1
|
Luu M, Krause FF, Monning H, Wempe A, Leister H, Mainieri L, Staudt S, Ziegler-Martin K, Mangold K, Kappelhoff N, Shaul YD, Göttig S, Plaza-Sirvent C, Schulte LN, Bekeredjian-Ding I, Schmitz I, Steinhoff U, Visekruna A. Dissecting the metabolic signaling pathways by which microbial molecules drive the differentiation of regulatory B cells. Mucosal Immunol 2025; 18:66-75. [PMID: 39265892 DOI: 10.1016/j.mucimm.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
The host-microbiome axis has been implicated in promoting anti-inflammatory immune responses. Yet, the underlying molecular mechanisms of commensal-mediated IL-10 production by regulatory B cells (Bregs) are not fully elucidated. Here, we demonstrate that bacterial CpG motifs trigger the signaling downstream of TLR9 promoting IκBNS-mediated expression of Blimp-1, a transcription regulator of IL-10. Surprisingly, this effect was counteracted by the NF-κB transcription factor c-Rel. A functional screen for intestinal bacterial species identified the commensal Clostridium sporogenes, secreting high amounts of short-chain fatty acids (SCFAs) and branched-chain fatty acids (BCFAs), as an amplifier of IL-10 production by promoting sustained mTOR signaling in B cells. Consequently, enhanced Breg functionality was achieved by combining CpG with the SCFA butyrate or the BCFA isovalerate thereby synergizing TLR- and mTOR-mediated pathways. Collectively, Bregs required two bacterial signals (butyrate and CpG) to elicit their full suppressive capacity and ameliorate T cell-mediated intestinal inflammation. Our study has dissected the molecular pathways induced by bacterial factors, which might contribute not only to better understanding of host-microbiome interactions, but also to exploration of new strategies for improvement of anti-inflammatory cellular therapy.
Collapse
Affiliation(s)
- Maik Luu
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany; Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany.
| | - Felix F Krause
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Heide Monning
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Anne Wempe
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Hanna Leister
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Lisa Mainieri
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Sarah Staudt
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kai Ziegler-Martin
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kira Mangold
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany; Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nora Kappelhoff
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Yoav D Shaul
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stephan Göttig
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt am Main, Germany
| | | | - Leon N Schulte
- Institute for Lung Research, Philipps-University Marburg, Marburg, Germany
| | | | - Ingo Schmitz
- Department of Molecular Immunology, Ruhr-University Bochum, Bochum, Germany
| | - Ulrich Steinhoff
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
2
|
Werlen G, Hernandez T, Jacinto E. Food for thought: Nutrient metabolism controlling early T cell development. Bioessays 2025; 47:e2400179. [PMID: 39504233 DOI: 10.1002/bies.202400179] [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: 07/16/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024]
Abstract
T cells develop in the thymus by expressing a diverse repertoire of either αβ- or γδ-T cell receptors (TCR). While many studies have elucidated how TCR signaling and gene expression control T cell ontogeny, the role of nutrient metabolism is just emerging. Here, we discuss how metabolic reprogramming and nutrient availability impact the fate of developing thymic T cells. We focus on how the PI3K/mTOR signaling mediates various extracellular inputs and how this signaling pathway controls metabolic rewiring during highly proliferative and anabolic developmental stages. We highlight the role of the hexosamine biosynthetic pathway that generates metabolites that are utilized for N- and O-linked glycosylation of proteins and how it impacts TCR expression during T cell ontogeny. We consider the dichotomy in metabolic needs during αβ- versus γδ-T cell lineage commitment as well as how metabolism is also coupled to molecular signaling that controls cell fate.
Collapse
Affiliation(s)
- Guy Werlen
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Tatiana Hernandez
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Estela Jacinto
- Department of Biochemistry and Molecular Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| |
Collapse
|
3
|
Yada Y, Matsumoto M, Inoue T, Baba A, Higuchi R, Kawai C, Yanagisawa M, Kitamura D, Ohga S, Kurosaki T, Baba Y. STIM-mediated calcium influx regulates maintenance and selection of germinal center B cells. J Exp Med 2024; 221:e20222178. [PMID: 37902601 PMCID: PMC10615893 DOI: 10.1084/jem.20222178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/02/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023] Open
Abstract
Positive selection of high-affinity germinal center (GC) B cells is driven by antigen internalization through their B cell receptor (BCR) and presentation to follicular helper T cells. However, the requirements of BCR signaling in GC B cells remain poorly understood. Store-operated Ca2+ entry, mediated by stromal interacting molecule 1 (STIM1) and STIM2, is the main Ca2+ influx pathway triggered by BCR engagement. Here, we showed that STIM-deficient B cells have reduced B cell competitiveness compared with wild-type B cells during GC responses. B cell-specific deletion of STIM proteins decreased the number of high-affinity B cells in the late phase of GC formation. STIM deficiency did not affect GC B cell proliferation and antigen presentation but led to the enhancement of apoptosis due to the impaired upregulation of anti-apoptotic Bcl2a1. STIM-mediated activation of NFAT was required for the expression of Bcl2a1 after BCR stimulation. These findings suggest that STIM-mediated survival signals after antigen capture regulate the optimal selection and maintenance of GC B cells.
Collapse
Affiliation(s)
- Yutaro Yada
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masanori Matsumoto
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akemi Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryota Higuchi
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Chie Kawai
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Daisuke Kitamura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
4
|
Giampaolo S, Chiarolla CM, Knöpper K, Vaeth M, Klein M, Muhammad A, Bopp T, Berberich-Siebelt F, Patra AK, Serfling E, Klein-Hessling S. NFATc1 induction by an intronic enhancer restricts NKT γδ cell formation. iScience 2023; 26:106234. [PMID: 36926655 PMCID: PMC10011748 DOI: 10.1016/j.isci.2023.106234] [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: 04/19/2022] [Revised: 06/08/2022] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
In thymus, the ablation of T cell receptor (TCR)-activated transcription factor NFATc1 or its inducible isoforms during the double-negative (DN) stages of thymocyte development leads to a marked increase in γδ thymocytes whereas the development of αβ thymocytes remains mostly unaffected. These γδ thymocytes are characterized by the upregulation of the promyelocytic leukemia zinc-finger factor (PLZF), the "master regulator" of natural killer T (NKT) cell development, and the acquisition of an NKT γδ cell phenotype with higher cell survival rates. The suppressive function of NFATc1 in NKT γδ cell formation critically depends on the remote enhancer E2, which is essential for the inducible expression of NFATc1 directed by its distal promoter P1. Thus, the enhancer deciphers a strong γδ TCR signal into the expression of inducible NFATc1 isoforms resulting in high levels of NFATc1 protein that are essential to control the numbers of NKT γδ cells.
Collapse
Affiliation(s)
- Sabrina Giampaolo
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Cristina M Chiarolla
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Konrad Knöpper
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-University Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany
| | - Martin Vaeth
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-University Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center, University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Azeem Muhammad
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Friederike Berberich-Siebelt
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Amiya K Patra
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany.,Peninsula Medical School, University of Plymouth, The John Bull Building, Plymouth Science Park, Research Way, Plymouth PL6 8BU, UK
| | - Edgar Serfling
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Stefan Klein-Hessling
- Institute of Pathology, Julius Maximilians University Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| |
Collapse
|
5
|
Duke-Cohan JS, Akitsu A, Mallis RJ, Messier CM, Lizotte PH, Aster JC, Hwang W, Lang MJ, Reinherz EL. Pre-T cell receptor self-MHC sampling restricts thymocyte dedifferentiation. Nature 2023; 613:565-574. [PMID: 36410718 PMCID: PMC9851994 DOI: 10.1038/s41586-022-05555-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/11/2022] [Indexed: 11/22/2022]
Abstract
Programming T cells to distinguish self from non-self is a vital, multi-step process that occurs in the thymus1-4. Signalling through the pre-T cell receptor (preTCR), a CD3-associated heterodimer comprising an invariant pTα chain and a clone-specific β chain, is a critical early checkpoint in thymocyte development within the αβ T cell lineage5,6. PreTCRs arrayed on CD4-CD8- double-negative thymocytes ligate peptides bound to major histocompatibility complex molecules (pMHC) on thymic stroma, similar to αβ T cell receptors that appear on CD4+CD8+ double-positive thymocytes, but via a different molecular docking strategy7-10. Here we show the consequences of these distinct interactions for thymocyte progression using synchronized fetal thymic progenitor cultures that differ in the presence or absence of pMHC on support stroma, and single-cell transcriptomes at key thymocyte developmental transitions. Although major histocompatibility complex (MHC)-negative stroma fosters αβ T cell differentiation, the absence of preTCR-pMHC interactions leads to deviant thymocyte transcriptional programming associated with dedifferentiation. Highly proliferative double-negative and double-positive thymocyte subsets emerge, with antecedent characteristics of T cell lymphoblastic and myeloid malignancies. Compensatory upregulation of diverse MHC class Ib proteins in B2m/H2-Ab1 MHC-knockout mice partially safeguards in vivo thymocyte progression, although disseminated double-positive thymic tumours may develop with ageing. Thus, as well as promoting β chain repertoire broadening for subsequent αβ T cell receptor utilization, preTCR-pMHC interactions limit cellular plasticity to facilitate normal thymocyte differentiation and proliferation that, if absent, introduce developmental vulnerabilities.
