1
|
Kübelbeck T, Wichmann NO, Raj T, Raj C, Ohnmacht C, Hövelmeyer N, Kramer D, Heissmeyer V. Regulation and Function of the Atypical IκBs-Bcl-3, IκB NS, and IκBζ-in Lymphocytes and Autoimmunity. Eur J Immunol 2025; 55:e202451273. [PMID: 40359334 PMCID: PMC12074568 DOI: 10.1002/eji.202451273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
Signaling pathways involving NF-κB transcription factors have essential roles in inflammation, immunity, cell proliferation, differentiation, and survival. Classical IκB proteins, such as IκBα and IκBβ, bind to NF-κB via ankyrin repeats to sequester NF-κB in the cytoplasm and thus suppress NF-κB activity. Unlike these constitutively expressed classical IκBs, the expression of the atypical IκBs Bcl-3, IκBNS, and IκBζ is induced in immune cells after recognition of antigens, pathogen-associated molecular patterns (PAMPs) or cytokines, upon which they localize to the nucleus and form complexes with transcription factors and regulators on the DNA. Atypical, nuclear IκBs have been proposed to modulate NF-κB activity in a context-dependent manner as they can either inhibit or increase gene expression of a subset of NF-κB target genes. This complexity may be related to the molecular function of atypical IκBs, which bind to different transcription factor complexes and form a bridge to different cofactors or epigenetic modifiers. Recent research has identified novel target genes of atypical IκBs that include chemokines, cytokines, and master regulators of lymphocyte differentiation, underscoring prominent roles in adaptive immune and autoimmune responses. Here, we summarize our current understanding of atypical IκBs in lymphocytes with a focus on their emerging role in autoimmunity.
Collapse
Affiliation(s)
- Tanja Kübelbeck
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg‐University of MainzMainzGermany
| | - Nina Olivera Wichmann
- Center of Allergy and Environment (ZAUM)Technical University and Helmholtz Zentrum MünchenMunichGermany
| | - Timsse Raj
- Institute for Immunology, Biomedical Center (BMC), Faculty of MedicineLudwig‐Maximilians‐Universität in MunichPlanegg‐MartinsriedGermany
| | - Cynthia Raj
- Institute for Molecular Medicine MainzUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM)Technical University and Helmholtz Zentrum MünchenMunichGermany
| | - Nadine Hövelmeyer
- Institute for Molecular Medicine MainzUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
- Research Centre for Immunotherapy (FZI)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Daniela Kramer
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg‐University of MainzMainzGermany
- Research Centre for Immunotherapy (FZI)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Vigo Heissmeyer
- Institute for Immunology, Biomedical Center (BMC), Faculty of MedicineLudwig‐Maximilians‐Universität in MunichPlanegg‐MartinsriedGermany
- Research Unit Molecular Immune RegulationMolecular Targets and Therapeutics CenterHelmholtz Zentrum MünchenMunichGermany
| |
Collapse
|
2
|
Wu J, Wang N. Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review). Mol Med Rep 2024; 30:221. [PMID: 39364736 PMCID: PMC11462401 DOI: 10.3892/mmr.2024.13344] [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: 02/17/2023] [Accepted: 11/11/2023] [Indexed: 10/05/2024] Open
Abstract
Glioblastoma (GBM) is the most common central nervous system malignancy in adults. GBM may be classified as grade IV diffuse astrocytoma according to the 2021 World Health Organization revised classification of central nervous system tumors, which means it is the most aggressive, invasive, undifferentiated type of tumor. Immune checkpoint blockade (ICB), particularly anti‑programmed cell death protein‑1 (PD‑1)/PD‑1 ligand‑1 immunotherapy, has been confirmed to be successful across several tumor types. However, in GBM, this treatment is still uncommon and the efficacy is unpredictable, and <10% of patients show long‑term responses. Recently, numerous studies have been conducted to explore what factors may indicate or affect the ICB response rate in GBM, including molecular alterations, immune expression signatures and immune infiltration. The present review aimed to summarize the current progress to improve the understanding of immunotherapy for GBM.
Collapse
Affiliation(s)
- Jianheng Wu
- Department of Neurosurgery, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Nannan Wang
- Department of Gastroenterology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| |
Collapse
|
3
|
Köhler A, Geiselhöringer AL, Kolland D, Kreft L, Wichmann N, Hils M, Pasztoi M, Zurkowski E, Vogt J, Kübelbeck T, Biedermann T, Schmitz I, Hansen W, Kramer D, Gaida MM, Schmidt-Weber CB, Hoevelmeyer N, Ohnmacht C. The atypical IκB family member Bcl3 determines differentiation and fate of intestinal RORγt + regulatory T-cell subsets. Mucosal Immunol 2024; 17:673-691. [PMID: 38663461 DOI: 10.1016/j.mucimm.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/07/2024] [Accepted: 04/16/2024] [Indexed: 05/09/2024]
Abstract
Peripherally-induced regulatory T cells (pTregs) expressing the retinoic acid receptor-related orphan-receptor gamma t (RORγt) are indispensable for intestinal immune homeostasis. Nuclear factor kappa family members regulate the differentiation of thymic Tregs and promote their survival in the periphery. However, the Treg intrinsic molecular mechanisms controlling the size of the pTregs in the intestine and associated lymphoid organs remain unclear. Here, we provide direct evidence that B-cell lymphoma 3 (Bcl3) limits the development of pTregs in a T cell-intrinsic manner. Moreover, the absence of Bcl3 allowed for the formation of an unusual intestinal Treg population co-expressing the transcription factors Helios and RORγt. The expanded RORγt+ Treg populations in the absence of Bcl3 displayed an activated phenotype and secreted high levels of the anti-inflammatory cytokines interleukin (IL)-10 and transforming growth factor beta. They were fully capable of suppressing effector T cells in a transfer colitis model despite an intrinsic bias to trans-differentiate toward T helper 17-like cells. Finally, we provide a Bcl3-dependent gene signature in pTregs including altered responsiveness to the cytokines IL-2, IL-6, and tumor necrosis factor alpha. Our results demonstrate that Bcl3 acts as a molecular switch to limit the expansion of different intestinal Treg subsets and may thus serve as a novel therapeutic target for inflammatory bowel disease by restoring intestinal immune tolerance.
