomical sites.
Collapse
Affiliation(s)
- Natalia Davidzohn
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liat Stoler-Barak
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amalie Grenov
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Bareket Dassa
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
14
|
Suzuki JI, Miki S, Ushijima M, Kodera Y. Regulation of immune response by S-1-propenylcysteine through autophagy-mediated protein degradation. Exp Ther Med 2019; 19:1570-1573. [PMID: 32010341 PMCID: PMC6966193 DOI: 10.3892/etm.2019.8392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a key event in cellular recycling processes due to its involvement in the intracellular degradation of proteins. It has been demonstrated that S−1-propenylcysteine (S1PC), a characteristic sulfur compound in aged garlic extract, induces the activation of autophagy. S1PC degrades the adaptor protein myeloid differentiation response protein 88 (MyD88) of downstream of Toll-like receptor (TLR) by activating autophagy in vitro and in vivo. The degradation of MyD88 inhibits the TLR signaling pathway, including the phosphorylation of interleukin 1 receptor associated kinase 4 (IRAK4) and nuclear factor (NF)-κB p65 in vitro, and eventually leads to the inhibition of interleukin (IL)-6 production in vitro and C-C motif chemokine ligand 2 (Ccl2) mRNA expression in vivo. S1PC also increases the level of intestinal immunoglobulin A (IgA) and the number of IgA-producing cells in Peyer's patches in vivo. In addition, S1PC triggers the mRNA expression of X-box binding protein 1 (Xbp1), an inducer of IgA-producing cell differentiation via the phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and the degradation of paired box protein 5 (Pax5), a suppressor of Xbp1 mRNA expression. The present review summarizes the mechanisms through which the activation of autophagy by S1PC modulates the immune response.
Collapse
Affiliation(s)
- Jun-Ichiro Suzuki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Satomi Miki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Mitsuyasu Ushijima
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Yukihiro Kodera
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| |
Collapse
|
15
|
Interleukin 21 Receptor/Ligand Interaction Is Linked to Disease Progression in Pancreatic Cancer. Cells 2019; 8:cells8091104. [PMID: 31540511 PMCID: PMC6770770 DOI: 10.3390/cells8091104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) displays a marked fibro-inflammatory microenvironment in which infiltrated immune cells fail to eliminate the tumor cells and often—rather paradoxically—promote tumor progression. Of special interest are tumor-promoting T cells that assume a Th17-like phenotype because their presence in PDAC tissue is associated with a poor prognosis. In that context, the role of IL-21, a major cytokine released by Th17-like cells, was assessed. In all tissue samples (n = 264) IL-21+ immune cells were detected by immunohistochemistry and high density of those cells was associated with poor prognosis. In the majority of patients (221/264), tumor cells expressed the receptor for IL-21 (IL-21R) and also a downstream target of IL-21, Blimp-1 (199/264). Blimp-1 expression closely correlated with IL-21R expression and multivariate analysis revealed that expression of both IL-21R and Blimp-1 was associated with shorter survival time of the patients. In vitro data using pancreatic tumor cells lines provided a possible explanation: IL-21 activated ERK and STAT3 pathways and upregulated Blimp-1. Moreover, IL-21 increased invasion of tumor cell lines in a Blimp-1-dependent manner. As an in vivo correlate, an avian xenograft model was used. Here again Blimp-1 expression was significantly upregulated in IL-21 stimulated tumor cells. In summary, our data showed an association of IL-21+ immune cell infiltration and IL-21 receptor expression in PDAC with poor survival, most likely due to an IL-21-mediated promotion of tumor cell invasion and enhanced colony formation, supporting the notion of the tumor-promoting abilities of the tumor microenvironment.
Collapse
|
16
|
Yamamoto H, Hayakawa F, Yasuda T, Odaira K, Minamikawa Y, Tange N, Hirano D, Kojima Y, Morishita T, Tsuzuki S, Naoe T, Kiyoi H. ZNF384-fusion proteins have high affinity for the transcriptional coactivator EP300 and aberrant transcriptional activities. FEBS Lett 2019; 593:2151-2161. [PMID: 31234226 DOI: 10.1002/1873-3468.13506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 11/07/2022]
Abstract
Zinc-finger protein 384 (ZNF384) fusion (Z-fusion) genes have recently been identified as recurrent fusion genes in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) and have been detected in 7-17% of Philadelphia chromosome-negative BCP-ALL cases. We selected SALL4 and ID2 as potential Z-fusion-specific transcriptional targets that might lead to the differentiation disorder of Z-fusion-positive ALL. The introduction of EP300-ZNF384 and SYNRG-ZNF384 induced the expression of these genes. Z-fusion proteins exhibited stronger transcriptional activities on the promoter or enhancer region of these genes than Wild-Z. Furthermore, GST pull-down assay revealed that Z-fusion proteins associated more strongly with EP300 than Wild-Z. Coexpression of EP300 specifically enhanced the transcriptional activities of Z-fusion proteins. We propose the increased EP300 binding of Z-fusion proteins as a mechanism for their increased transcriptional activities.