Collapse
Affiliation(s)
- Jonathan S Duke-Cohan
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Aoi Akitsu
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Robert J Mallis
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Cameron M Messier
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Patrick H Lizotte
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wonmuk Hwang
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX, USA
- Department of Physics and Astronomy, Texas A&M University, College Station, TX, USA
| | - Matthew J Lang
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Ellis L Reinherz
- Laboratory of Immunobiology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Badrani JH, Strohm AN, Lacasa L, Civello B, Cavagnero K, Haung YA, Amadeo M, Naji LH, Lund SJ, Leng A, Kim H, Baum RE, Khorram N, Mondal M, Seumois G, Pilotte J, Vanderklish PW, McGee HM, Doherty TA. RNA-binding protein RBM3 intrinsically suppresses lung innate lymphoid cell activation and inflammation partially through CysLT1R. Nat Commun 2022; 13:4435. [PMID: 35908044 PMCID: PMC9338970 DOI: 10.1038/s41467-022-32176-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
Innate lymphoid cells (ILC) promote lung inflammation in asthma through cytokine production. RNA-binding proteins (RBPs) are critical post-transcriptional regulators, although less is known about RBPs in ILC biology. Here, we demonstrate that RNA-binding motif 3 (RBM3) is highly expressed in lung ILCs and is further induced by alarmins TSLP and IL-33. Rbm3-/- and Rbm3-/-Rag2-/- mice exposed to asthma-associated Alternaria allergen develop enhanced eosinophilic lung inflammation and ILC activation. IL-33 stimulation studies in vivo and in vitro show that RBM3 suppressed lung ILC responses. Further, Rbm3-/- ILCs from bone marrow chimeric mice display increased ILC cytokine production suggesting an ILC-intrinsic suppressive function of RBM3. RNA-sequencing of Rbm3-/- lung ILCs demonstrates increased expression of type 2/17 cytokines and cysteinyl leukotriene 1 receptor (CysLT1R). Finally, Rbm3-/-Cyslt1r-/- mice show dependence on CysLT1R for accumulation of ST2+IL-17+ ILCs. Thus, RBM3 intrinsically regulates lung ILCs during allergen-induced type 2 inflammation that is partially dependent on CysLT1R.
Collapse
Affiliation(s)
- Jana H. Badrani
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Allyssa N. Strohm
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,Veterans Affairs San Diego Health Care System, La Jolla, CA USA
| | - Lee Lacasa
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Blake Civello
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Kellen Cavagnero
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Yung-An Haung
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.145695.a0000 0004 1798 0922Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Michael Amadeo
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Luay H. Naji
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Sean J. Lund
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Anthea Leng
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Hyojoung Kim
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Rachel E. Baum
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Naseem Khorram
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Monalisa Mondal
- grid.185006.a0000 0004 0461 3162La Jolla Institute, La Jolla, CA USA
| | - Grégory Seumois
- grid.185006.a0000 0004 0461 3162La Jolla Institute, La Jolla, CA USA
| | - Julie Pilotte
- grid.214007.00000000122199231The Scripps Research Institute, La Jolla, CA USA
| | | | - Heather M. McGee
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,grid.250671.70000 0001 0662 7144NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute, La Jolla, CA USA ,grid.410425.60000 0004 0421 8357Departments of Radiation Oncology and Immuno-Oncology, City of Hope, Duarte, CA USA ,Department of Molecular Medicine, La Jolla, CA USA
| | - Taylor A. Doherty
- grid.266100.30000 0001 2107 4242Divison of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA USA ,Veterans Affairs San Diego Health Care System, La Jolla, CA USA
| |
Collapse
|
7
|
Last but not least: BFL-1 as an emerging target for anti-cancer therapies. Biochem Soc Trans 2022; 50:1119-1128. [PMID: 35900226 PMCID: PMC9444066 DOI: 10.1042/bst20220153] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
BFL-1 is an understudied pro-survival BCL-2 protein. The expression of BFL-1 is reported in many cancers, but it is yet to be clarified whether high transcript expression also always correlates with a pro-survival function. However, recent applications of BH3-mimetics for the treatment of blood cancers identified BFL-1 as a potential resistance factor in this type of cancer. Hence, understanding the role of BFL-1 in human cancers and how its up-regulation leads to therapy resistance has become an area of great clinical relevance. In addition, deletion of the murine homologue of BFL-1, called A1, in mice showed only minimal impacts on the well-being of these animals, suggesting drugs targeting BFL-1 would exhibit limited on-target toxicities. BFL-1 therefore represents a good clinical cancer target. Currently, no effective BFL-1 inhibitors exist, which is likely due to the underappreciation of BFL-1 as a potential target in the clinic and lack of understanding of the BFL-1 protein. In this review, the roles of BFL-1 in the development of different types of cancers and drug resistant mechanisms are discussed and some recent advances in the generation of BFL-1 inhibitors highlighted.
Collapse
|
8
|
Zheng Q, Gan G, Gao X, Luo Q, Chen F. Targeting the IDO-BCL2A1-Cytochrome c Pathway Promotes Apoptosis in Oral Squamous Cell Carcinoma. Onco Targets Ther 2021; 14:1673-1687. [PMID: 33707952 PMCID: PMC7942955 DOI: 10.2147/ott.s288692] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/04/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Indolamine 2,3-dioxygenase (IDO) is the rate limiting enzyme of tryptophan degradation and is a negative prognostic factor in oral squamous cell carcinoma (OSCC) patients, while the underlying molecular mechanism remains unclear. This research aimed to explore the IDO expression and its biological functions in OSCC. Materials and Methods IDO expression was analyzed by qPCR, Western blots, and immunohistochemistry (IHC) in OSCC cell lines and tissue specimens. Tryptophan and kynurenine content were determined by UPLC-MS/MS in serum samples of OSCC patients and healthy controls. Oncomine databases and Kaplan-Meier survival analyses were used to identify the IDO expression and its correlation with OSCC prognosis. Cell counting, CCK8 assay, flow cytometry, cell cycle, and EdU incorporation assays were used to assess the effect of IDO inhibition on OSCC growth either by shRNA or the IDO-specific inhibitor (epacadostat) in vitro. An OSCC xenograft mouse model was established to verify the predicted function of IDO inhibition in vivo. Mechanistically, an 84-gene apoptosis PCR array and rescue experiment were used to characterize the underlying mechanism involved in IDO-regulated apoptosis in OSCC. Results IDO expression was upregulated in OSCC cell lines and tissues and was negatively correlated with OSCC progression. Lentivirus-mediated IDO knockdown and epacadostat significantly reduced viability and promoted apoptosis of OSCC cells in vitro and in vivo. The apoptosis PCR array identified BCL2 related protein A1 (BCL2A1) as the most obviously changed gene at the transcriptional level. IDO inhibition downregulated BCL2A1 expression, increased the expression and translocation of cytochrome c, thus promoted apoptosis in OSCC. Overexpression of BCL2A1 reversed the pro-apoptotic effect of IDO inhibition. Conclusion The present results revealed that IDO directly affect the growth of OSCC cells by regulating BCL2A1 expression. IDO and the IDO-BCL2A1-cytochrome c axis may be potential therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Qiaoping Zheng
- Department of Clinical Immunology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Guifang Gan
- Department of Clinical Immunology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Xianfu Gao
- Shanghai Profleader Biotech Co., Ltd., Shanghai, People's Republic of China
| | - Qingqiong Luo
- Department of Clinical Immunology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Fuxiang Chen
- Department of Clinical Immunology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| |
Collapse
|
9
|
Seumois G, Ramírez-Suástegui C, Schmiedel BJ, Liang S, Peters B, Sette A, Vijayanand P. Single-cell transcriptomic analysis of allergen-specific T cells in allergy and asthma. Sci Immunol 2021; 5:5/48/eaba6087. [PMID: 32532832 DOI: 10.1126/sciimmunol.aba6087] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/22/2020] [Indexed: 12/19/2022]
Abstract
CD4+ T helper (TH) cells and regulatory T (Treg) cells that respond to common allergens play an important role in driving and dampening airway inflammation in patients with asthma. Until recently, direct, unbiased molecular analysis of allergen-reactive TH and Treg cells has not been possible. To better understand the diversity of these T cell subsets in allergy and asthma, we analyzed the single-cell transcriptome of ~50,000 house dust mite (HDM) allergen-reactive TH cells and Treg cells from asthmatics with HDM allergy and from three control groups: asthmatics without HDM allergy and nonasthmatics with and without HDM allergy. Our analyses show that HDM allergen-reactive TH and Treg cells are highly heterogeneous and certain subsets are quantitatively and qualitatively different in individuals with HDM-reactive asthma. The number of interleukin-9 (IL-9)-expressing HDM-reactive TH cells is greater in asthmatics with HDM allergy compared with nonasthmatics with HDM allergy, and this IL-9-expressing TH subset displays enhanced pathogenic properties. More HDM-reactive TH and Treg cells expressing the interferon response signature (THIFNR and TregIFNR) are present in asthmatics without HDM allergy compared with those with HDM allergy. In cells from these subsets (THIFNR and TregIFNR), expression of TNFSF10 was enriched; its product, tumor necrosis factor-related apoptosis-inducing ligand, dampens activation of TH cells. These findings suggest that the THIFNR and TregIFNR subsets may dampen allergic responses, which may help explain why only some people develop TH2 responses to nearly ubiquitous allergens.
Collapse
Affiliation(s)
- Grégory Seumois
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | - Shu Liang
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Pandurangan Vijayanand
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA. .,Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA.,Clinical and Experimental Sciences, National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Faculty of Medicine, University of Southampton, Southampton SO166YD, UK
| |
Collapse
|
10
|
Role of Notch Receptors in Hematologic Malignancies. Cells 2020; 10:cells10010016. [PMID: 33374160 PMCID: PMC7823720 DOI: 10.3390/cells10010016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Notch receptors are single-pass transmembrane proteins that play a critical role in cell fate decisions and have been implicated in the regulation of many developmental processes. The human Notch family comprises of four receptors (Notch 1 to 4) and five ligands. Their signaling can regulate extremely basic cellular processes such as differentiation, proliferation and death. Notch is also involved in hematopoiesis and angiogenesis, and increasing evidence suggests that these genes are involved and frequently deregulated in several human malignancies, contributing to cell autonomous activities that may be either oncogenic or tumor suppressive. It was recently proposed that Notch signaling could play an active role in promoting and sustaining a broad spectrum of lymphoid malignancies as well as mutations in Notch family members that are present in several disorders of T- and B-cells, which could be responsible for altering the related signaling. Therefore, different Notch pathway molecules could be considered as potential therapeutic targets for hematological cancers. In this review, we will summarize and discuss compelling evidence pointing to Notch receptors as pleiotropic regulators of hematologic malignancies biology, first describing the physiological role of their signaling in T- and B-cell development and homeostasis, in order to fully understand the pathological alterations reported.