Collapse
Affiliation(s)
- Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany
| | - Anna-Lena Geiselhöringer
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany
| | - Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany
| | - Luisa Kreft
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany
| | - Nina Wichmann
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany
| | - Miriam Hils
- Department of Dermatology and Allergy Biederstein, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Maria Pasztoi
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany
| | - Elena Zurkowski
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Johannes Vogt
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Tanja Kübelbeck
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Ingo Schmitz
- Department of Molecular Immunology, Ruhr University Bochum, Bochum, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Daniela Kramer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Matthias M Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, Mainz, Germany; TRON, Translational Oncology at the University Medical Center, JGU-Mainz, Mainz, Germany; Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany; Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Nadine Hoevelmeyer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Germany.
| |
Collapse
|
4
|
Seaton G, Smith H, Brancale A, Westwell AD, Clarkson R. Multifaceted roles for BCL3 in cancer: a proto-oncogene comes of age. Mol Cancer 2024; 23:7. [PMID: 38195591 PMCID: PMC10775530 DOI: 10.1186/s12943-023-01922-8] [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: 10/27/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Abstract
In the early 1990's a group of unrelated genes were identified from the sites of recurring translocations in B-cell lymphomas. Despite sharing the nomenclature 'Bcl', and an association with blood-borne cancer, these genes have unrelated functions. Of these genes, BCL2 is best known as a key cancer target involved in the regulation of caspases and other cell viability mechanisms. BCL3 on the other hand was originally identified as a non-canonical regulator of NF-kB transcription factor pathways - a signaling mechanism associated with important cell outcomes including many of the hallmarks of cancer. Most of the early investigations into BCL3 function have since focused on its role in NF-kB mediated cell proliferation, inflammation/immunity and cancer. However, recent evidence is coming to light that this protein directly interacts with and modulates a number of other signaling pathways including DNA damage repair, WNT/β-catenin, AKT, TGFβ/SMAD3 and STAT3 - all of which have key roles in cancer development, metastatic progression and treatment of solid tumours. Here we review the direct evidence demonstrating BCL3's central role in a transcriptional network of signaling pathways that modulate cancer biology and treatment response in a range of solid tumour types and propose common mechanisms of action of BCL3 which may be exploited in the future to target its oncogenic effects for patient benefit.
Collapse
Affiliation(s)
- Gillian Seaton
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Hannah Smith
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Andrea Brancale
- UCT Prague, Technická 5, 166 28, 6 - Dejvice, IČO: 60461337, Prague, Czech Republic
| | - Andrew D Westwell
- Cardiff University School of Pharmacy and Pharmaceutical Sciences, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Richard Clarkson
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
5
|
Merino-Vico A, van Hamburg JP, Tuijnenburg P, Frazzei G, Al-Soudi A, Bonasia CG, Helder B, Rutgers A, Abdulahad WH, Stegeman CA, Sanders JS, Bergamaschi L, Lyons PA, Bijma T, van Keep L, Wesenhagen K, Jongejan A, Olsson H, de Vries N, Kuijpers TW, Heeringa P, Tas SW. Targeting NF-κB signaling in B cells as a potential new treatment modality for ANCA-associated vasculitis. J Autoimmun 2024; 142:103133. [PMID: 37931331 DOI: 10.1016/j.jaut.2023.103133] [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: 07/18/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023]
Abstract
B lineage cells are critically involved in ANCA-associated vasculitis (AAV), evidenced by alterations in circulating B cell subsets and beneficial clinical effects of rituximab (anti-CD20) therapy. This treatment renders a long-term, peripheral B cell depletion, but allows for the survival of long-lived plasma cells. Therefore, there is an unmet need for more reversible and full B lineage cell targeting approaches. To find potential novel therapeutic targets, RNA sequencing of CD27+ memory B cells of patients with active AAV was performed, revealing an upregulated NF-κB-associated gene signature. NF-κB signaling pathways act downstream of various B cell surface receptors, including the BCR, CD40, BAFFR and TLRs, and are essential for B cell responses. Here we demonstrate that novel pharmacological inhibitors of NF-κB inducing kinase (NIK, non-canonical NF-κB signaling) and inhibitor-of-κB-kinase-β (IKKβ, canonical NF-κB signaling) can effectively inhibit NF-κB signaling in B cells, whereas T cell responses were largely unaffected. Moreover, both inhibitors significantly reduced B cell proliferation, differentiation and production of antibodies, including proteinase-3 (PR3) autoantibodies, in B lineage cells of AAV patients. These findings indicate that targeting NF-κB, particularly NIK, may be an effective, novel B lineage cell targeted therapy for AAV and other autoimmune diseases with prominent B cell involvement.
Collapse
Affiliation(s)
- Ana Merino-Vico
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul Tuijnenburg
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Giulia Frazzei
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Aram Al-Soudi
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Carlo G Bonasia
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Boy Helder
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Abraham Rutgers
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Wayel H Abdulahad
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Coen A Stegeman
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Jan-Stephan Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Laura Bergamaschi
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffre Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Paul A Lyons
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffre Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Theo Bijma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Laura van Keep
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Kirsten Wesenhagen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science, Bioinformatics Laboratory, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Henric Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Niek de Vries
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 EA11, 9713, GZ, Groningen, the Netherlands
| | - Sander W Tas
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Wang H, Zhang G, Dong L, Chen L, Liang L, Ge L, Gai D, Shen X. Identification and study of cuproptosis-related genes in prognostic model of multiple myeloma. Hematology 2023; 28:2249217. [PMID: 37610069 DOI: 10.1080/16078454.2023.2249217] [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: 03/31/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a highly heterogeneous disease. Cuproptosis is a novel mode of death that is closely associated with several diseases, such as hepatocellular carcinoma. However, its role in MM is unknown. METHODS MM transcriptomic and clinical data were obtained from UCSC Xena and gene expression omnibus (GEO) databases. Following MM samples were divided into different subtypes based on the cuproptosis genes, the differentially expressed genes (DEGs) among different subtypes, namely, candidate cuproptosis related genes were analyzed by univariate Cox and least absolute shrinkage and selection operator (LASSO) regression to construct a cuproptosis-related risk model. After the independent prognostic analysis was performed, a nomogram was constructed. Finally, Functional enrichment analysis and immune infiltration analysis were performed in the high- and low-risk groups, potential therapeutic agents were then predicted. RESULTS The 784 MM samples in UCSC Xena cohorts were divided into three different subtypes, and 4 out of 346 candidate cuproptosis related genes, namely CDKN2A, BCL3, KCNA3 and TTC14 were used to construct a risk model. Risk score was considered a reliable independent prognostic factor for MM patients. It was investigated that the pathway of cell cycle was significantly enriched in the high-risk group. In addition, immune score, ESTIMATE score and cytolytic activity were significantly different between different risk groups, as well as 13 immune cells such as memory B cells. Nine drugs were predicted in our study. CONCLUSION A cuproptosis-related prognostic model was constructed, which may have a potential guiding role in the treatment of MM.