Collapse
Affiliation(s)
- Hideyuki Yamamoto
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Fumihiko Hayakawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan.,Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Takahiko Yasuda
- Clinical Research Center, Nagoya Medical Center, National Hospital Organization, Nagoya, Japan
| | - Koya Odaira
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Yuka Minamikawa
- Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Naoyuki Tange
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Daiki Hirano
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Yuki Kojima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Takanobu Morishita
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, School of Medicine, Aichi Medical University, Japan
| | - Tomoki Naoe
- Nagoya Medical Center, National Hospital Organization, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
17
|
Chromosomal translocation-mediated evasion from miRNA induces strong MEF2D fusion protein expression, causing inhibition of PAX5 transcriptional activity. Oncogene 2018; 38:2263-2274. [PMID: 30478446 DOI: 10.1038/s41388-018-0573-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 08/29/2018] [Accepted: 10/11/2018] [Indexed: 12/22/2022]
Abstract
MEF2D fusion genes are newly discovered recurrent gene abnormalities that are detected in approximately 5% of acute lymphoblastic leukemia cases. We previously demonstrated that the vector-driven expression of MEF2D fusion proteins was markedly stronger than that of wild-type MEF2D; however, the underlying mechanisms and significance of this expression have yet to be clarified. We herein showed that the strong expression of MEF2D fusion proteins was caused by the loss of the target site of miRNA due to gene translocation. We identified the target region of miRNA located in the coding region and selected miR-122 as a candidate of the responsible miRNA. Mutations at a putative binding site of miR-122 increased MEF2D expression, while the transfection of its miRNA mimic reduced the expression of wild-type MEF2D, but not MEF2D fusion proteins. We also found that MEF2D fusion proteins inhibited the transcriptional activity of PAX5, a B-cell differentiation regulator in a manner that depended on fusion-specific strong expression and an association with histone deacetylase 4, which may lead to the differentiation disorders of B cells. Our results provide novel insights into the mechanisms underlying leukemia development by MEF2D fusion genes and the involvement of the deregulation of miRNA-mediated repression in cancer development.
Collapse
|
18
|
Sud A, Thomsen H, Orlando G, Försti A, Law PJ, Broderick P, Cooke R, Hariri F, Pastinen T, Easton DF, Pharoah PDP, Dunning AM, Peto J, Canzian F, Eeles R, Kote-Jarai ZS, Muir K, Pashayan N, Campa D, Hoffmann P, Nöthen MM, Jöckel KH, von Strandmann EP, Swerdlow AJ, Engert A, Orr N, Hemminki K, Houlston RS. Genome-wide association study implicates immune dysfunction in the development of Hodgkin lymphoma. Blood 2018; 132:2040-2052. [PMID: 30194254 PMCID: PMC6236462 DOI: 10.1182/blood-2018-06-855296] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023] Open
Abstract
To further our understanding of inherited susceptibility to Hodgkin lymphoma (HL), we performed a meta-analysis of 7 genome-wide association studies totaling 5325 HL cases and 22 423 control patients. We identify 5 new HL risk loci at 6p21.31 (rs649775; P = 2.11 × 10-10), 6q23.3 (rs1002658; P = 2.97 × 10-8), 11q23.1 (rs7111520; P = 1.44 × 10-11), 16p11.2 (rs6565176; P = 4.00 × 10-8), and 20q13.12 (rs2425752; P = 2.01 × 10-8). Integration of gene expression, histone modification, and in situ promoter capture Hi-C data at the 5 new and 13 known risk loci implicates dysfunction of the germinal center reaction, disrupted T-cell differentiation and function, and constitutive NF-κB activation as mechanisms of predisposition. These data provide further insights into the genetic susceptibility and biology of HL.