Collapse
|
11
|
Abou-Fadel J, Vasquez M, Grajeda B, Ellis C, Zhang J. Systems-wide analysis unravels the new roles of CCM signal complex (CSC). Heliyon 2019; 5:e02899. [PMID: 31872111 PMCID: PMC6909108 DOI: 10.1016/j.heliyon.2019.e02899] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/17/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are characterized by abnormally dilated intracranial capillaries that result in increased susceptibility to stroke. Three genes have been identified as causes of CCMs; KRIT1 (CCM1), MGC4607 (CCM2) and PDCD10 (CCM3); one of them is disrupted in most CCM cases. It was demonstrated that both CCM1 and CCM3 bind to CCM2 to form a CCM signaling complex (CSC) to modulate angiogenesis. In this report, we deployed both RNA-seq and proteomic analysis of perturbed CSC after depletion of one of three CCM genes to generate interactomes for system-wide studies. Our results demonstrated a unique portrait detailing alterations in angiogenesis and vascular integrity. Interestingly, only in-direct overlapped alterations between RNA and protein levels were detected, supporting the existence of multiple layers of regulation in CSC cascades. Notably, this is the first report identifying that both β4 integrin and CAV1 signaling are downstream of CSC, conveying the angiogenic signaling. Our results provide a global view of signal transduction modulated by the CSC, identifies novel regulatory signaling networks and key cellular factors associated with CSC.
Collapse
Affiliation(s)
- Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Mariana Vasquez
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Brian Grajeda
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Cameron Ellis
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| |
Collapse
|
12
|
An integrated transcriptional switch at the β-selection checkpoint determines T cell survival, development and leukaemogenesis. Biochem Soc Trans 2019; 47:1077-1089. [DOI: 10.1042/bst20180414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023]
Abstract
Abstract
In T cell development, a pivotal decision-making stage, termed β-selection, integrates a TCRβ checkpoint to coordinate survival, proliferation and differentiation to an αβ T cell. Here, we review how transcriptional regulation coordinates fate determination in early T cell development to enable β-selection. Errors in this transcription control can trigger T cell acute lymphoblastic leukaemia. We describe how the β-selection checkpoint goes awry in leukaemic transformation.
Collapse
|
13
|
Saadi W, Kermezli Y, Dao LTM, Mathieu E, Santiago-Algarra D, Manosalva I, Torres M, Belhocine M, Pradel L, Loriod B, Aribi M, Puthier D, Spicuglia S. A critical regulator of Bcl2 revealed by systematic transcript discovery of lncRNAs associated with T-cell differentiation. Sci Rep 2019; 9:4707. [PMID: 30886319 PMCID: PMC6423290 DOI: 10.1038/s41598-019-41247-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/01/2019] [Indexed: 12/30/2022] Open
Abstract
Normal T-cell differentiation requires a complex regulatory network which supports a series of maturation steps, including lineage commitment, T-cell receptor (TCR) gene rearrangement, and thymic positive and negative selection. However, the underlying molecular mechanisms are difficult to assess due to limited T-cell models. Here we explore the use of the pro-T-cell line P5424 to study early T-cell differentiation. Stimulation of P5424 cells by the calcium ionophore ionomycin together with PMA resulted in gene regulation of T-cell differentiation and activation markers, partially mimicking the CD4-CD8- double negative (DN) to double positive (DP) transition and some aspects of subsequent T-cell maturation and activation. Global analysis of gene expression, along with kinetic experiments, revealed a significant association between the dynamic expression of coding genes and neighbor lncRNAs including many newly-discovered transcripts, thus suggesting potential co-regulation. CRISPR/Cas9-mediated genetic deletion of Robnr, an inducible lncRNA located downstream of the anti-apoptotic gene Bcl2, demonstrated a critical role of the Robnr locus in the induction of Bcl2. Thus, the pro-T-cell line P5424 is a powerful model system to characterize regulatory networks involved in early T-cell differentiation and maturation.
Collapse
Affiliation(s)
- Wiam Saadi
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Yasmina Kermezli
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Lan T M Dao
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Vinmec Research Institute of Stem cell and Gene technology (VRISG), Hanoi, Vietnam
| | - Evelyne Mathieu
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - David Santiago-Algarra
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Iris Manosalva
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Magali Torres
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Mohamed Belhocine
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Molecular Biology and Genetics Laboratory, Dubai, United Arab Emirates
| | - Lydie Pradel
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Beatrice Loriod
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Denis Puthier
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France. .,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.
| | - Salvatore Spicuglia
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France. .,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.
| |
Collapse
|
14
|
Olivas-Aguirre M, Pottosin I, Dobrovinskaya O. Mitochondria as emerging targets for therapies against T cell acute lymphoblastic leukemia. J Leukoc Biol 2019; 105:935-946. [PMID: 30698851 DOI: 10.1002/jlb.5vmr0818-330rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/29/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) comprises a heterogeneous group of hematologic malignancies, arising from diverse genetic alterations in the early lymphocyte development. T-cell subtype of ALL (T-ALL) accounts for about 15% and 25% of ALL in children and adults, respectively. Being less frequent among ALL subtypes, T-ALL represents a high-risk factor for poor prognosis due to its aggressiveness and resistance to common antileukemic drugs. Mitochondria were widely explored recently as a target for anticancer treatment because they are involved in a metabolic reprogramming of a cancer cell and play key roles in reactive oxygen species generation, Ca2+ signaling, and cell death induction. Accordingly, a new class of anticancer compounds named mitocans has been developed, which target mitochondria at distinct crucial points to promote their dysfunction and subsequent cell death. The present review analyses the role of mitochondria in malignant reprogramming and emerging therapeutic strategies targeting mitochondria as an "Achilles' heel" in T-ALL, with an emphasis on BH3 mimetics, sequestering pro-survival BCL proteins and voltage-dependent anion channel (VDAC)1-directed drugs, which promote the suppression of aerobic glycolysis, VDAC1 closure, mitochondrial Ca2+ overload, stoppage of the oxidative phosphorylation, oxidative stress, and release of proapoptotic factors.
Collapse
Affiliation(s)
- Miguel Olivas-Aguirre
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Colima, Mexico
| | - Igor Pottosin
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Colima, Mexico
| | - Oxana Dobrovinskaya
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Colima, Mexico
| |
Collapse
|
15
|
Chatterjee S, Chakraborty P, Daenthanasanmak A, Iamsawat S, Andrejeva G, Luevano LA, Wolf M, Baliga U, Krieg C, Beeson CC, Mehrotra M, Hill EG, Rathmell JC, Yu XZ, Kraft AS, Mehrotra S. Targeting PIM Kinase with PD1 Inhibition Improves Immunotherapeutic Antitumor T-cell Response. Clin Cancer Res 2018; 25:1036-1049. [PMID: 30327305 DOI: 10.1158/1078-0432.ccr-18-0706] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/29/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023]
Abstract
PURPOSE Adoptive T-cell therapy (ACT) of cancer, which involves the infusion of ex vivo-engineered tumor epitope reactive autologous T cells into the tumor-bearing host, is a potential treatment modality for cancer. However, the durable antitumor response following ACT is hampered either by loss of effector function or survival of the antitumor T cells. Therefore, strategies to improve the persistence and sustain the effector function of the antitumor T cells are of immense importance. Given the role of metabolism in determining the therapeutic efficacy of T cells, we hypothesize that inhibition of PIM kinases, a family of serine/threonine kinase that promote cell-cycle transition, cell growth, and regulate mTORC1 activity, can improve the potency of T cells in controlling tumor. EXPERIMENTAL DESIGN The role of PIM kinases in T cells was studied either by genetic ablation (PIM1-/-PIM2-/-PIM3-/-) or its pharmacologic inhibition (pan-PIM kinase inhibitor, PimKi). Murine melanoma B16 was established subcutaneously and treated by transferring tumor epitope gp100-reactive T cells along with treatment regimen that involved inhibiting PIM kinases, anti-PD1 or both. RESULTS With inhibition of PIM kinases, T cells had significant reduction in their uptake of glucose, and upregulated expression of memory-associated genes that inversely correlate with glycolysis. In addition, the expression of CD38, which negatively regulates the metabolic fitness of the T cells, was also reduced in PimKi-treated cells. Importantly, the efficacy of antitumor T-cell therapy was markedly improved by inhibiting PIM kinases in tumor-bearing mice receiving ACT, and further enhanced by adding anti-PD1 antibody to this combination. CONCLUSIONS This study highlights the potential therapeutic significance of combinatorial strategies where ACT and inhibition of signaling kinase with checkpoint blockade could improve tumor control.
Collapse
Affiliation(s)
- Shilpak Chatterjee
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Anusara Daenthanasanmak
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Supinya Iamsawat
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Gabriela Andrejeva
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Libia A Luevano
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Melissa Wolf
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Uday Baliga
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Carsten Krieg
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Craig C Beeson
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Meenal Mehrotra
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Elizabeth G Hill
- Department of Public Health, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Jeffery C Rathmell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Andrew S Kraft
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
16
|
Li KP, Shanmuganad S, Carroll K, Katz JD, Jordan MB, Hildeman DA. Dying to protect: cell death and the control of T-cell homeostasis. Immunol Rev 2017; 277:21-43. [PMID: 28462527 PMCID: PMC5416827 DOI: 10.1111/imr.12538] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 02/07/2023]
Abstract
T cells play a critical role in immune responses as they specifically recognize peptide/MHC complexes with their T-cell receptors and initiate adaptive immune responses. While T cells are critical for performing appropriate effector functions and maintaining immune memory, they also can cause autoimmunity or neoplasia if misdirected or dysregulated. Thus, T cells must be tightly regulated from their development onward. Maintenance of appropriate T-cell homeostasis is essential to promote protective immunity and limit autoimmunity and neoplasia. This review will focus on the role of cell death in maintenance of T-cell homeostasis and outline novel therapeutic strategies tailored to manipulate cell death to limit T-cell survival (eg, autoimmunity and transplantation) or enhance T-cell survival (eg, vaccination and immune deficiency).