Collapse
Affiliation(s)
- Haili Wang
- Shanxi Medical University, Taiyuan, People's Republic of China
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Guoxiang Zhang
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Lu Dong
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Lu Chen
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Li Liang
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Li Ge
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Dongzheng Gai
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| | - Xuliang Shen
- Shanxi Medical University, Taiyuan, People's Republic of China
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, People's Republic of China
| |
Collapse
|
7
|
Pecharromán I, Solé L, Álvarez‐Villanueva D, Lobo‐Jarne T, Alonso‐Marañón J, Bertran J, Guillén Y, Montoto Á, Martínez‐Iniesta M, García‐Hernández V, Giménez G, Salazar R, Santos C, Garrido M, Borràs E, Sabidó E, Bonfill‐Teixidor E, Iurlaro R, Seoane J, Villanueva A, Iglesias M, Bigas A, Espinosa L. IκB kinase-α coordinates BRD4 and JAK/STAT signaling to subvert DNA damage-based anticancer therapy. EMBO J 2023; 42:e114719. [PMID: 37737566 PMCID: PMC10620764 DOI: 10.15252/embj.2023114719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/28/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023] Open
Abstract
Activation of the IκB kinase (IKK) complex has recurrently been linked to colorectal cancer (CRC) initiation and progression. However, identification of downstream effectors other than NF-κB has remained elusive. Here, analysis of IKK-dependent substrates in CRC cells after UV treatment revealed that phosphorylation of BRD4 by IKK-α is required for its chromatin-binding at target genes upon DNA damage. Moreover, IKK-α induces the NF-κB-dependent transcription of the cytokine LIF, leading to STAT3 activation, association with BRD4 and recruitment to specific target genes. IKK-α abrogation results in defective BRD4 and STAT3 functions and consequently irreparable DNA damage and apoptotic cell death upon different stimuli. Simultaneous inhibition of BRAF-dependent IKK-α activity, BRD4, and the JAK/STAT pathway enhanced the therapeutic potential of 5-fluorouracil combined with irinotecan in CRC cells and is curative in a chemotherapy-resistant xenograft model. Finally, coordinated expression of LIF and IKK-α is a poor prognosis marker for CRC patients. Our data uncover a functional link between IKK-α, BRD4, and JAK/STAT signaling with clinical relevance.
Collapse
Affiliation(s)
- Irene Pecharromán
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Laura Solé
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Daniel Álvarez‐Villanueva
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet del LlobregatBarcelonaSpain
| | - Teresa Lobo‐Jarne
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Josune Alonso‐Marañón
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Joan Bertran
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
- Faculty of Science and TechnologyUniversity of Vic – Central University of CataloniaVicSpain
| | - Yolanda Guillén
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Ángela Montoto
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - María Martínez‐Iniesta
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet del LlobregatBarcelonaSpain
| | - Violeta García‐Hernández
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Gemma Giménez
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Ramon Salazar
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)‐CIBERONCL'Hospitalet de LlobregatBarcelonaSpain
| | - Cristina Santos
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)‐CIBERONCL'Hospitalet de LlobregatBarcelonaSpain
| | - Marta Garrido
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Eva Borràs
- Proteomics Unit, Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Proteomics UnitUniversitat Pompeu FabraBarcelonaSpain
| | - Eduard Sabidó
- Proteomics Unit, Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Proteomics UnitUniversitat Pompeu FabraBarcelonaSpain
| | - Ester Bonfill‐Teixidor
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONCVall d'Hebron University Hospital, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Raffaella Iurlaro
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONCVall d'Hebron University Hospital, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONCVall d'Hebron University Hospital, Universitat Autònoma de BarcelonaBarcelonaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet del LlobregatBarcelonaSpain
- Xenopat S.L., Parc Cientific de Barcelona (PCB)BarcelonaSpain
| | - Mar Iglesias
- Department of Pathology, Institut Mar d'Investigacions Mèdiques, CIBERONCUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Anna Bigas
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
- Josep Carreras Leukemia Research InstituteBadalonaSpain
| | - Lluís Espinosa
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| |
Collapse
|
8
|
Inagaki T, Wang KH, Kumar A, Izumiya C, Miura H, Komaki S, Davis RR, Tepper CG, Katano H, Shimoda M, Izumiya Y. KSHV vIL-6 enhances inflammatory responses by epigenetic reprogramming. PLoS Pathog 2023; 19:e1011771. [PMID: 37934757 PMCID: PMC10656005 DOI: 10.1371/journal.ppat.1011771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/17/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023] Open
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) inflammatory cytokine syndrome (KICS) is a newly described chronic inflammatory disease condition caused by KSHV infection and is characterized by high KSHV viral load and sustained elevations of serum KSHV-encoded IL-6 (vIL-6) and human IL-6 (hIL-6). KICS has significant immortality and greater risks of other complications, including malignancies. Although prolonged inflammatory vIL-6 exposure by persistent KSHV infection is expected to have key roles in subsequent disease development, the biological effects of prolonged vIL-6 exposure remain elusive. Using thiol(SH)-linked alkylation for the metabolic (SLAM) sequencing and Cleavage Under Target & Release Using Nuclease analysis (CUT&RUN), we studied the effect of prolonged vIL-6 exposure in chromatin landscape and resulting cytokine production. The studies showed that prolonged vIL-6 exposure increased Bromodomain containing 4 (BRD4) and histone H3 lysine 27 acetylation co-occupancies on chromatin, and the recruitment sites were frequently co-localized with poised RNA polymerase II with associated enzymes. Increased BRD4 recruitment on promoters was associated with increased and prolonged NF-κB p65 binding after the lipopolysaccharide stimulation. The p65 binding resulted in quicker and sustained transcription bursts from the promoters; this mechanism increased total amounts of hIL-6 and IL-10 in tissue culture. Pretreatment with the BRD4 inhibitors, OTX015 and MZ1, eliminated the enhanced inflammatory cytokine production. These findings suggest that persistent vIL-6 exposure may establish a chromatin landscape favorable for the reactivation of inflammatory responses in monocytes. This epigenetic memory may explain the greater risk of chronic inflammatory disease development in KSHV-infected individuals.