Collapse
Affiliation(s)
- Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Hauke Thomsen
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Giulia Orlando
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Peter Broderick
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Rosie Cooke
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Fadi Hariri
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, McGill University, Montreal, QC, Canada
| | - Tomi Pastinen
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, McGill University, Montreal, QC, Canada
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, and
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, and
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, and
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center, Heidelberg, Germany
| | - Rosalind Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - ZSofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Kenneth Muir
- Institute of Population Health, University of Manchester, Manchester, United Kingdom
- Division of Health Sciences, Warwick Medical School, Warwick University, Coventry, United Kingdom
| | - Nora Pashayan
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- Department of Applied Health Research, University College London, London, United Kingdom
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Per Hoffmann
- Human Genomic Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Human Genetics and
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics and
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | | | - Elke Pogge von Strandmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Division of Breast Cancer Research, The Institute of Cancer Research, London, United Kingdom; and
| | - Andreas Engert
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Nick Orr
- Division of Breast Cancer Research, The Institute of Cancer Research, London, United Kingdom; and
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
19
|
Effect of glutamine on lymphoplasmacytic lymphoma, especially on the viewpoint of the differentiation into vulnerable subpopulation. Pathol Res Pract 2018; 214:1667-1674. [PMID: 30173943 DOI: 10.1016/j.prp.2018.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/07/2018] [Accepted: 08/18/2018] [Indexed: 11/24/2022]
Abstract
Glutamine (Gln) is important not only for cell proliferation but also for differentiation. Although Gln is essential for plasmacytic differentiation of lymphocytes, no study has been done on the effect of Gln on differentiation of tumor cells, such as lymphoma. Here we examined the effect of Gln on plasmacytic differentiation of lymphoplasmacytic lymphoma (LPL) with its cell lines, MWCL-1 and RPCI-WM1. Gln promoted plasmacytic differentiation of LPL, and p38 MAPK signaling pathway mediated such differentiation. We previously reported that the subpopulation with plasmacytic differentiation was vulnerable to apoptosis in LPL. Although it is difficult to lead these findings to the radical therapy, they might help the treatment of LPL, in which stimulation of p38 MAPK by Gln induced differentiation of LPL into vulnerable subpopulation.
Collapse
|
20
|
Ikuse T, Ohtsuka Y, Obayashi N, Jimbo K, Aoyagi Y, Kudo T, Asaoka D, Hojo M, Nagahara A, Watanabe S, Blanchard TG, Czinn SJ, Shimizu T. Host response genes associated with nodular gastritis in Helicobacter pylori infection. Pediatr Int 2018; 60:446-454. [PMID: 29415337 DOI: 10.1111/ped.13527] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/19/2017] [Accepted: 02/01/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Chronic Helicobacter pylori infection in children induces lymphoid hyperplasia called nodular gastritis (NG) at the antral gastric mucosa. The aim of this study was to evaluate genes in gastric biopsy on microarray analysis, to identify molecules associated with NG on comparison with NG-negative pediatric corpus tissue and with H. pylori-infected adult tissue with atrophic gastritis (AG). METHODS Eight pediatric and six adult H. pylori-infected patients, as well as six pediatric and six adult uninfected patients were evaluated. All infected adults had AG. NG was observed in the antrum of all eight pediatric patients and in the corpus of three patients. Adult and uninfected patients were free of NG; that is, only pediatric H. pylori-infected patients had NG. Total RNA was purified from gastric biopsy, and microarray analysis was performed to compare gene expression between groups. The three infected children with NG in both the antrum and corpus were excluded from analysis of corpus samples. RESULTS The number of genes significantly up- or downregulated (fold change >3, P < 0.01) compared with uninfected controls varied widely: 72 in pediatric antrum, 45 in pediatric corpus, 103 in adult antrum and 71 in adult corpus. Nineteen genes had significantly altered expression in the antrum of NG tissue compared with NG-negative pediatric corpus tissue and adult AG tissue. The CD20 B-cell specific differentiation antigen had the most pronounced increase. Previously described regulators of NG development were not predominantly upregulated in the NG mucosa. CONCLUSIONS CD20 overexpression may play an important role in lymphoid follicle enlargement and NG.
Collapse
Affiliation(s)
- Tamaki Ikuse
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yoshikazu Ohtsuka
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naho Obayashi
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keisuke Jimbo
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yo Aoyagi
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takahiro Kudo
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Daisuke Asaoka
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mariko Hojo
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihito Nagahara
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sumio Watanabe
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Thomas G Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Steven J Czinn
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Toshiaki Shimizu
- Department of Pediatric and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Affiliation(s)
- Niklas Engels
- Institute of Cellular & Molecular Immunology; University Medical Center Göttingen; Göttingen Germany
| | - Jürgen Wienands
- Institute of Cellular & Molecular Immunology; University Medical Center Göttingen; Göttingen Germany
| |
Collapse
|
22
|
Dysregulation of Blimp1 transcriptional repressor unleashes p130Cas/ErbB2 breast cancer invasion. Sci Rep 2017; 7:1145. [PMID: 28442738 PMCID: PMC5430666 DOI: 10.1038/s41598-017-01332-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/28/2017] [Indexed: 12/29/2022] Open
Abstract
ErbB2 overexpression is detected in approximately 20% of breast cancers and is correlated with poor survival. It was previously shown that the adaptor protein p130Cas/BCAR1 is a crucial mediator of ErbB2 transformation and that its overexpression confers invasive properties to ErbB2-positive human mammary epithelial cells. We herein prove, for the first time, that the transcriptional repressor Blimp1 is a novel mediator of p130Cas/ErbB2-mediated invasiveness. Indeed, high Blimp1 expression levels are detected in invasive p130Cas/ErbB2 cells and correlate with metastatic status in human breast cancer patients. The present study, by using 2D and 3D breast cancer models, shows that the increased Blimp1 expression depends on both MAPK activation and miR-23b downmodulation. Moreover, we demonstrate that Blimp1 triggers cell invasion and metastasis formation via its effects on focal adhesion and survival signaling. These findings unravel the previously unidentified role that transcriptional repressor Blimp1 plays in the control of breast cancer invasiveness.