Collapse
Affiliation(s)
- Kun-Po Li
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sharmila Shanmuganad
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kaitlin Carroll
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jonathan D. Katz
- Division of Immunobiology, Cincinnati, OH 45229, USA
- Division of Endocrinology, Diabetes Research Center, Cincinnati, OH 45229, USA
| | - Michael B. Jordan
- Division of Immunobiology, Cincinnati, OH 45229, USA
- Division of Bone Marrow Transplantation and Immune Deficiency, Department of Pediatrics, Cincinnati Children’s Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | |
Collapse
|
17
|
Daley SR, Teh C, Hu DY, Strasser A, Gray DH. Cell death and thymic tolerance. Immunol Rev 2017; 277:9-20. [DOI: 10.1111/imr.12532] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Stephen R. Daley
- Infection and Immunity Program; Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology; Monash University; Melbourne VIC Australia
| | - Charis Teh
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| | | | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| | - Daniel H.D. Gray
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| |
Collapse
|
18
|
Martinez-Gonzalez I, Mathä L, Steer CA, Takei F. Immunological Memory of Group 2 Innate Lymphoid Cells. Trends Immunol 2017; 38:423-431. [PMID: 28416448 DOI: 10.1016/j.it.2017.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 01/25/2023]
Abstract
Immunological memory has long been described as a property of the adaptive immune system that results in potent responses on exposure to an antigen encountered previously. While this definition appears to exclude cells that do not express antigen receptors, recent studies have shown that innate immune cells, including natural killer (NK) cells, macrophages, and, more recently, group 2 innate lymphoid cells (ILC2s) can record previous activations and respond more vigorously on reactivation. Here we review the similarities and differences between these forms of memory and the underlying mechanisms. Based on these insights, we propose to revise the definition of immunological memory, as the capacity to remember being previously activated and respond more efficiently on reactivation regardless of antigen specificity.
Collapse
Affiliation(s)
- Itziar Martinez-Gonzalez
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Laura Mathä
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Catherine A Steer
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Fumio Takei
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada; Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
19
|
Tuzlak S, Schenk RL, Vasanthakumar A, Preston SP, Haschka MD, Zotos D, Kallies A, Strasser A, Villunger A, Herold MJ. The BCL-2 pro-survival protein A1 is dispensable for T cell homeostasis on viral infection. Cell Death Differ 2017; 24:523-533. [PMID: 28085151 DOI: 10.1038/cdd.2016.155] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/08/2016] [Accepted: 12/01/2016] [Indexed: 12/31/2022] Open
Abstract
The physiological role of the pro-survival BCL-2 family member A1 has been debated for a long time. Strong mRNA induction in T cells on T cell receptor (TCR)-engagement suggested a major role of A1 in the survival of activated T cells. However, the investigation of the physiological roles of A1 was complicated by the quadruplication of the A1 gene locus in mice, making A1 gene targeting very difficult. Here, we used the recently generated A1-/- mouse model to examine the role of A1 in T cell immunity. We confirmed rapid and strong induction of A1 protein in response to TCR/CD3 stimulation in CD4+ as well as CD8+ T cells. Surprisingly, on infection with the acute influenza HKx31 or the lymphocytic choriomeningitis virus docile strains mice lacking A1 did not show any impairment in the expansion, survival, or effector function of cytotoxic T cells. Furthermore, the ability of A1-/- mice to generate antigen-specific memory T cells or to provide adequate CD4-dependent help to B cells was not impaired. These results suggest functional redundancy of A1 with other pro-survival BCL-2 family members in the control of T cell-dependent immune responses.
Collapse
Affiliation(s)
- Selma Tuzlak
- Division of Developmental Immunology, BIOCENTER, Medical University Innsbruck, Innsbruck, Austria.,The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia
| | - Robyn L Schenk
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Ajithkumar Vasanthakumar
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Simon P Preston
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Manuel D Haschka
- Division of Developmental Immunology, BIOCENTER, Medical University Innsbruck, Innsbruck, Austria
| | - Dimitra Zotos
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Axel Kallies
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Andreas Strasser
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| | - Andreas Villunger
- Division of Developmental Immunology, BIOCENTER, Medical University Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Marco J Herold
- The Walter & Eliza Hall Institute for Medical Research, Parkville, Melbourne, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3050, Australia
| |
Collapse
|
20
|
Characterisation of mice lacking all functional isoforms of the pro-survival BCL-2 family member A1 reveals minor defects in the haematopoietic compartment. Cell Death Differ 2017; 24:534-545. [PMID: 28085150 DOI: 10.1038/cdd.2016.156] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/08/2016] [Accepted: 12/01/2016] [Indexed: 12/31/2022] Open
Abstract
The pro-survival proteins of the BCL-2 family regulate the survival of all cells, and genetic deletion models for these proteins have revealed which specific BCL-2 family member(s) is/are critical for the survival of particular cell types. A1 is a pro-survival BCL-2-like protein that is expressed predominantly in haematopoietic cells, and here we describe the characterisation of a novel mouse strain that lacks all three functional isoforms of A1 (A1-a, A1-b and A1-d). Surprisingly, complete loss of A1 caused only minor defects, with significant, although relatively small, decreases in γδTCR T cells, antigen-experienced conventional as well as regulatory CD4 T cells and conventional dendritic cells (cDCs). When examining these cell types in tissue culture, only cDC survival was significantly impaired by the loss of A1. Therefore, A1 appears to be a surprisingly redundant pro-survival protein in the haematopoietic system and other tissues, suggesting that its targeting in cancer may be readily tolerated.
Collapse
|
21
|
Pham K, Shimoni R, Charnley M, Ludford-Menting MJ, Hawkins ED, Ramsbottom K, Oliaro J, Izon D, Ting SB, Reynolds J, Lythe G, Molina-Paris C, Melichar H, Robey E, Humbert PO, Gu M, Russell SM. Asymmetric cell division during T cell development controls downstream fate. J Cell Biol 2015; 210:933-50. [PMID: 26370500 PMCID: PMC4576854 DOI: 10.1083/jcb.201502053] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
T cell precursors undergo asymmetric cell division after T cell receptor genomic recombination, with stromal cell cues controlling the differential inheritance of fate determinants Numb and α-Adaptin by the daughters of a dividing DN3a T cell precursor. During mammalian T cell development, the requirement for expansion of many individual T cell clones, rather than merely expansion of the entire T cell population, suggests a possible role for asymmetric cell division (ACD). We show that ACD of developing T cells controls cell fate through differential inheritance of cell fate determinants Numb and α-Adaptin. ACD occurs specifically during the β-selection stage of T cell development, and subsequent divisions are predominantly symmetric. ACD is controlled by interaction with stromal cells and chemokine receptor signaling and uses a conserved network of polarity regulators. The disruption of polarity by deletion of the polarity regulator, Scribble, or the altered inheritance of fate determinants impacts subsequent fate decisions to influence the numbers of DN4 cells arising after the β-selection checkpoint. These findings indicate that ACD enables the thymic microenvironment to orchestrate fate decisions related to differentiation and self-renewal.
Collapse
Affiliation(s)
- Kim Pham
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Raz Shimoni
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Mirren Charnley
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia Industrial Research Institute Swinburne, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Mandy J Ludford-Menting
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Edwin D Hawkins
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Kelly Ramsbottom
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Jane Oliaro
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - David Izon
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Stephen B Ting
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia
| | - Joseph Reynolds
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, England, UK
| | - Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, England, UK
| | - Carmen Molina-Paris
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, England, UK
| | - Heather Melichar
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Ellen Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Patrick O Humbert
- Cell Cycle and Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Min Gu
- Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Sarah M Russell
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia Centre for Micro-Photonics, Faculty of Science, Engineering, and Technology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia Department of Pathology, University of Melbourne, Parkville, Victoria 3010, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
22
|
Sochalska M, Ottina E, Tuzlak S, Herzog S, Herold M, Villunger A. Conditional knockdown of BCL2A1 reveals rate-limiting roles in BCR-dependent B-cell survival. Cell Death Differ 2015; 23:628-39. [PMID: 26450454 PMCID: PMC4986635 DOI: 10.1038/cdd.2015.130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/19/2015] [Accepted: 08/27/2015] [Indexed: 11/09/2022] Open
Abstract
Bcl2 family proteins control mitochondrial apoptosis and its members exert critical cell type and differentiation stage-specific functions, acting as barriers against autoimmunity or transformation. Anti-apoptotic Bcl2a1/Bfl1/A1 is frequently deregulated in different types of blood cancers in humans but its physiological role is poorly understood as quadruplication of the Bcl2a1 gene locus in mice hampers conventional gene targeting strategies. Transgenic overexpression of A1, deletion of the A1-a paralogue or constitutive knockdown in the hematopoietic compartment of mice by RNAi suggested rate-limiting roles in lymphocyte development, granulopoiesis and mast cell activation. Here we report on the consequences of conditional knockdown of A1 protein expression using a reverse transactivator (rtTA)-driven approach that highlights a critical role for this Bcl2 family member in the maintenance of mature B-cell homeostasis. Furthermore, we define the A1/Bim (Bcl-2 interacting mediator of cell death) axis as a target of key kinases mediating B-cell receptor (BCR)-dependent survival signals, such as, spleen tyrosine kinase (Syk) and Brutons tyrosine kinase (Btk). As such, A1 represents a putative target for the treatment of B-cell-related pathologies depending on hyperactivation of BCR-emanating survival signals and loss of A1 expression accounts, in part, for the pro-apoptotic effects of Syk- or Btk inhibitors that rely on the ‘BH3-only' protein Bim for cell killing.
Collapse
Affiliation(s)
- M Sochalska
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - E Ottina
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - S Tuzlak
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - S Herzog
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - M Herold
- The Walter and Eliza Hall Institute for Medical Research, University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - A Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
23
|
López-Rodríguez C, Aramburu J, Berga-Bolaños R. Transcription factors and target genes of pre-TCR signaling. Cell Mol Life Sci 2015; 72:2305-21. [PMID: 25702312 PMCID: PMC11113633 DOI: 10.1007/s00018-015-1864-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/22/2015] [Accepted: 02/16/2015] [Indexed: 11/27/2022]
Abstract
Almost 30 years ago pioneering work by the laboratories of Harald von Boehmer and Susumo Tonegawa provided the first indications that developing thymocytes could assemble a functional TCRβ chain-containing receptor complex, the pre-TCR, before TCRα expression. The discovery and study of the pre-TCR complex revealed paradigms of signaling pathways in control of cell survival and proliferation, and culminated in the recognition of the multifunctional nature of this receptor. As a receptor integrated in a dynamic developmental process, the pre-TCR must be viewed not only in the light of the biological outcomes it promotes, but also in context with those molecular processes that drive its expression in thymocytes. This review article focuses on transcription factors and target genes activated by the pre-TCR to drive its different outcomes.