Collapse
Affiliation(s)
- Tomoki Inagaki
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Kang-Hsin Wang
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Ashish Kumar
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Chie Izumiya
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Hiroki Miura
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Somayeh Komaki
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Ryan R. Davis
- Department of Pathology and Laboratory Medicine, School of Medicine, UC Davis, Sacramento, California, United States of America
| | - Clifford G. Tepper
- Department of Biochemistry and Molecular Medicine, School of Medicine, UC Davis, Sacramento, California, United States of America
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Michiko Shimoda
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, School of Medicine, UC Davis, Sacramento, California, United States of America
| |
Collapse
|
9
|
Brandstoetter T, Schmoellerl J, Grausenburger R, Kollmann S, Doma E, Huuhtanen J, Klampfl T, Eder T, Grebien F, Hoermann G, Zuber J, Mustjoki S, Maurer B, Sexl V. SBNO2 is a critical mediator of STAT3-driven hematological malignancies. Blood 2023; 141:1831-1845. [PMID: 36630607 PMCID: PMC10646773 DOI: 10.1182/blood.2022018494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/12/2022] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Gain-of-function mutations in the signal transducer and activator of transcription 3 (STAT3) gene are recurrently identified in patients with large granular lymphocytic leukemia (LGLL) and in some cases of natural killer (NK)/T-cell and adult T-cell leukemia/lymphoma. To understand the consequences and molecular mechanisms contributing to disease development and oncogenic transformation, we developed murine hematopoietic stem and progenitor cell models that express mutated STAT3Y640F. These cells show accelerated proliferation and enhanced self-renewal potential. We integrated gene expression analyses and chromatin occupancy profiling of STAT3Y640F-transformed cells with data from patients with T-LGLL. This approach uncovered a conserved set of direct transcriptional targets of STAT3Y640F. Among these, strawberry notch homolog 2 (SBNO2) represents an essential transcriptional target, which was identified by a comparative genome-wide CRISPR/Cas9-based loss-of-function screen. The STAT3-SBNO2 axis is also present in NK-cell leukemia, T-cell non-Hodgkin lymphoma, and NPM-ALK-rearranged T-cell anaplastic large cell lymphoma (T-ALCL), which are driven by STAT3-hyperactivation/mutation. In patients with NPM-ALK+ T-ALCL, high SBNO2 expression correlates with shorter relapse-free and overall survival. Our findings identify SBNO2 as a potential therapeutic intervention site for STAT3-driven hematopoietic malignancies.
Collapse
Affiliation(s)
- Tania Brandstoetter
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eszter Doma
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Thorsten Klampfl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Eder
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Johannes Zuber
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
Salameh L, Bhamidimarri PM, Saheb Sharif-Askari N, Dairi Y, Hammoudeh SM, Mahdami A, Alsharhan M, Tirmazy SH, Rawat SS, Busch H, Hamid Q, Al Heialy S, Hamoudi R, Mahboub B. In Silico Bioinformatics Followed by Molecular Validation Using Archival FFPE Tissue Biopsies Identifies a Panel of Transcripts Associated with Severe Asthma and Lung Cancer. Cancers (Basel) 2022; 14:1663. [PMID: 35406434 PMCID: PMC8996975 DOI: 10.3390/cancers14071663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 12/04/2022] Open
Abstract
Severe asthma and lung cancer are both heterogeneous pathological diseases affecting the lung tissue. Whilst there are a few studies that suggest an association between asthma and lung cancer, to the best of our knowledge, this is the first study to identify common genes involved in both severe asthma and lung cancer. Publicly available transcriptomic data for 23 epithelial brushings from severe asthmatics and 55 samples of formalin-fixed paraffin-embedded (FFPE) lung cancer tissue at relatively early stages were analyzed by absolute gene set enrichment analysis (GSEA) in comparison to 37 healthy bronchial tissue samples. The key pathways enriched in asthmatic patients included adhesion, extracellular matrix, and epithelial cell proliferation, which contribute to tissue remodeling. In the lung cancer dataset, the main pathways identified were receptor tyrosine kinase signaling, wound healing, and growth factor response, representing the early cancer pathways. Analysis of the enriched genes derived from the pathway analysis identified seven genes expressed in both the asthma and lung cancer sets: BCL3, POSTN, PPARD, STAT1, MYC, CD44, and FOSB. The differential expression of these genes was validated in vitro in the cell lines retrieved from different lung cancer and severe asthma patients using real-time PCR. The effect of the expression of the seven genes identified in the study on the overall survival of lung cancer patients (n = 1925) was assessed using a Kaplan-Meier plot. In vivo validation performed in the archival biopsies obtained from patients diagnosed with both the disease conditions provided interesting insights into the pathogenesis of severe asthma and lung cancer, as indicated by the differential expression pattern of the seven transcripts in the mixed group as compared to the asthmatics and lung cancer samples alone.
Collapse
Affiliation(s)
- Laila Salameh
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
- Dubai Health Authority, Dubai 4545, United Arab Emirates; (Y.D.); (M.A.); (S.H.T.); (B.M.)
| | - Poorna Manasa Bhamidimarri
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
| | - Narjes Saheb Sharif-Askari
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
| | - Youssef Dairi
- Dubai Health Authority, Dubai 4545, United Arab Emirates; (Y.D.); (M.A.); (S.H.T.); (B.M.)
| | - Sarah Musa Hammoudeh
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
| | - Amena Mahdami
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
| | - Mouza Alsharhan
- Dubai Health Authority, Dubai 4545, United Arab Emirates; (Y.D.); (M.A.); (S.H.T.); (B.M.)
| | - Syed Hammad Tirmazy
- Dubai Health Authority, Dubai 4545, United Arab Emirates; (Y.D.); (M.A.); (S.H.T.); (B.M.)
| | - Surendra Singh Rawat
- Collage of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (S.S.R.); (S.A.H.)
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck 23562, Germany;
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
- Meakins-Christie Laboratories, Research Institute of the McGill University Healthy Center, Faculty of Medicine, Montreal, QC H3A 0G4, Canada
| | - Saba Al Heialy
- Collage of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (S.S.R.); (S.A.H.)
- Meakins-Christie Laboratories, Research Institute of the McGill University Healthy Center, Faculty of Medicine, Montreal, QC H3A 0G4, Canada
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; (L.S.); (P.M.B.); (N.S.S.-A.); (S.M.H.); (A.M.); (Q.H.)