Collapse
|
23
|
Wada N, Ikeda JI, Tanaka H, Sakakita H, Hori M, Ikehara Y, Morii E. Effect of plasma-activated medium on the decrease of tumorigenic population in lymphoma. Pathol Res Pract 2017; 213:773-777. [PMID: 28554762 DOI: 10.1016/j.prp.2017.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/08/2017] [Accepted: 04/09/2017] [Indexed: 10/19/2022]
Abstract
Nonequilibrium atmospheric pressure plasma (NEAPP) is a novel approach for blood coagulation, wound healing, and tumor elimination. NEAPP not only directly but also indirectly affects living cells via the medium exposed to NEAPP-yielding devises, called plasma-activated medium (PAM). The conservable and portable PAM serves as an alternative and advantageous approach over direct NEAPP. Here we examined the effect of PAM on lymphoplasmacytic lymphoma (LPL) cell lines. We found that PAM induced plasma cell differentiation and reduced tumorigenic population. PAM increased the expression level of PRDM1α, which is a transcription factor promoting plasma cell differentiation, suggesting that plasma cell differentiation of LPL might be mediated by PRDM1α. We previously reported that plasma cell component of LPL is vulnerable to apoptosis and less tumorigenic. These findings suggested that PAM treatment might become a novel therapy against LPL by inducing the transition from tumorigenic to non-tumorigenic population.
Collapse
Affiliation(s)
- Naoki Wada
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Jun-Ichiro Ikeda
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Hiromasa Tanaka
- Institute of Innovation for Future Society, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Hajime Sakakita
- Electronics and Photonics Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, 305-8568, Japan
| | - Masaru Hori
- Institute of Innovation for Future Society, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Yuzuru Ikehara
- Biotechnology Research Institute for Drug Discovery, AIST, Tsukuba, 305-8568, Japan; Departement of Pathology, Graduate School of Medicine, Chiba University, Chiba, 107-0052, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan.
| |
Collapse
|
24
|
Inagaki Y, Hayakawa F, Hirano D, Kojima Y, Morishita T, Yasuda T, Naoe T, Kiyoi H. PAX5 tyrosine phosphorylation by SYK co-operatively functions with its serine phosphorylation to cancel the PAX5-dependent repression of BLIMP1: A mechanism for antigen-triggered plasma cell differentiation. Biochem Biophys Res Commun 2016; 475:176-81. [DOI: 10.1016/j.bbrc.2016.05.067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
|
25
|
Wang JZ, Zhang YH, Guo XH, Zhang HY, Zhang Y. The double-edge role of B cells in mediating antitumor T-cell immunity: Pharmacological strategies for cancer immunotherapy. Int Immunopharmacol 2016; 36:73-85. [PMID: 27111515 DOI: 10.1016/j.intimp.2016.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022]
Abstract
Emerging evidence reveals the controversial role of B cells in antitumor immunity, but the underlying mechanisms have to be explored. Three latest articles published in the issue 521 of Nature in 2015 reconfirmed the puzzling topic and put forward some explanations of how B cells regulate antitumor T-cell responses both positively and negatively. This paper attempts to demonstrate that different B-cell subpopulations have distinct immunological properties and that they are involved in either antitumor responses or immunosuppression. Recent studies supporting the positive and negative roles of B cells in tumor development were summarized comprehensively. Several specific B-cell subpopulations, such as IgG(+), IgA(+), IL-10(+), and regulatory B cells, were described in detail. The mechanisms underlying the controversial B-cell effects were mainly attributed to different B-cell subpopulations, different B-cell-derived cytokines, direct B cell-T cell interaction, different cancer categories, and different malignant stages, and the immunological interaction between B cells and T cells is mediated by dendritic cells. Promising B-cell-based antitumor strategies were proposed and novel B-cell regulators were summarized to present interesting therapeutic targets. Future investigations are needed to make sure that B-cell-based pharmacological strategies benefit cancer immunotherapy substantially.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China.
| | - Yu-Hua Zhang
- Department of Library, Hebei University of Engineering, Handan 056038, PR China
| | - Xin-Hua Guo
- Department of Medicine, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| | - Hong-Yan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| | - Yuan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| |
Collapse
|
26
|
Abstract
The generation of antigen-specific neutralizing antibodies and memory B cells is one of the most important immune protections of the host and is the basis for successful vaccination strategies. The protective antibodies, secreted by preexisting long-lived plasma cells and reactivated antigen-experienced memory B cells, constitute the main humoral immune defense. Distinct from the primary antibody response, the humoral memory response is generated much faster and with greater magnitude, and it produces antibodies with higher affinity and variable isotypes. Humoral immunity is critically dependent on the germinal center where high-affinity memory B cells and plasma cells are generated. In this chapter, we focus on recent advances in our understanding of the molecular mechanisms that govern fate decision for memory B cells and plasma cells and the mechanisms that maintain the long-lived plasma-cell pool, with emphasis on how the transcription factor Blimp-1 (B lymphocyte-induced maturation protein-1) helps regulate the above-mentioned immunoregulatory steps to ensure the production and maintenance of antibody-secreting plasma cells as well as how it directs memory cell vs plasma-cell fate. We also discuss the molecular basis of Blimp-1 action and how its expression is regulated.