Collapse
Affiliation(s)
- Cristina López-Rodríguez
- Immunology Unit, Department of Experimental and Health Sciences and Barcelona Biomedical Research Park, Universitat Pompeu Fabra, C/Doctor Aiguader Nº88, 08003, Barcelona, Barcelona, Spain,
| | | | | |
Collapse
|
24
|
Lauritsen JPH, Boding L, Buus TB, Kongsbak M, Levring TB, Rode AKO, Bonefeld CM, Geisler C. Fine-tuning of T-cell development by the CD3γ di-leucine-based TCR-sorting motif. Int Immunol 2015; 27:393-404. [DOI: 10.1093/intimm/dxv022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/24/2015] [Indexed: 01/13/2023] Open
|
25
|
Sochalska M, Tuzlak S, Egle A, Villunger A. Lessons from gain- and loss-of-function models of pro-survival Bcl2 family proteins: implications for targeted therapy. FEBS J 2015; 282:834-849. [PMID: 25559680 PMCID: PMC4562365 DOI: 10.1111/febs.13188] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/22/2014] [Accepted: 01/02/2015] [Indexed: 01/23/2023]
Abstract
Cell survival depends on the maintenance of mitochondrial integrity controlled by a well-balanced interplay between anti- and pro-apoptotic B cell lymphoma 2 (Bcl2) family members. Given their frequent deregulation in human pathologies, including autoimmunity and cancer, significant research efforts have increased our molecular understanding of how Bcl2 proteins control cell death. This has fostered the development of small non-peptidic compounds, so-called BH3-mimetics, that show excellent prospects of passing clinical trials and entering daily use for targeted therapy. Possible limitations in clinical application may, to a certain degree, be predicted from loss-of-function phenotypes gathered from studies using gene-modified mice that we attempt to summarize and discuss in this context.
Collapse
Affiliation(s)
- Maja Sochalska
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Austria
| | - Selma Tuzlak
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Austria
| | - Alexander Egle
- Laboratory for Immunological and Molecular Cancer Research, Third Medical Department, Paracelsus Medical University, Salzburg, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Austria
| |
Collapse
|
26
|
Hind CK, Carter MJ, Harris CL, Chan HTC, James S, Cragg MS. Role of the pro-survival molecule Bfl-1 in melanoma. Int J Biochem Cell Biol 2014; 59:94-102. [PMID: 25486183 DOI: 10.1016/j.biocel.2014.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/11/2014] [Accepted: 11/28/2014] [Indexed: 11/16/2022]
Abstract
Bfl-1 is a pro-survival Bcl-2 family member overexpressed in a subset of chemoresistant tumours, including melanoma. Here, we characterised the expression and regulation of Bfl-1 in normal and malignant melanocytes and determined its role in protecting these cells from chemotherapy-induced apoptosis. Bfl-1 was mitochondrially resident in both resting and apoptotic cells and experienced regulation by the proteasome and NFκB pathways. siRNA-mediated knockdown enhanced sensitivity towards various relevant drug treatments, with forced overexpression of Bfl-1 protective. These findings identify Bfl-1 as a contributor towards therapeutic resistance in melanoma cells and support the use of NFκB inhibitors alongside current treatment strategies.
Collapse
Affiliation(s)
- C K Hind
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | - M J Carter
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | - C L Harris
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | - H T C Chan
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | - S James
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | - M S Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
27
|
Loss of IP3R-dependent Ca2+ signalling in thymocytes leads to aberrant development and acute lymphoblastic leukemia. Nat Commun 2014; 5:4814. [PMID: 25215520 DOI: 10.1038/ncomms5814] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/25/2014] [Indexed: 01/28/2023] Open
Abstract
Calcium ions (Ca(2+)) function as universal second messengers in eukaryotic cells, including immune cells. Ca(2+) is crucial for peripheral T-lymphocyte activation and effector functions, and influences thymocyte selection and motility in the developing thymus. However, the role of Ca(2+) signalling in early T-lymphocyte development is not well understood. Here we show that the inositol triphosphate receptors (IP3Rs) Ca(2+) ion channels are required for proliferation, survival and developmental progression of T-lymphocyte precursors. Our studies indicate that signalling via IP3Rs represses Sox13, an antagonist of the developmentally important transcription factor Tcf-1. In the absence of IP3R-mediated Ca(2+) signalling, repression of key Notch transcriptional targets--including Hes1--fail to occur in post β-selection thymocytes, and mice develop aggressive T-cell malignancies that resemble human T-cell acute lymphoblastic leukemia (T-ALL). These data indicate that IP3R-mediated Ca(2+) signalling reinforces Tcf-1 activity to both ensure normal development and prevent thymocyte neoplasia.
Collapse
|
28
|
Hampson P, Wang K, Ersvær E, McCormack E, Schüler J, Fiebig HH, Gjertsen BT, Bruserud Ø, Lord JM. Up-regulation of anti-apoptotic genes confers resistance to the novel anti-leukaemic compound PEP005 in primary AML cells. Oncoscience 2014; 1:529-39. [PMID: 25594060 PMCID: PMC4278328 DOI: 10.18632/oncoscience.71] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/02/2014] [Indexed: 11/25/2022] Open
Abstract
We showed previously that PEP005 induced apoptosis in leukaemic cell lines and blasts from patients with acute myeloid leukaemia (AML). Here we assess the anti-leukeamic effects of PEP005 in vivo and determine the mechanism of resistance of PEP005 non-responsive cells. We used 2 human xenograft mouse models of AML to assess the anti-leukaemic effects of PEP005 in vivo. Expression microarray analysis of primary AML blasts following treatment with PEP005 was used to determine patterns of gene expression that conferred resistance. PEP005 significantly reduced tumour burden in two human leukaemia mouse xenograft models. We also assessed responsiveness of 33 AML samples to PEP005, with 78% of the samples entering apoptosis at 100nM. Resistance to PEP005 was not restricted to a particular AML subtype. Expression microarray analysis of resistant samples following treatment with PEP005 revealed a significant up regulation of the anti-apoptotic genes Bcl-2A1, Mcl-1, and PHLDA1 which was verified using RT-PCR. We conclude that PEP005 shows broad efficacy against AML subtypes and that up regulation of anti-apoptotic genes underlies resistance to this agent and could be used to screen for patients unlikely to benefit from a therapeutic regime involving PEP005.
Collapse
Affiliation(s)
- Peter Hampson
- School of Immunity and Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Keqing Wang
- Department of Biosciences, University of Aston, Birmingham, B4 7ET
| | - Elisabeth Ersvær
- Institute of Medicine, University of Bergen, Bergen, NO-5020, Norway
| | - Emmet McCormack
- Institute of Medicine, University of Bergen, Bergen, NO-5020, Norway
| | | | | | | | - Øystein Bruserud
- Institute of Medicine, University of Bergen, Bergen, NO-5020, Norway
| | - Janet M Lord
- School of Immunity and Infection, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
29
|
Gerondakis S, Fulford TS, Messina NL, Grumont RJ. NF-κB control of T cell development. Nat Immunol 2014; 15:15-25. [PMID: 24352326 DOI: 10.1038/ni.2785] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 11/12/2013] [Indexed: 12/12/2022]
Abstract
The NF-κB signal transduction pathway is best known as a major regulator of innate and adaptive immune responses, yet there is a growing appreciation of its importance in immune cell development, particularly of T lineage cells. In this Review, we discuss how the temporal regulation of NF-κB controls the stepwise differentiation and antigen-dependent selection of conventional and specialized subsets of T cells in response to T cell receptor and costimulatory, cytokine and growth factor signals.
Collapse
Affiliation(s)
- Steve Gerondakis
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Thomas S Fulford
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Nicole L Messina
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Raelene J Grumont
- The Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University Central Clinical School, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
NFAT5 induction by the pre-T-cell receptor serves as a selective survival signal in T-lymphocyte development. Proc Natl Acad Sci U S A 2013; 110:16091-6. [PMID: 24043824 DOI: 10.1073/pnas.1215934110] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The Rel-like transcription factors nuclear factor kappa B (NF-κB) and the calcineurin-dependent nuclear factor of activated T cells (NFATc) control specific points of thymocyte maturation. Thymocytes also express a distinct member of the Rel family, the calcineurin-independent, osmostress response regulator NFAT5. Here we show that IKKβ regulates the expression of NFAT5 in thymocytes, which in turn contributes to the survival of T-cell receptor αβ thymocytes and the transition from the β-selection checkpoint to the double-positive stage in an osmostress-independent manner. NFAT5-deficient thymocytes had normal expression and proximal signaling of the pre-T-cell receptor but exhibited a partial defect in β-chain allelic exclusion and increased apoptosis. Further analysis showed that NFAT5 regulated the expression of the prosurvival factors A1 and Bcl2 and attenuated the proapoptotic p53/Noxa axis. These findings position NFAT5 as a target of the IKKβ/NF-κB pathway in thymocytes and as a downstream effector of the prosurvival role of the pre-T-cell receptor.
Collapse
|
31
|
Lei F, Song J, Haque R, Haque M, Xiong X, Fang D, Croft M, Song J. Regulation of A1 by OX40 contributes to CD8(+) T cell survival and anti-tumor activity. PLoS One 2013; 8:e70635. [PMID: 23936461 PMCID: PMC3731243 DOI: 10.1371/journal.pone.0070635] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 06/25/2013] [Indexed: 12/28/2022] Open
Abstract
The TNFR family member OX40 (CD134) is critical for optimal clonal expansion and survival of T cells. However, the intracellular targets of OX40 in CD8 T cells are not fully understood. Here we show that A1, a Bcl-2 family protein, is regulated by OX40 in effector CD8 T cells. In contrast to wild-type T cells, OX40-deficient CD8 T cells failed to maintain A1 expression driven by antigen. Conversely, enforced OX40 stimulation promoted A1 expression. In both situations, the expression of A1 directly correlated with CD8 T cell survival. In addition, exogenous expression of A1 in OX40-deficient CD8 T cells reversed their survival defect in vitro and in vivo. Moreover, forced expression of A1 in CD8 T cells from OX40-deficient mice restored the ability of these T cells to suppress tumor growth in a murine model. These results indicate that OX40 signals regulate CD8 T cell survival at least in part through maintaining expression of the anti-apoptotic molecule A1, and provide new insight into the mechanism by which OX40 may impact anti-tumor immunity.