- Division of Surgery and Interventional Science, University College London, London NW3 2QG, UK
| | - Bassam Mahboub
- Dubai Health Authority, Dubai 4545, United Arab Emirates; (Y.D.); (M.A.); (S.H.T.); (B.M.)
| |
Collapse
|
11
|
Sung SE, Seo MS, Kim YI, Kang KK, Choi JH, Lee S, Sung M, Yim SG, Lim JH, Seok HG, Yang SY, Lee GW. Human Epidural AD-MSC Exosomes Improve Function Recovery after Spinal Cord Injury in Rats. Biomedicines 2022; 10:678. [PMID: 35327480 PMCID: PMC8945172 DOI: 10.3390/biomedicines10030678] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 02/08/2023] Open
Abstract
Spinal cord injury (SCI) interferes with the normal function of the autonomic nervous system by blocking circuits between the sensory and motor nerves. Although many studies focus on functional recovery after neurological injury, effective neuroregeneration is still being explored. Recently, extracellular vesicles such as exosomes have emerged as cell-free therapeutic agents owing to their ability of cell-to-cell communication. In particular, exosomes released from mesenchymal stem cells (MSCs) have the potential for tissue regeneration and exhibit therapeutic effectiveness in neurological disorders. In this study, we isolated exosomes from human epidural adipose tissue-derived MSCs (hEpi AD-MSCs) using the tangential flow filtration method. The isolated exosomes were analyzed for size, concentration, shape, and major surface markers using nanoparticle tracking analysis, transmission electron microscopy, and flow cytometry. To evaluate their effect on SCI recovery, hEpi AD-MSC exosomes were injected intravenously in SCI-induced rats. hEpi AD-MSC exosomes improved the locomotor function of SCI-induced rats. The results of histopathological and cytokine assays showed that hEpi AD-MSC exosomes regulated inflammatory response. Genetic profiling of the rat spinal cord tissues revealed changes in the expression of inflammation-related genes after exosome administration. Collectively, hEpi AD-MSC exosomes are effective in restoring spinal functions by reducing the inflammatory response.
Collapse
Affiliation(s)
- Soo-Eun Sung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
- Department of Biomaterials Science (BK21 Four Program), Pusan National University, Miryang 50463, Korea;
| | - Min-Soo Seo
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | | | - Kyung-Ku Kang
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Joo-Hee Choi
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Sijoon Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Minkyoung Sung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Sang-Gu Yim
- Department of Biomaterials Science (BK21 Four Program), Pusan National University, Miryang 50463, Korea;
| | - Ju-Hyeon Lim
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170, Hyochung-ro, Namgu, Daegu 42415, Korea; (J.-H.L.); (H.-G.S.)
- Company Culture Team, Kolmar Korea Holdings 61, Heolleung-ro 8 gill, Seocho-gu, Seoul 06800, Korea
| | - Hyun-Gyu Seok
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170, Hyochung-ro, Namgu, Daegu 42415, Korea; (J.-H.L.); (H.-G.S.)
| | - Seung-Yun Yang
- Department of Biomaterials Science (BK21 Four Program), Pusan National University, Miryang 50463, Korea;
| | - Gun-Woo Lee
- Cellexobio, Co., Ltd., Daegu 42415, Korea;
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170, Hyochung-ro, Namgu, Daegu 42415, Korea; (J.-H.L.); (H.-G.S.)
| |
Collapse
|
12
|
Liu H, Zeng L, Yang Y, Guo C, Wang H. Bcl-3: A Double-Edged Sword in Immune Cells and Inflammation. Front Immunol 2022; 13:847699. [PMID: 35355979 PMCID: PMC8959985 DOI: 10.3389/fimmu.2022.847699] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
Abstract
The NF-κB transcription factor family controls the transcription of many genes and regulates a number of pivotal biological processes. Its activity is regulated by the IκB family of proteins. Bcl-3 is an atypical member of the IκB protein family that regulates the activity of nuclear factor NF-κB. It can promote or inhibit the expression of NF-κB target genes according to the received cell type and stimulation, impacting various cell functions, such as proliferation and differentiation, induction of apoptosis and immune response. Bcl-3 is also regarded as an environment-dependent cell response regulator that has dual roles in the development of B cells and the differentiation, survival and proliferation of Th cells. Moreover, it also showed a contradictory role in inflammation. At present, in addition to the work aimed at studying the molecular mechanism of Bcl-3, an increasing number of studies have focused on the effects of Bcl-3 on inflammation, immunity and malignant tumors in vivo. In this review, we focus on the latest progress of Bcl-3 in the regulation of the NF-κB pathway and its extensive physiological role in inflammation and immune cells, which may help to provide new ideas and targets for the early diagnosis or targeted treatment of various inflammatory diseases, immunodeficiency diseases and malignant tumors.
Collapse
Affiliation(s)
- Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Lin Zeng
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlei Guo
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
13
|
Wong TY, Menaga S, Huang CYF, Ho SHA, Gan SC, Lim YM. 2-Methoxy-1,4-naphthoquinone (MNQ) regulates cancer key genes of MAPK, PI3K, and NF-κB pathways in Raji cells. Genomics Inform 2022; 20:e7. [PMID: 35399006 PMCID: PMC9001993 DOI: 10.5808/gi.21041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 03/10/2022] [Indexed: 11/20/2022] Open
Abstract
2-Methoxy-1,4-naphthoquinone (MNQ) has been shown to cause cytotoxic towards various cancer cell lines. This study is designed to investigate the regulatory effect of MNQ on the key cancer genes in mitogen-activated protein kinase, phosphoinositide 3-kinase, and nuclear factor кB signaling pathways. The expression levels of the genes were compared at different time point using polymerase chain reaction arrays and Ingenuity Pathway Analysis was performed to identify gene networks that are most significant to key cancer genes. A total of 43 differentially expressed genes were identified with 21 up-regulated and 22 down-regulated genes. Up-regulated genes were involved in apoptosis, cell cycle and act as tumor suppressor while down-regulated genes were involved in anti-apoptosis, angiogenesis, cell cycle and act as transcription factor as well as proto-oncogenes. MNQ exhibited multiple regulatory effects on the cancer key genes that targeting at cell proliferation, cell differentiation, cell transformation, apoptosis, reduce inflammatory responses, inhibits angiogenesis and metastasis.