Collapse
|
27
|
Suzuki JI, Yamaguchi T, Matsutomo T, Amano H, Morihara N, Kodera Y. S-1-Propenylcysteine promotes the differentiation of B cells into IgA-producing cells by the induction of Erk1/2-dependent Xbp1 expression in Peyer's patches. Nutrition 2016; 32:884-9. [PMID: 27134204 DOI: 10.1016/j.nut.2016.01.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/19/2016] [Accepted: 01/30/2016] [Indexed: 01/28/2023]
Abstract
OBJECTIVES S-Allylcysteine (SAC) and S-1-propenylcysteine (S1PC) are the characteristic sulfur-containing amino acids in aged garlic extract. In this study, we investigated the effect of SAC and S1PC on intestinal immunoglobulin (Ig)A production to gain insight into the immunomodulatory effect of aged garlic extract. METHODS In vitro study: Mouse splenic lymphocytes were treated with S1PC (0.1 and 0.3 mM) or SAC (0.1 and 0.3 mM) for 3 d, and IgA concentration in the culture medium was examined. In vivo study: Mice were orally administrated S1PC (7.5, 15, and 30 mg/kg) for 5 d and the IgA level in the intestinal lavage fluids as well as the population of IgA-producing cells in Peyer's patches were measured using mouse IgA enzyme-linked immunosorbent assay quantification set and flow cytometer, respectively. RESULTS S1PC enhanced IgA production in mouse splenic lymphocytes in culture. However, SAC was ineffective. In addition, oral administration of S1PC to mice increased the IgA level and number of IgA-producing cells in Peyer's Patches. Furthermore, S1PC induced the expression of X-box binding protein 1 (Xbp1) mRNA, an inducer of plasma cell differentiation, in Peyer's patches. This induction was accompanied by the degradation of paired box protein 5 and the activation of mitogen activated protein/extracellular signal-regulated kinase signaling pathway. CONCLUSION These results suggest that S1PC increases IgA-producing cells via the enhancement of Erk1/2-mediated Xbp1 expression in the intestine.
Collapse
Affiliation(s)
- Jun-Ichiro Suzuki
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan.
| | - Takako Yamaguchi
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| | - Toshiaki Matsutomo
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| | - Hirotaka Amano
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| | - Naoaki Morihara
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| | - Yukihiro Kodera
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Hiroshima, Japan
| |
Collapse
|
28
|
Méndez A, Mendoza L. A Network Model to Describe the Terminal Differentiation of B Cells. PLoS Comput Biol 2016; 12:e1004696. [PMID: 26751566 PMCID: PMC4720151 DOI: 10.1371/journal.pcbi.1004696] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 12/07/2015] [Indexed: 01/31/2023] Open
Abstract
Terminal differentiation of B cells is an essential process for the humoral immune response in vertebrates and is achieved by the concerted action of several transcription factors in response to antigen recognition and extracellular signals provided by T-helper cells. While there is a wealth of experimental data regarding the molecular and cellular signals involved in this process, there is no general consensus regarding the structure and dynamical properties of the underlying regulatory network controlling this process. We developed a dynamical model of the regulatory network controlling terminal differentiation of B cells. The structure of the network was inferred from experimental data available in the literature, and its dynamical behavior was analyzed by modeling the network both as a discrete and a continuous dynamical systems. The steady states of these models are consistent with the patterns of activation reported for the Naive, GC, Mem, and PC cell types. Moreover, the models are able to describe the patterns of differentiation from the precursor Naive to any of the GC, Mem, or PC cell types in response to a specific set of extracellular signals. We simulated all possible single loss- and gain-of-function mutants, corroborating the importance of Pax5, Bcl6, Bach2, Irf4, and Blimp1 as key regulators of B cell differentiation process. The model is able to represent the directional nature of terminal B cell differentiation and qualitatively describes key differentiation events from a precursor cell to terminally differentiated B cells. Generation of antibody-producing cells through terminal B cell differentiation represents a good model to study the formation of multiple effector cells from a progenitor cell type. This process is controlled by the action of several molecules that maintain cell type specific programs in response to cytokines, antigen recognition and the direct contact with T helper cells, forming a complex regulatory network. While there is a large body of experimental data regarding some of the key molecules involved in this process and there have been several efforts to reconstruct the underlying regulatory network, a general consensus about the structure and dynamical behavior of this network is lacking. Moreover, it is not well understood how this network controls the establishment of specific B cell expression patterns and how it responds to specific external signals. We present a model of the regulatory network controlling terminal B cell differentiation and analyze its dynamical behavior under normal and mutant conditions. The model recovers the patterns of differentiation of B cells and describes a large set of gain- and loss-of-function mutants. This model provides an unified framework to generate qualitative descriptions to interpret the role of intra- and extracellular regulators of B cell differentiation.