Collapse
Affiliation(s)
- Fengyang Lei
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Jianyong Song
- Center of Irradiation, The Third Military Medical University, Chongqing, China
| | - Rizwanul Haque
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Mohammad Haque
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Xiaofang Xiong
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Jianxun Song
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
Bonzheim I, Irmler M, Klier-Richter M, Steinhilber J, Anastasov N, Schäfer S, Adam P, Beckers J, Raffeld M, Fend F, Quintanilla-Martinez L. Identification of C/EBPβ target genes in ALK+ anaplastic large cell lymphoma (ALCL) by gene expression profiling and chromatin immunoprecipitation. PLoS One 2013; 8:e64544. [PMID: 23741337 PMCID: PMC3669320 DOI: 10.1371/journal.pone.0064544] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/16/2013] [Indexed: 12/16/2022] Open
Abstract
C/EBPβ (CCAAT enhancer binding protein) is a transcription factor that plays a crucial role in survival and transformation of ALK+ anaplastic large cell lymphoma (ALCL). The aim of this study was to identify the downstream targets of C/EBPβ responsible for ALK-mediated oncogenesis. C/EBPβ was knocked down in ALK+ ALCL cell lines with a C/EBPβ-shRNA, followed by gene expression profiling (GEP). GEP analysis revealed a reproducible signature of genes that were significantly regulated by C/EBPβ. Classification into biological categories revealed overrepresentation of genes involved in the immune response, apoptosis and cell proliferation. Transcriptional regulation by C/EBPβ was found in 6 of 11 (BCL2A1, G0S2, TRIB1, S100A9, DDX21 and DDIT4) genes investigated by chromatin immunoprecipitation. We demonstrated that BCL2A1, G0S2 and DDX21 play a crucial role in survival and proliferation of ALK+ ALCL cells. DDX21, a gene involved in rRNA biogenesis, was found differentially overexpressed in primary ALK+ ALCL cases. All three candidate genes were validated in primary ALCL cases by either immunohistochemistry or RT-qPCR. In conclusion, we identified and validated several key C/EBPβ-regulated genes with major impact on survival and cell growth in ALK+ ALCL, supporting the central role of C/EBPβ in ALK-mediated oncogenesis.
Collapse
MESH Headings
- CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors
- CCAAT-Enhancer-Binding Protein-beta/genetics
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
- Carcinogenesis/pathology
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Cell Survival/genetics
- Chromatin Immunoprecipitation
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Minor Histocompatibility Antigens
- Promoter Regions, Genetic
- Protein Binding
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- Irina Bonzheim
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
- Institute of Pathology, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, Neuherberg, Germany
| | - Margit Klier-Richter
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
- Institute of Pathology, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, Neuherberg, Germany
| | - Julia Steinhilber
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
| | - Nataša Anastasov
- Institute of Pathology, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, Neuherberg, Germany
| | - Sabine Schäfer
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
| | - Patrick Adam
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Experimental Genetics, Technische Universität München, Freising-Weihenstephan, Germany
| | - Mark Raffeld
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, University Hospital Tübingen and Comprehensive Cancer Center, Eberhard-Karls-University, Tübingen, Germany
- Institute of Pathology, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, Neuherberg, Germany
- * E-mail:
| |
Collapse
|
33
|
Xiong J, Parker BL, Dalheimer SL, Yankee TM. Interleukin-7 supports survival of T-cell receptor-β-expressing CD4(-) CD8(-) double-negative thymocytes. Immunology 2013; 138:382-91. [PMID: 23215679 DOI: 10.1111/imm.12050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 11/30/2012] [Accepted: 12/05/2012] [Indexed: 01/17/2023] Open
Abstract
Among the milestones that occur during T-cell development in the thymus is the expression of T-cell receptor-β (TCR-β) and the formation of the pre-TCR complex. Signals emanating from the pre-TCR trigger survival, proliferation and differentiation of T-cell precursors. Although the pre-TCR is essential for these cell outcomes, other receptors, such as Notch and CXCR4, also contribute. Whether interleukin-7 (IL-7) participates in promoting the survival or proliferation of pre-TCR-expressing cells is controversial. We used in vitro and in vivo models of T-cell development to examine the function of IL-7 in TCR-β-expressing thymocytes. Culturing TCR-β-expressing CD4(-) CD8(-) double-negative thymocytes in an in vitro model of T-cell development revealed that IL-7 reduced the frequency of CD4(+) CD8(+) double-positive thymocytes at the time of harvest. The mechanism for this change in the percentage of double-positive cells was that IL-7 promoted the survival of thymocytes that had not yet differentiated. By preserving the double-negative population, IL-7 reduced the frequency of double-positive thymocytes. Interleukin-7 was not required for proliferation in the in vitro system. To follow this observation, we examined mice lacking CD127 (IL-7Rα). In addition to the known effect of CD127 deficiency on T-cell development before TCR-β expression, CD127 deficiency also impaired the development of TCR-β-expressing double-negative thymocytes. Specifically, we found that Bcl-2 expression and cell cycle progression were reduced in TCR-β-expressing double-negative thymocytes in mice lacking CD127. We conclude that IL-7 continues to function after TCR-β is expressed by promoting the survival of TCR-β-expressing double-negative thymocytes.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
34
|
Renault TT, Chipuk JE. Getting away with murder: how does the BCL-2 family of proteins kill with immunity? Ann N Y Acad Sci 2013; 1285:59-79. [PMID: 23527542 DOI: 10.1111/nyas.12045] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The adult human body produces approximately one million white blood cells every second. However, only a small fraction of the cells will survive because the majority is eliminated through a genetically controlled form of cell death known as apoptosis. This review places into perspective recent studies pertaining to the BCL-2 family of proteins as critical regulators of the development and function of the immune system, with particular attention on B cell and T cell biology. Here we discuss how elegant murine model systems have revealed the major contributions of the BCL-2 family in establishing an effective immune system. Moreover, we highlight some key regulatory pathways that influence the expression, function, and stability of individual BCL-2 family members, and discuss their role in immunity. From lethal mechanisms to more gentle ones, the final portion of the review discusses the nonapoptotic functions of the BCL-2 family and how they pertain to the control of immunity.
Collapse
Affiliation(s)
- Thibaud T Renault
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
35
|
Venigalla RKC, McGuire VA, Clarke R, Patterson-Kane JC, Najafov A, Toth R, McCarthy PC, Simeons F, Stojanovski L, Arthur JSC. PDK1 regulates VDJ recombination, cell-cycle exit and survival during B-cell development. EMBO J 2013; 32:1008-22. [PMID: 23463102 PMCID: PMC3616287 DOI: 10.1038/emboj.2013.40] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 01/30/2013] [Indexed: 01/25/2023] Open
Abstract
Phosphoinositide-dependent kinase-1 (PDK1) controls the activation of a subset of AGC kinases. Using a conditional knockout of PDK1 in haematopoietic cells, we demonstrate that PDK1 is essential for B cell development. B-cell progenitors lacking PDK1 arrested at the transition of pro-B to pre-B cells, due to a cell autonomous defect. Loss of PDK1 decreased the expression of the IgH chain in pro-B cells due to impaired recombination of the IgH distal variable segments, a process coordinated by the transcription factor Pax5. The expression of Pax5 in pre-B cells was decreased in PDK1 knockouts, which correlated with reduced expression of the Pax5 target genes IRF4, IRF8 and Aiolos. As a result, Ccnd3 is upregulated in PDK1 knockout pre-B cells and they have an impaired ability to undergo cell-cycle arrest, a necessary event for Ig light chain rearrangement. Instead, these cells underwent apoptosis that correlated with diminished expression of the pro-survival gene Bcl2A1. Reintroduction of both Pax5 and Bcl2A1 together into PDK1 knockout pro-B cells restored their ability to differentiate in vitro into mature B cells.
Collapse
Affiliation(s)
- Ram K C Venigalla
- MRC Protein Phosphorylation Unit, Sir James Black Centre, College of Life Sciences, University of Dundee, Dundee, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Gerondakis S, Banerjee A, Grigoriadis G, Vasanthakumar A, Gugasyan R, Sidwell T, Grumont RJ. NF-κB subunit specificity in hemopoiesis. Immunol Rev 2012; 246:272-85. [PMID: 22435561 DOI: 10.1111/j.1600-065x.2011.01090.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although the diverse functions served by the nuclear factor-κB (NF-κB) pathway in virtually all cell types are typically employed to deal with stress responses, NF-κB transcription factors also play key roles in the development of hemopoietic cells. This review focuses on how NF-κB transcription factors control various aspects of thymic T-cell and myeloid cell differentiation that include its roles in hemopoietic precursors, conventional αβ T cells, CD4(+) regulatory T cells, natural killer T cells, γδ T cells, macrophages, and dendritic cells.
Collapse
|
37
|
Ottina E, Tischner D, Herold MJ, Villunger A. A1/Bfl-1 in leukocyte development and cell death. Exp Cell Res 2012; 318:1291-303. [PMID: 22342458 PMCID: PMC3405526 DOI: 10.1016/j.yexcr.2012.01.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 12/17/2022]
Abstract
The function of the anti-apoptotic Bcl-2 family member Bcl2a1/Bfl-1/A1 is poorly understood due to the lack of appropriate loss-of-function mouse models and redundant effects with other Bcl-2 pro-survival proteins upon overexpression. Expression analysis of A1 suggests predominant roles in leukocyte development, their survival upon viral or bacterial infection, as well as during allergic reactions. In addition, A1 has been implicated in autoimmunity and the pathology and therapy resistance of hematological as well as solid tumors that may aberrantly express this protein. In this review, we aim to summarize current knowledge on A1 biology, focusing on its role in the immune system and compare it to that of other pro-survival Bcl-2 proteins.