Collapse
Affiliation(s)
- Teck Yew Wong
- Centre for Cancer Research, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor, Malaysia
| | - Subramaniam Menaga
- Centre for Cancer Research, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor, Malaysia
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Siong Hock Anthony Ho
- School of Biosciences, Taylor's University, Lakeside Campus 1, 47500 Subang Jaya, Malaysia
| | - Seng Chiew Gan
- Department of Pre-Clinical Sciences Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Malaysia
| | - Yang Mooi Lim
- Centre for Cancer Research, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor, Malaysia.,Department of Pre-Clinical Sciences Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Malaysia
| |
Collapse
|
14
|
Soares-Lima SC, Gonzaga IM, Camuzi D, Nicolau-Neto P, Vieira da Silva R, Guaraldi S, Ferreira MA, Hernandez-Vargas H, Herceg Z, Ribeiro Pinto LF. IL6 and BCL3 Expression Are Potential Biomarkers in Esophageal Squamous Cell Carcinoma. Front Oncol 2021; 11:722417. [PMID: 34422669 PMCID: PMC8371528 DOI: 10.3389/fonc.2021.722417] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/22/2021] [Indexed: 12/09/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) ranks among the most lethal tumors worldwide, as a consequence of late detection and poor treatment response, evidencing the need for diagnosis anticipation and new therapeutic targets. First, we investigated the IL6 gene and protein expression in the esophagus of individuals without esophageal disorders (healthy), ESCC, and non-tumoral surrounding tissue (NTST). Our results showed that IL6 mRNA and protein expression is upregulated in tumor cells relative to NTST. In the TCGA dataset, we identified a set of genes whose expression was correlated with IL6 mRNA levels, including the antiapoptotic gene BCL3. By using an immortalized esophageal cell line, we confirmed that IL6 was capable of inducing BCL3 expression in esophageal cells. BCL3 mRNA and protein are overexpressed in ESCC and NTST compared to healthy esophagus, and BCL3 mRNA could distinguish the morphologically normal samples (healthy and NTST) with 100% sensitivity and 95.12% specificity. The spatial intratumoral heterogeneity of both IL6 and BCL3 expression was evaluated, corroborating IL6 upregulation throughout the tumor, while tumor and NTST showed a consistent increase of BCL3 expression relative to the healthy esophagus. Our study shows that IL6 overexpression seems to be a key event in ESCC carcinogenesis, contributing to ESCC through a homogeneous antiapoptotic signalling via BCL3 overexpression, thus suggesting anti-IL6 therapies to be further considered for ESCC treatment. Finally, our data support the use of BCL3 mRNA expression as a potential biomarker for ESCC detection.
Collapse
Affiliation(s)
| | - Isabela Martins Gonzaga
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Diego Camuzi
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Pedro Nicolau-Neto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Raissa Vieira da Silva
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Simone Guaraldi
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
- Seção de Endoscopia, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | | | - Hector Hernandez-Vargas
- Cancer Research Centre of Lyon (CRCL), Inserm U 1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Zdenko Herceg
- Epigenetics Group, Mechanisms of Carcinogenesis Section, International Agency for Research on Cancer (IARC), Lyon, France
| | | |
Collapse
|
15
|
Yang N, Xiong Y, Wang Y, Yi Y, Zhu J, Ma F, Li J, Liu H. ADAP Y571 Phosphorylation Is Required to Prime STAT3 for Activation in TLR4-Stimulated Macrophages. THE JOURNAL OF IMMUNOLOGY 2021; 206:814-826. [PMID: 33431658 DOI: 10.4049/jimmunol.2000569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022]
Abstract
Adhesion and degranulation-promoting adapter protein (ADAP), originally identified as an essential adaptor molecule in TCR signaling and T cell adhesion, has emerged as a critical regulator in innate immune cells such as macrophages; however, its role in macrophage polarization and inflammatory responses remains unknown. In this study, we show that ADAP plays an essential role in TLR4-mediated mouse macrophage polarization via modulation of STAT3 activity. Macrophages from ADAP-deficient mice exhibit enhanced M1 polarization, expression of proinflammatory cytokines and capacity in inducing Th1 responses, but decreased levels of anti-inflammatory cytokines in response to TLR4 activation by LPS. Furthermore, overexpression of ADAP enhances, whereas loss of ADAP reduces, the LPS-mediated phosphorylation and activity of STAT3, suggesting ADAP acts as a coactivator of STAT3 activity and function. Furthermore, the coactivator function of ADAP mostly depends on the tyrosine phosphorylation at Y571 in the motif YDSL induced by LPS. Mutation of Y571 to F severely impairs the stimulating effect of ADAP on STAT3 activity and the ability of ADAP to inhibit M1-like polarization in TLR4-activated mouse macrophages. Moreover, ADAP interacts with STAT3, and loss of ADAP renders mouse macrophages less sensitive to IL-6 stimulation for STAT3 phosphorylation. Collectively, our findings revealed an additional layer of regulation of TLR4-mediated mouse macrophage plasticity whereby ADAP phosphorylation on Y571 is required to prime STAT3 for activation in TLR4-stimulated mouse macrophages.
Collapse
Affiliation(s)
- Naiqi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu Province, China.,Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu Province, China
| | - Yiwei Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Yan Wang
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu Province, China
| | - Yulan Yi
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu Province, China
| | - Jingfei Zhu
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; and.,Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Feng Ma
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; and.,Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Jing Li
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu Province, China
| | - Hebin Liu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu Province, China; .,Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu Province, China
| |
Collapse
|
16
|
Martinez-Fabregas J, Wang L, Pohler E, Cozzani A, Wilmes S, Kazemian M, Mitra S, Moraga I. CDK8 Fine-Tunes IL-6 Transcriptional Activities by Limiting STAT3 Resident Time at the Gene Loci. Cell Rep 2020; 33:108545. [PMID: 33357429 PMCID: PMC7773550 DOI: 10.1016/j.celrep.2020.108545] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/28/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokines are highly pleiotropic ligands that regulate the immune response. Here, using interleukin-6 (IL-6) as a model system, we perform detailed phosphoproteomic and transcriptomic studies in human CD4+ T helper 1 (Th-1) cells to address the molecular bases defining cytokine functional pleiotropy. We identify CDK8 as a negative regulator of STAT3 transcriptional activities, which interacts with STAT3 upon IL-6 stimulation. Inhibition of CDK8 activity, using specific small molecule inhibitors, reduces the IL-6-induced phosphoproteome by 23% in Th-1 cells, including STAT3 S727 phosphorylation. STAT3 binding to target DNA sites in the genome is increased upon CDK8 inhibition, which results in a concomitant increase in STAT3-mediated transcriptional activity. Importantly, inhibition of CDK8 activity under Th-17 polarizing conditions results in an enhancement of Th-17 differentiation. Our results support a model where CDK8 regulates STAT3 transcriptional processivity by modulation of its gene loci resident time, critically contributing to diversification of IL-6 responses. CDK8 regulates IL-6-mediated STAT3 S727 phosphorylation in primary human T cells CDK8 controls STAT3 activity by limiting its resident time at gene loci CDK8 inhibition increases IL-6-mediated Th17 differentiation
Collapse
Affiliation(s)
| | - Luopin Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Elizabeth Pohler
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Adeline Cozzani
- Université de Lille, INSERM UMR1277 CNRS UMR9020-CANTHER and Institut pour la Recherche sur le Cancer de Lille (IRCL), Lille, France
| | - Stephan Wilmes
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Majid Kazemian
- Department of Computer Science, Purdue University, West Lafayette, IN, USA; Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
| | - Suman Mitra
- Université de Lille, INSERM UMR1277 CNRS UMR9020-CANTHER and Institut pour la Recherche sur le Cancer de Lille (IRCL), Lille, France.