Collapse
Affiliation(s)
- Akram Méndez
- Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- C3, Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad de México, México
- * E-mail:
| |
Collapse
|
29
|
Wang X, Ma K, Chen M, Ko KH, Zheng BJ, Lu L. IL-17A Promotes Pulmonary B-1a Cell Differentiation via Induction of Blimp-1 Expression during Influenza Virus Infection. PLoS Pathog 2016; 12:e1005367. [PMID: 26735852 PMCID: PMC4703366 DOI: 10.1371/journal.ppat.1005367] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/04/2015] [Indexed: 12/11/2022] Open
Abstract
B-1 cells play a critical role in early protection during influenza infections by producing natural IgM antibodies. However, the underlying mechanisms involved in regulating this process are largely unknown. Here we found that during influenza infection pleural cavity B-1a cells rapidly infiltrated lungs, where they underwent plasmacytic differentiation with enhanced IgM production. This process was promoted by IL-17A signaling via induction of Blimp-1 expression and NF-κB activation in B-1a cells. Deficiency of IL-17A led to severely impaired B-1a-derived antibody production in the respiratory tract, resulting in a deficiency in viral clearance. Transfer of B-1a-derived natural antibodies rescued Il17a-/- mice from otherwise lethal infections. Together, we identify a critical function of IL-17A in promoting the plasmacytic differentiation of B-1a cells. Our findings provide new insights into the mechanisms underlying the regulation of pulmonary B-1a cell response against influenza infection. Influenza infection is highly localized in respiratory tract where immune response is triggered to provide protection from primary infection. Although natural IgM antibodies produced by B-1a cells have long been recognized as first-line protection against influenza, it remains unclear whether B-1a cell response occurs in the lung and what molecular mechanisms regulate this process. We show that airway exposure to influenza causes migration of B-1a cells to lungs for further differentiation into plasma cells with enhanced production of protective IgM antibodies. IL-17A critically regulates this process by driving differentiation of B-1a cells to high-rate IgM producing plasma cells in situ. Thus, IL-17A is a key factor in the local inflammatory milieu that modulates early humoral immunity afforded by B-1a cells.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Pathology and Center of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Kongyang Ma
- Department of Pathology and Center of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Miao Chen
- Department of Pathology and Center of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - King-Hung Ko
- Department of Pathology and Center of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Bo-Jian Zheng
- Department of Pathology and Center of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Liwei Lu
- Department of Pathology and Center of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- * E-mail:
| |
Collapse
|
30
|
Recaldin T, Fear DJ. Transcription factors regulating B cell fate in the germinal centre. Clin Exp Immunol 2015; 183:65-75. [PMID: 26352785 DOI: 10.1111/cei.12702] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2015] [Indexed: 12/27/2022] Open
Abstract
Diversification of the antibody repertoire is essential for the normal operation of the vertebrate adaptive immune system. Following antigen encounter, B cells are activated, proliferate rapidly and undergo two diversification events; somatic hypermutation (followed by selection), which enhances the affinity of the antibody for its cognate antigen, and class-switch recombination, which alters the effector functions of the antibody to adapt the response to the challenge faced. B cells must then differentiate into antibody-secreting plasma cells or long-lived memory B cells. These activities take place in specialized immunological environments called germinal centres, usually located in the secondary lymphoid organs. To complete the germinal centre activities successfully, a B cell adopts a transcriptional programme that allows it to migrate to specific sites within the germinal centre, proliferate, modify its DNA recombination and repair pathways, alter its apoptotic potential and finally undergo terminal differentiation. To co-ordinate these processes, B cells employ a number of 'master regulator' transcription factors which mediate wholesale transcriptomic changes. These master transcription factors are mutually antagonistic and form a complex regulatory network to maintain distinct gene expression programs. Within this network, multiple points of positive and negative feedback ensure the expression of the 'master regulators', augmented by a number of 'secondary' factors that reinforce these networks and sense the progress of the immune response. In this review we will discuss the different activities B cells must undertake to mount a successful T cell-dependent immune response and describe how a regulatory network of transcription factors controls these processes.