Collapse
Affiliation(s)
- Eleonora Ottina
- Division of Developmental Immunology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Denise Tischner
- Division of Developmental Immunology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Marco J. Herold
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
- Corresponding author at: Division of Developmental Immunology, BIOCENTER, Innsbruck Medical University, A-6020 Innsbruck, Austria. Fax: + 43 512 9003 73960.
| |
Collapse
|
38
|
Chang CF, D'Souza WN, Ch'en IL, Pages G, Pouyssegur J, Hedrick SM. Polar opposites: Erk direction of CD4 T cell subsets. THE JOURNAL OF IMMUNOLOGY 2012; 189:721-31. [PMID: 22675204 DOI: 10.4049/jimmunol.1103015] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Effective immune responses depend upon appropriate T cell differentiation in accord with the nature of an infectious agent, and the contingency of differentiation depends minimally on TCR, coreceptor, and cytokine signals. In this reverse genetic study, we show that the MAPK Erk2 is not essential for T cell proliferation in the presence of optimum costimulation. Instead, it has opposite effects on T-bet and Gata3 expression and, hence, on Th1 and Th2 differentiation. Alternatively, in the presence of TGF-β, the Erk pathway suppresses a large program of gene expression, effectively limiting the differentiation of Foxp3(+) regulatory T cells. In the latter case, the mechanisms involved include suppression of Gata3 and Foxp3, induction of Tbx21, phosphorylation of Smad2,3, and possibly suppression of Socs2, a positive inducer of Stat5 signaling. Consequently, loss of Erk2 severely impeded Th1 differentiation while enhancing the development of Foxp3(+)-induced T regulatory cells. Selected profiles of gene expression under multiple conditions of T cell activation illustrate the opposing consequences of Erk pathway signaling.
Collapse
|
39
|
Targeting antiapoptotic A1/Bfl-1 by in vivo RNAi reveals multiple roles in leukocyte development in mice. Blood 2012; 119:6032-42. [PMID: 22581448 DOI: 10.1182/blood-2011-12-399089] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gene-targeting studies in mice have identified the essential roles of most prosurvival Bcl-2 family members in normal physiology and under conditions of stress. The function of one member, Bcl2a1/Bfl-1/A1, is only poorly understood because of quadruplication of its gene locus in mice, hindering conventional knockout studies. To overcome this problem, we generated mouse models allowing traceable constitutive or reversible ablation of A1 in the hematopoietic system by RNA interference. Knockdown of A1 impaired early stages of T-cell differentiation, B-cell homeostasis, and sensitized transitional as well as follicular B cells to apoptosis induced by ligation of the B-cell receptor. As a consequence, B-cell proliferation in response to mitogens was severely impaired, whereas that of T cells appeared unaffected. Furthermore, depending on the extent of A1 knockdown, granulocytes showed increased spontaneous death in culture or failed to accumulate in significant numbers in vivo. These models highlight the critical role of A1 in leukocyte development and homeostasis, constituting valuable tools for investigating presumed roles of this Bcl-2 family member in immunity, tumorigenesis, and drug resistance.
Collapse
|
40
|
Tischner D, Gaggl I, Peschel I, Kaufmann M, Tuzlak S, Drach M, Thuille N, Villunger A, Jan Wiegers G. Defective cell death signalling along the Bcl-2 regulated apoptosis pathway compromises Treg cell development and limits their functionality in mice. J Autoimmun 2012; 38:59-69. [PMID: 22257939 PMCID: PMC3314992 DOI: 10.1016/j.jaut.2011.12.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 12/16/2011] [Accepted: 12/19/2011] [Indexed: 01/07/2023]
Abstract
The Bcl-2 regulated apoptosis pathway is critical for the elimination of autoreactive lymphocytes, thereby precluding autoimmunity. T cells escaping this process can be kept in check by regulatory T (Treg) cells expressing the transcription and lineage commitment factor Foxp3. Despite the well-established role of Bcl-2 family proteins in shaping the immune system and their frequent deregulation in autoimmune pathologies, it is poorly understood how these proteins affect Treg cell development and function. Here we compared the relative expression of a panel of 40 apoptosis-associated genes in Treg vs. conventional CD4+ T cells. Physiological significance of key-changes was validated using gene-modified mice lacking or overexpressing pro- or anti-apoptotic Bcl-2 family members. We define a key role for the Bim/Bcl-2 axis in Treg cell development, homeostasis and function but exclude a role for apoptosis induction in responder T cells as relevant suppression mechanism. Notably, only lack of the pro-apoptotic BH3-only protein Bim or Bcl-2 overexpression led to accumulation of Treg cells while loss of pro-apoptotic Bad, Bmf, Puma or Noxa had no effect. Remarkably, apoptosis resistant Treg cells showed reduced suppressive capacity in a model of T cell-driven colitis, posing a caveat for the use of such long-lived cells in possible therapeutic settings.
Collapse
Affiliation(s)
- Denise Tischner
- Biocenter, Division of Developmental Immunology, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
B-cell lymphoma 2 (BCL2) proteins are important cell death regulators, whose main function is to control the release of cytochrome c from mitochondria in the intrinsic apoptotic pathway. They comprise both pro- and anti-apoptotic proteins, which interact in various ways to induce or prevent pore formation in the outer mitochondrial membrane. Due to their central function in the apoptotic machinery, BCL2 proteins are often deregulated in cancer. To this end, many anti-apoptotic BCL2 proteins have been identified as important cellular oncogenes and attractive targets for anti-cancer therapy. In this review, the existing knowledge on B-cell lymphoma 2-related protein A1 (BCL2A1)/Bcl-2-related gene expressed in fetal liver (Bfl-1), one of the less extensively studied anti-apoptotic BCL2 proteins, is summarized. BCL2A1 is a highly regulated nuclear factor κB (NF-κB) target gene that exerts important pro-survival functions. In a physiological context, BCL2A1 is mainly expressed in the hematopoietic system, where it facilitates survival of selected leukocytes subsets and inflammation. However, BCL2A1 is overexpressed in a variety of cancer cells, including hematological malignancies and solid tumors, and may contribute to tumor progression. Therefore, the development of small molecule inhibitors of BCL2A1 may be a promising approach mainly to sensitize tumor cells for apoptosis and thus improve the efficiency of anti-cancer therapy.
Collapse
Affiliation(s)
- M Vogler
- MRC Toxicology Unit, University of Leicester, Leicester, UK.
| |
Collapse
|
42
|
The NF-κB1 transcription factor prevents the intrathymic development of CD8 T cells with memory properties. EMBO J 2011; 31:692-706. [PMID: 22124325 DOI: 10.1038/emboj.2011.435] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 11/04/2011] [Indexed: 12/29/2022] Open
Abstract
The role of specific members of the NF-κB family of transcription factors in CD8 T-cell selection and development is largely unknown. Here, we show that mice lacking NF-κB1 develop a unique population of conventional CD8 single-positive (SP) thymocytes with memory T cell-like properties that populate peripheral immune organs. Development of this memory-like population is not due to PLZF(+) thymocytes and instead coincides with changes in CD8 T-cell selection. These include a reduction in the efficiency of negative selection and a dependence on MHC class Ia or Ib expressed by haematopoietic cells. These findings indicate that NF-κB1 regulates multiple events in the thymus that collectively inhibit the excess development of CD8(+) thymocytes with memory cell characteristics.
Collapse
|
43
|
Pharmacological blockade of Bcl-2, Bcl-x(L) and Bcl-w by the BH3 mimetic ABT-737 has only minor impact on tumour development in p53-deficient mice. Cell Death Differ 2011; 19:623-32. [PMID: 21997189 DOI: 10.1038/cdd.2011.133] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The tumour suppressor p53 transcriptionally regulates a range of target genes that control cell growth and survival. Mutations of p53 have been implicated in the development of approximately 50% of human cancers, including those instigated by exposure to mutagens. Although numerically rare, cancers can arise as a consequence of inherited mutations, such as in the Li-Fraumeni syndrome, which is caused by mutation of one p53 allele. Gene-targeted mice deficient for p53 have been generated to study this familial cancer syndrome. On a C57BL/6 background, p53-deficient mice develop primarily thymic lymphoma and more rarely sarcoma. Evasion of apoptosis is considered to be essential for neoplastic transformation. As proteins of the Bcl-2 family are the critical regulators of apoptosis, we investigated the role of the pro-survival members Bcl-2, Bcl-x(L) and Bcl-w in cancer development in p53(+/-) and p53(-/-) mice by testing whether ABT-737, a pharmacological inhibitor of these proteins, could prevent or delay tumourigenesis. Our studies showed that ABT-737 prophylaxis only caused a minor delay and reduction in γ-radiation-induced thymic lymphoma development in p53(-/-) mice, but this was accompanied by a concomitant increase in sarcoma. These data show that, collectively, Bcl-2, Bcl-x(L) and Bcl-w have only minor roles in thymic lymphoma development elicited by defects in p53, and this may indicate that Mcl-1 and/or A1 may feature more prominently in this process.
Collapse
|
44
|
Villaudy J, Wencker M, Gadot N, Gillet NA, Scoazec JY, Gazzolo L, Manz MG, Bangham CRM, Dodon MD. HTLV-1 propels thymic human T cell development in "human immune system" Rag2⁻/⁻ gamma c⁻/⁻ mice. PLoS Pathog 2011; 7:e1002231. [PMID: 21909275 PMCID: PMC3164654 DOI: 10.1371/journal.ppat.1002231] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 07/07/2011] [Indexed: 01/06/2023] Open
Abstract
Alteration of early haematopoietic development is thought to be responsible for the onset of immature leukemias and lymphomas. We have previously demonstrated that Tax(HTLV-1) interferes with ß-selection, an important checkpoint of early thymopoiesis, indicating that human T-cell leukemia virus type 1 (HTLV-1) infection has the potential to perturb thymic human αβ T-cell development. To verify that inference and to clarify the impact of HTLV-1 infection on human T-cell development, we investigated the in vivo effects of HTLV-1 infection in a "Human Immune System" (HIS) Rag2⁻/⁻γ(c)⁻/⁻ mouse model. These mice were infected with HTLV-1, at a time when the three main subpopulations of human thymocytes have been detected. In all but two inoculated mice, the HTLV-1 provirus was found integrated in thymocytes; the proviral load increased with the length of the infection period. In the HTLV-1-infected mice we observed alterations in human T-cell development, the extent of which correlated with the proviral load. Thus, in the thymus of HTLV-1-infected HIS Rag2⁻/⁻γc⁻/⁻ mice, mature single-positive (SP) CD4⁺ and CD8⁺ cells were most numerous, at the expense of immature and double-positive (DP) thymocytes. These SP cells also accumulated in the spleen. Human lymphocytes from thymus and spleen were activated, as shown by the expression of CD25: this activation was correlated with the presence of tax mRNA and with increased expression of NF-kB dependent genes such as bfl-1, an anti-apoptotic gene, in thymocytes. Finally, hepato-splenomegaly, lymphadenopathy and lymphoma/thymoma, in which Tax was detected, were observed in HTLV-1-infected mice, several months after HTLV-1 infection. These results demonstrate the potential of the HIS Rag2⁻/⁻γ(c)⁻/⁻ animal model to elucidate the initial steps of the leukemogenic process induced by HTLV-1.