| | - Ignacio Moraga
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
17
|
Carcinoma-Associated Fibroblasts Promote Growth of Sox2-Expressing Breast Cancer Cells. Cancers (Basel) 2020; 12:cancers12113435. [PMID: 33228022 PMCID: PMC7699386 DOI: 10.3390/cancers12113435] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The tumor microenvironment has a strong impact on the behavior of tumor cells. One major cell type residing in the tumor microenvironment is the carcinoma-associated fibroblast (CAF). We were interested in the effect of CAFs on Sox2 (sex determining region Y (SRY)-box 2), which not only is an essential embryonal stem cell transcription factor, but also plays a role in cancer stem cell activity. We found that long-term exposure of ERα-positive breast cancer cells to the cocktail of CAF-secreted factors strongly increased Sox2 expression involving tumor-related proteins and signaling pathways. However, Sox2 was not only present in those tumor cells that express stem cell markers, but was equally abundant in other tumor cells. By being widely expressed, Sox2 may have functions in non-stem cells. In fact, Sox2 was found to regulate ERα expression, to act anti-apoptotically, to promote cellular growth and to protect cells against the anti-estrogen fulvestrant. Abstract CAFs (Carcinoma-associated fibroblasts) play an important role in cancer progression. For instance, they promote resistance to anti-estrogens, such as fulvestrant. Here, we show that, in ERα-positive breast cancer cell lines, the cocktail of factors secreted by CAFs (CAF-CM) induce the expression of the embryonal stem cell transcription factor Sox2 (sex determining region Y (SRY)-box 2). Long-term exposure to CAF-CM was able to give rise to very high Sox2 levels both in the absence and presence of fulvestrant. IL-6 (interleukin-6), a major component of CAF-CM, failed to raise Sox2 expression. In MCF-7 sublines established in the presence of CAF-CM, almost all cells showed Sox2 expression, whereas long-term treatment of T47D cells with CAF-CM resulted in a ~60-fold increase in the proportions of two distinct populations of Sox2 high and low expresser cells. Exposure of BT474 cells to CAF-CM raised the fraction of Sox2 high expresser cells by ~3-fold. Cell sorting based on CD44 and CD24 expression or ALDH (aldehyde dehydrogenase) activity revealed that most Sox2 high expresser cells were not CD44hi/CD24lo- or ALDH-positive cells suggesting that they were not CSCs (cancer stem cells), though CD44 played a role in Sox2 expression. Functionally, Sox2 was found to protect CAF-CM-treated cells against apoptosis and to allow higher growth activity in the presence of fulvestrant. Mechanistically, the key drivers of Sox2 expression was found to be STAT3 (Signal transducer and activator of transcription 3), Bcl-3 (B-cell lymphoma 3) and the PI3K (Phosphoinositide 3-kinase)/AKT pathway, whose activities/expression can all be upregulated by CAF-CM. These data suggest that CAF-CM induces Sox2 expression in non-CSCs by activating proteins involved in growth control and drug resistance, leading to higher protection against apoptosis.
Collapse
|
18
|
Chen X, Wang C, Jiang Y, Wang Q, Tao Y, Zhang H, Zhao Y, Hu Y, Li C, Ye D, Liu D, Jiang W, Chin EY, Chen S, Liu Y, Wang M, Liu S, Zhang X. Bcl-3 promotes Wnt signaling by maintaining the acetylation of β-catenin at lysine 49 in colorectal cancer. Signal Transduct Target Ther 2020; 5:52. [PMID: 32355204 PMCID: PMC7193563 DOI: 10.1038/s41392-020-0138-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/24/2019] [Accepted: 02/18/2020] [Indexed: 11/09/2022] Open
Abstract
Wnt/β-catenin signaling plays a critical role in colorectal cancer (CRC) tumorigenesis and the homeostasis of colorectal cancer stem cells (CSCs), but its molecular mechanism remains unclear. B-cell lymphoma 3 (Bcl-3), a member of the IκB family, is overexpressed in CRC and promotes tumorigenicity. Here, we report a novel function of Bcl-3 in maintaining colorectal CSC homeostasis by activating Wnt/β-catenin signaling. Silencing Bcl-3 suppresses the self-renewal capacity of colorectal CSCs and sensitizes CRC cells to chemotherapeutic drugs through a decrease in Wnt/β-catenin signaling. Moreover, our data show that Bcl-3 is a crucial component of Wnt/β-catenin signaling and is essential for β-catenin transcriptional activity in CRC cells. Interestingly, Wnt3a increases the level and nuclear translocation of Bcl-3, which binds directly to β-catenin and enhances the acetylation of β-catenin at lysine 49 (Ac-K49-β-catenin) and transcriptional activity. Bcl-3 depletion decreases the Ac-K49-β-catenin level by increasing the level of histone deacetylase 1 to remove acetyl groups from β-catenin, thus interrupting Wnt/β-catenin activity. In CRC clinical specimens, Bcl-3 expression negatively correlates with the overall survival of CRC patients. A significantly positive correlation was found between the expression of Bcl-3 and Ac-K49-β-catenin. Collectively, our data reveal that Bcl-3 plays a crucial role in CRC chemoresistance and colorectal CSC maintenance via its modulation of the Ac-K49-β-catenin, which serves as a promising therapeutic target for CRC.