Collapse
Affiliation(s)
- T Recaldin
- Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - D J Fear
- Division of Asthma, Allergy and Lung Biology, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| |
Collapse
|
31
|
Mendoza L, Méndez A. A dynamical model of the regulatory network controlling lymphopoiesis. Biosystems 2015; 137:26-33. [PMID: 26408858 DOI: 10.1016/j.biosystems.2015.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/22/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022]
Abstract
Due to the large number of diseases associated to a malfunction of the hematopoietic system, there is an interest in knowing the molecular mechanisms controlling the differentiation of blood cell lineages. However, the structure and dynamical properties of the underlying regulatory network controlling this process is not well understood. This manuscript presents a regulatory network of 81 nodes, representing several types of molecules that regulate each other during the process of lymphopoiesis. The regulatory interactions were inferred mostly from published experimental data. However, 15 out of 159 regulatory interactions are predictions arising from the present study. The network is modelled as a continuous dynamical system, in the form of a set of differential equations. The dynamical behaviour of the model describes the differentiation process from the common lymphocyte precursor (CLP) to several mature B and T cell types; namely, plasma cell (PC), cytotoxic T lymphocyte (CTL), T helper 1 (Th1), Th2, Th17, and T regulatory (Treg) cells. The model qualitatively recapitulates key cellular differentiation events, being able to represent the directional and branched nature of lymphopoiesis, going from a multipotent progenitor to fully differentiated cell types.
Collapse
Affiliation(s)
- Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, Mexico.
| | - Akram Méndez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, Mexico; Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, México, Mexico
| |
Collapse
|
32
|
Kometani K, Kurosaki T. Differentiation and maintenance of long-lived plasma cells. Curr Opin Immunol 2015; 33:64-9. [DOI: 10.1016/j.coi.2015.01.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/16/2015] [Accepted: 01/28/2015] [Indexed: 01/03/2023]
|
33
|
White CA, Pone EJ, Lam T, Tat C, Hayama KL, Li G, Zan H, Casali P. Histone deacetylase inhibitors upregulate B cell microRNAs that silence AID and Blimp-1 expression for epigenetic modulation of antibody and autoantibody responses. THE JOURNAL OF IMMUNOLOGY 2014; 193:5933-50. [PMID: 25392531 DOI: 10.4049/jimmunol.1401702] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Class-switch DNA recombination (CSR) and somatic hypermutation (SHM), which require activation-induced cytidine deaminase (AID), and plasma cell differentiation, which requires B lymphocyte-induced maturation protein-1 (Blimp-1), are critical for the generation of class-switched and hypermutated (mature) Ab and autoantibody responses. We show that histone deacetylase inhibitors valproic acid and butyrate dampened AICDA/Aicda (AID) and PRDM1/Prdm1 (Blimp-1) mRNAs by upregulating miR-155, miR-181b, and miR-361 to silence AICDA/Aicda, and miR-23b, miR-30a, and miR-125b to silence PRDM1/Prdm1, in human and mouse B cells. This led to downregulation of AID, Blimp-1, and X-box binding protein 1, thereby inhibiting CSR, SHM, and plasma cell differentiation without altering B cell viability or proliferation. The selectivity of histone deacetylase inhibitor-mediated silencing of AICDA/Aicda and PRDM1/Prdm1 was emphasized by unchanged expression of HoxC4 and Irf4 (important inducers/modulators of AICDA/Aicda), Rev1 and Ung (central elements for CSR/SHM), and Bcl6, Bach2, or Pax5 (repressors of PRDM1/Prdm1 expression), as well as unchanged expression of miR-19a/b, miR-20a, and miR-25, which are not known to regulate AICDA/Aicda or PRDM1/Prdm1. Through these B cell-intrinsic epigenetic mechanisms, valproic acid blunted class-switched and hypermutated T-dependent and T-independent Ab responses in C57BL/6 mice. In addition, it decreased class-switched and hypermutated autoantibodies, ameliorated disease, and extended survival in lupus MRL/Fas(lpr/lpr) mice. Our findings outline epigenetic mechanisms that modulate expression of an enzyme (AID) and transcription factors (Blimp-1 and X-box binding protein 1) that are critical to the B cell differentiation processes that underpin Ab and autoantibody responses. They also provide therapeutic proof-of-principle in autoantibody-mediated autoimmunity.