Collapse
Affiliation(s)
- Julien Villaudy
- Virologie Humaine, INSERM-U758, Lyon, France
- Ecole Normale Supérieure, Lyon, France
- UMS3444 BioSciences Lyon-Gerland, Lyon, France
| | - Mélanie Wencker
- Virologie Humaine, INSERM-U758, Lyon, France
- Ecole Normale Supérieure, Lyon, France
- UMS3444 BioSciences Lyon-Gerland, Lyon, France
- Cancer Research UK, London Research Institute and King's College, London, United Kingdom
| | - Nicolas Gadot
- Anipath, UFR Médecine Lyon-RTH Laennec, Lyon, France
| | - Nicolas A. Gillet
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London, United Kingdom
- Molecular and Cellular Epigenetics, Interdisciplinary Cluster for Applied Genoproteomics (GIGA) of University of Liège (ULg), Liège, Belgium
| | | | - Louis Gazzolo
- Virologie Humaine, INSERM-U758, Lyon, France
- Ecole Normale Supérieure, Lyon, France
- UMS3444 BioSciences Lyon-Gerland, Lyon, France
| | - Markus G. Manz
- Institute for Research in Biomedicine (IRB), Bellinzona, Switzerland
- University and University Hospital Zürich, Division of Hematology, Zürich, Switzerland
| | - Charles R. M. Bangham
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London, United Kingdom
| | - Madeleine Duc Dodon
- Virologie Humaine, INSERM-U758, Lyon, France
- Ecole Normale Supérieure, Lyon, France
- UMS3444 BioSciences Lyon-Gerland, Lyon, France
- * E-mail:
| |
Collapse
|
45
|
Abstract
During the development and normal function of T lymphocytes, the cells are subject to several checkpoints at which they must "decide" to live or die. At these critical times and during homeostasis, the molecules that regulate the classical apoptotic pathways and survival pathways such as autophagy have critical roles in controlling this decision. Our laboratory has focused on the roles of apoptotic and autophagic proteins in T lymphocyte development and function. Using genetic models in mice and in vitro analyses of T cell functions, we have outlined critical roles for the Bcl-2 family (regulators of the intrinsic pathway of apoptosis), c-FLIP (an anti-apoptotic protein in the extrinsic pathway of apoptosis), and autophagy in T lymphocytes.
Collapse
Affiliation(s)
- Alexis Dunkle
- Department of Immunology, Duke University Medical Center, Box 3010, Durham, NC 27710, USA
| | | |
Collapse
|
46
|
Castiello L, Sabatino M, Jin P, Clayberger C, Marincola FM, Krensky AM, Stroncek DF. Monocyte-derived DC maturation strategies and related pathways: a transcriptional view. Cancer Immunol Immunother 2011; 60:457-66. [PMID: 21258790 PMCID: PMC3086891 DOI: 10.1007/s00262-010-0954-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 11/30/2010] [Indexed: 12/17/2022]
Abstract
Ex vivo production of highly stimulator mature dendritic cells (DCs) for cellular therapy has been used to treat different pathological conditions with the aim of inducing a specific immune response. In the last decade, several protocols have been developed to mature monocyte-derived DCs: each one has led to the generation of DCs showing different phenotypes and stimulatory abilities, but it is not yet known which one is the best for inducing effective immune responses. We grouped several different maturation protocols according to the downstream pathways they activated and reviewed the shared features at a transcriptomic level to reveal the potential of DCs matured by each protocol to develop Th-polarized immune responses.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Marianna Sabatino
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ping Jin
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Carol Clayberger
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Francesco M. Marincola
- Infectious Disease and Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, and Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD 20892 USA
| | - Alan M. Krensky
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - David F. Stroncek
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
47
|
Abstract
The link between evasion of apoptosis and the development of cellular hyperplasia and ultimately cancer is implicitly clear if one considers how many cells are produced each day and, hence, how many cells must die to make room for the new ones (reviewed in Raff, 1996). Furthermore, cells are frequently experiencing noxious stimuli that can cause lesions in their DNA and faults in DNA replication can occur during cellular proliferation. Such DNA damage needs to be repaired efficiently or cells with irreparable damage must be killed to prevent subsequent division of aberrant cells that may fuel tumorigenesis (reviewed in Weinberg, 2007). The detection of genetic lesions in human cancers that activate prosurvival genes or disable proapoptotic genes have provided the first evidence that defects in programmed cell death can cause cancer (Tagawa et al., 2005; Tsujimoto et al., 1984; Vaux, Cory, and Adams, 1988) and this concept was proven by studies with genetically modified mice (Egle et al., 2004b; Strasser et al., 1990a). It is therefore now widely accepted that evasion of apoptosis is a requirement for both neoplastic transformation and sustained growth of cancer cells (reviewed in Cory and Adams, 2002; Hanahan and Weinberg, 2000; Weinberg, 2007). Importantly, apoptosis is also a major contributor to anticancer therapy-induced killing of tumor cells (reviewed in Cory and Adams, 2002; Cragg et al., 2009). Consequently, a detailed understanding of apoptotic cell death will help to better comprehend the complexities of tumorigenesis and should assist with the development of improved targeted therapies for cancer based on the direct activation of the apoptotic machinery (reviewed in Lessene, Czabotar, and Colman, 2008).
Collapse
Affiliation(s)
- Gemma Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
48
|
Xiong J, Armato MA, Yankee TM. Immature single-positive CD8+ thymocytes represent the transition from Notch-dependent to Notch-independent T-cell development. Int Immunol 2010; 23:55-64. [PMID: 21148236 DOI: 10.1093/intimm/dxq457] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Early in T-cell development, cells proceed through stages that are critically dependent on signaling through the Notch receptor. As cells mature, thymocytes transition from being Notch dependent to being Notch independent, but the stage of development during which this transition occurs is unknown. We used an in vitro differentiation system in which thymocytes can be cultured in the presence or absence of a Notch ligand to identify the stage of development in which thymocytes transition from being Notch responsive to Notch non-responsive. We identified the immature single-positive (ISP) CD8(+) stage of T-cell development as being this transition point. ISP thymocytes were responsive to Notch, but ISP cells responded to Notch ligation in a manner that was distinct from the response by double-negative (DN) thymocytes. Fewer ISP thymocytes proliferated and more ISP cells died in culture than DN thymocytes. Further, fewer double-positive (DP) thymocytes generated by culturing ISP thymocytes were in the S, G2 or M phase of the cell cycle as compared with DP thymocytes derived from DN thymocytes. These data indicate that the DP population created varied depending on the input population. In summary, the data presented here indicate that ISP thymocytes responded to Notch differently than DN thymocytes and ISP thymocytes represent the transition stage from Notch-dependent survival and proliferation to Notch-independent survival and proliferation.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, 3025 Wahl Hall West and Mailstop 3029, Kansas City, KS 66160, USA
| | | | | |
Collapse
|
49
|
Hernandez JB, Newton RH, Walsh CM. Life and death in the thymus--cell death signaling during T cell development. Curr Opin Cell Biol 2010; 22:865-71. [PMID: 20810263 PMCID: PMC2993787 DOI: 10.1016/j.ceb.2010.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 07/31/2010] [Accepted: 08/05/2010] [Indexed: 12/27/2022]
Abstract
The thymus is an organ vital to proper T cell development, and the regulation of cell survival and death contributes significantly to its efficient function. Vital to many of the developmental processes that occur in the thymus, control over cell survival and death is orchestrated by several signaling processes. In this review, we focus on the regulation of death in early thymocytes known as CD4/CD8 double negative cells, including the roles of interleukin-7 and Bcl-2 family members in this developmental stage. We next consider the survival and death of later thymocytes that express both CD4 and CD8, the 'double-positive' thymocytes. These findings are discussed within the context of recent studies demonstrating the existence of caspase-independent cell death pathways.
Collapse
Affiliation(s)
| | | | - Craig M. Walsh
- The Institute for Immunology and the Department of Molecular Biology and Biochemistry, University of California, Irvine
| |
Collapse
|
50
|
Deng Y, Johnson DR, Guan X, Ang CY, Ai J, Perkins EJ. In vitro gene regulatory networks predict in vivo function of liver. BMC SYSTEMS BIOLOGY 2010; 4:153. [PMID: 21073692 PMCID: PMC2998496 DOI: 10.1186/1752-0509-4-153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 11/12/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Evolution of toxicity testing is predicated upon using in vitro cell based systems to rapidly screen and predict how a chemical might cause toxicity to an organ in vivo. However, the degree to which we can extend in vitro results to in vivo activity and possible mechanisms of action remains to be fully addressed. RESULTS Here we use the nitroaromatic 2,4,6-trinitrotoluene (TNT) as a model chemical to compare and determine how we might extrapolate from in vitro data to in vivo effects. We found 341 transcripts differentially expressed in common among in vitro and in vivo assays in response to TNT. The major functional term corresponding to these transcripts was cell cycle. Similarly modulated common pathways were identified between in vitro and in vivo. Furthermore, we uncovered the conserved common transcriptional gene regulatory networks between in vitro and in vivo cellular liver systems that responded to TNT exposure, which mainly contain 2 subnetwork modules: PTTG1 and PIR centered networks. Interestingly, all 7 genes in the PTTG1 module were involved in cell cycle and downregulated by TNT both in vitro and in vivo. CONCLUSIONS The results of our investigation of TNT effects on gene expression in liver suggest that gene regulatory networks obtained from an in vitro system can predict in vivo function and mechanisms. Inhibiting PTTG1 and its targeted cell cycle related genes could be key mechanism for TNT induced liver toxicity.
Collapse
Affiliation(s)
- Youping Deng
- Rush University Medical Center, Chicago, IL 60612, USA.
| | | | | | | | | | | |
Collapse
|