Collapse
Affiliation(s)
- Clayton A White
- Department of Microbiology and Immunology, University of Texas School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and Institute for Immunology, University of California, Irvine, CA 92697
| | - Egest J Pone
- Institute for Immunology, University of California, Irvine, CA 92697
| | - Tonika Lam
- Department of Microbiology and Immunology, University of Texas School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and Institute for Immunology, University of California, Irvine, CA 92697
| | - Connie Tat
- Department of Microbiology and Immunology, University of Texas School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and Institute for Immunology, University of California, Irvine, CA 92697
| | - Ken L Hayama
- Institute for Immunology, University of California, Irvine, CA 92697
| | - Guideng Li
- Department of Microbiology and Immunology, University of Texas School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and Institute for Immunology, University of California, Irvine, CA 92697
| | - Hong Zan
- Department of Microbiology and Immunology, University of Texas School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and Institute for Immunology, University of California, Irvine, CA 92697
| | - Paolo Casali
- Department of Microbiology and Immunology, University of Texas School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; and Institute for Immunology, University of California, Irvine, CA 92697
| |
Collapse
|
34
|
Abstract
Continued generation of new B cells within the bone marrow is required throughout life. However, in old age, B lymphopoiesis is inhibited at multiple developmental stages from hematopoietic stem cells through the late stages of new B cell generation. While changes in B cell precursor subsets, as well as alterations in the supporting bone marrow microenvironment, in old age have been known for the last 20 years, only more recently have insights into the cellular and molecular mechanisms responsible become clarified. Our recent discovery that B cells in aged mice are pro-inflammatory and can diminish B cell generation within the bone marrow suggests a potential mechanism of inappropriate "B cell feedback" which contributes to a bone marrow microenvironment unfavorable to B lymphopoiesis. We hypothesize that the consequences of a pro-inflammatory microenvironment in old age are (1) reduced B cell generation and (2) alteration in the "read-out" of the antibody repertoire. Both of these likely ensue from reduced expression of the surrogate light chain (λ5 + VpreB) and consequently reduced expression of the pre-B cell receptor (preBCR), critical to pre-B cell expansion and Vh selection. In old age, B cell development may progressively be diverted into a preBCR-compromised pathway. These abnormalities in B lymphopoiesis likely contribute to the poor humoral immunity seen in old age.
Collapse
Affiliation(s)
- Richard L Riley
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, P.O. Box 016960 (R-138), Miami, FL, 33101, USA,
| |
Collapse
|
35
|
Purssell E. Cyclooxygenase inhibitors inhibit antibody response through interference with MAPK/ERK pathways and BLIMP-1 inhibition. Med Hypotheses 2014; 83:372-7. [PMID: 25012778 DOI: 10.1016/j.mehy.2014.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 06/09/2014] [Accepted: 06/17/2014] [Indexed: 11/16/2022]
Abstract
Fever is a common symptom of illness in children, and although not harmful in itself, fever and its associated symptoms are often treated with antipyretic drugs. A number of national and other guidelines now recommend against their routine use; a conclusion that was initially supported by a study showing that the prophylactic use of paracetamol might reduce antibody response to some vaccine antigens, although data from booster vaccinations are more equivocal. Although in vivo data on the cause of this inhibition are scarce, in vitro data suggests that the cause may be due to inhibition of the mitogen activated protein kinase/extracellular regulated protein kinase pathways, and a subsequent reduction in the process of plasma cell differentiation at the beginning of the antibody response. This suggests that in high-risk patients these drugs could be avoided in the early part of an infection when plasma-cell differentiation is occurring. More data are needed to define this period; until then existing data support the recommendation against the routine use of these drugs.
Collapse
Affiliation(s)
- E Purssell
- King's College London, James Clerk Maxwell Building, 57 Waterloo Road, London SE1 8WA, United Kingdom.
| |
Collapse
|
36
|
Wheeler ML, Dong MB, Brink R, Zhong XP, DeFranco AL. Diacylglycerol kinase ζ limits B cell antigen receptor-dependent activation of ERK signaling to inhibit early antibody responses. Sci Signal 2013; 6:ra91. [PMID: 24129701 DOI: 10.1126/scisignal.2004189] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Signaling downstream of the B cell antigen receptor (BCR) is tightly regulated to enable cells to gauge the strength and duration of antigen-receptor interactions and to respond appropriately. We investigated whether metabolism of the second messenger diacylglycerol (DAG) by members of the family of DAG kinases (DGKs) played a role in modulating the magnitude of signaling by DAG downstream of the BCR. In the absence of DGKζ, the threshold for BCR signaling, measured as activation of the Ras-extracellular signal-regulated kinase (ERK) pathway, was markedly reduced in mature follicular B cells, which resulted in enhanced responses to antigen in vitro and in vivo. Inhibition of DAG signaling by DGKζ limited the number of antibody-secreting cells that were generated early in response to T cell-independent type 2 antigens, as well as to T cell-dependent antigens. Furthermore, the effect of loss of DGKζ closely resembled the effect of increasing the affinity of the BCR for antigen during the T cell-dependent antibody response. These results suggest that the magnitude of DAG signaling is important for translating the affinity of the BCR for antigen into the amount of antibody produced during the early stages of an immune response.
Collapse
Affiliation(s)
- Matthew L Wheeler
- 1Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
37
|
Pagni F, Bosisio FM, Sala E, Cattoretti G, Isimbaldi G, Coppola S, Nespoli L, Carpenedo M. The plasmablasts in Castleman disease. Am J Clin Pathol 2013; 139:555-9. [PMID: 23525622 DOI: 10.1309/ajcp5mcr0etaakzk] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|