Collapse
Affiliation(s)
- Xi Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Chen Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yuhang Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Qi Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Yu Tao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Haohao Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Yongxu Zhao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Yiming Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Cuifeng Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Deji Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Dandan Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Wenxia Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Eugene Y Chin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Sheng Chen
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Mingliang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sanhong Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Xiaoren Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China.
| |
Collapse
|
19
|
Collard TJ, Fallatah HM, Greenhough A, Paraskeva C, Williams AC. BCL‑3 promotes cyclooxygenase‑2/prostaglandin E2 signalling in colorectal cancer. Int J Oncol 2020; 56:1304-1313. [PMID: 32319612 DOI: 10.3892/ijo.2020.5013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/26/2020] [Indexed: 11/05/2022] Open
Abstract
First discovered as an oncogene in leukaemia, recent reports highlight an emerging role for the proto‑oncogene BCL‑3 in solid tumours. Importantly, BCL‑3 expression is upregulated in >30% of colorectal cancer cases and is reported to be associated with a poor prognosis. However, the mechanism by which BCL‑3 regulates tumorigenesis in the large intestine is yet to be fully elucidated. In the present study, it was shown for the first time that knocking down BCL‑3 expression suppressed cyclooxygenase‑2 (COX‑2)/prostaglandin E2 (PGE2) signalling in colorectal cancer cells, a pathway known to drive several of the hallmarks of cancer. RNAi‑mediated suppression of BCL‑3 expression decreased COX‑2 expression in colorectal cancer cells both at the mRNA and protein level. This reduction in COX‑2 expression resulted in a significant and functional reduction (30‑50%) in the quantity of pro‑tumorigenic PGE2 produced by the cancer cells, as shown by enzyme linked immunoassays and medium exchange experiments. In addition, inhibition of BCL‑3 expression also significantly suppressed cytokine‑induced (TNF‑α or IL‑1β) COX‑2 expression. Taken together, the results of the present study identified a novel role for BCL‑3 in colorectal cancer and suggested that expression of BCL‑3 may be a key determinant in the COX‑2‑meditated response to inflammatory cytokines in colorectal tumour cells. These results suggest that targeting BCL‑3 to suppress PGE2 synthesis may represent an alternative or complementary approach to using non‑steroidal anti‑inflammatory drugs [(NSAIDs), which inhibit cyclooxygenase activity and suppress the conversion of arachidonic acid to prostaglandin], for prevention and/or recurrence in PGE2‑driven tumorigenesis.
Collapse
Affiliation(s)
- Tracey Jane Collard
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Hafsah Mohammed Fallatah
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Alexander Greenhough
- Health and Applied Sciences, University of The West of England, Bristol BS16 1QY, United Kingdom
| | - Christos Paraskeva
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Ann Caroline Williams
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
20
|
Jin J, He X, Silva E. Stable intronic sequence RNAs (sisRNAs) are selected regions in introns with distinct properties. BMC Genomics 2020; 21:287. [PMID: 32264855 PMCID: PMC7137253 DOI: 10.1186/s12864-020-6687-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 03/18/2020] [Indexed: 12/31/2022] Open
Abstract
Background Stable introns and intronic fragments make up the largest population of RNA in the oocyte nucleus of the frog Xenopus tropicalis. These stable intronic sequence RNAs (sisRNAs) persist through the onset of zygotic transcription when synchronous cell division has ended, and the developing embryo consists of approximately 8000 cells. Despite their abundance, the sequence properties and biological function of sisRNAs are just beginning to be understood. Results To characterize this population of non-coding RNA, we identified all of the sisRNAs in the X. tropicalis oocyte nucleus using published high-throughput RNA sequencing data. Our analysis revealed that sisRNAs, have an average length of ~ 360 nt, are widely expressed from genes with multiple introns, and are derived from specific regions of introns that are GC and TG rich, while CpG poor. They are enriched in introns at both ends of transcripts but preferentially at the 3′ end. The consensus binding sites of specific transcription factors such as Stat3 are enriched in sisRNAs, suggesting an association between sisRNAs and transcription factors involved in early development. Evolutionary conservation analysis of sisRNA sequences in seven vertebrate genomes indicates that sisRNAs are as conserved as other parts of introns, but much less conserved than exons. Conclusion In total, our results indicate sisRNAs are selected intron regions with distinct properties and may play a role in gene expression regulation.
Collapse
Affiliation(s)
- Jing Jin
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.,Department of Biology, Georgetown University, 37th and O Sts, NW, Washington DC, 20057, USA
| | - Ximiao He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Elena Silva
- Department of Biology, Georgetown University, 37th and O Sts, NW, Washington DC, 20057, USA.
| |
Collapse
|
21
|
Vennou KE, Piovani D, Kontou PI, Bonovas S, Bagos PG. Multiple outcome meta-analysis of gene-expression data in inflammatory bowel disease. Genomics 2020; 112:1761-1767. [DOI: 10.1016/j.ygeno.2019.09.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/02/2023]
|
22
|
Dittmer A, Lange T, Leyh B, Dittmer J. Protein‑ and growth‑modulatory effects of carcinoma‑associated fibroblasts on breast cancer cells: Role of interleukin‑6. Int J Oncol 2019; 56:258-272. [PMID: 31789400 PMCID: PMC6910226 DOI: 10.3892/ijo.2019.4918] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023] Open
Abstract
Carcinoma-associated fibroblasts (CAFs) secrete factors that increase the expression and/or activities of proteins in breast cancer cells and induce resistance to anti-estrogens, such as fulvestrant. A major factor is interleukin-6 (IL-6). This study demonstrated that, across estrogen receptor (ER) α-positive and -negative cell lines, recombinant human IL-6 (rhIL-6) mimicked most of the CAF-conditioned medium (CM)-induced changes in protein expression patterns; however, in most cases, it failed to recapitulate CAF-CM-triggered alterations in ERK1/2 and AKT activities. The ability of rhIL-6 to induce fulvestrant resistance was dependent upon the culture conditions. In 3D, but not in 2D cultures, rhIL-6 increased the survival of fulvestrant-treated cells, although not to the same extent as observed with CAF-CM. In 2D cultures, rhIL-6 acted in a pro-apoptotic manner and decreased the expression of ATP-binding cassette transporter G2 (ABCG2). The inhibition of the PI3K/AKT pathway had similar effects on apoptosis and ABCG2 expression, linking the failure of rhIL-6 to induce fulvestrant resistance to its inability to activate the PI3K/AKT pathway. In 3D cultures, both CAF-CM and rhIL-6 acted in an anti-apoptotic manner. These activities are likely independent on the PI3K/AKT pathway and ABCG2. Experiments on ERα-negative breast cancer cells revealed a growth-inhibitory effects of both CAF-CM and rhIL-6, which coincided with a reduction in the c-Myc level. These data suggest that IL-6 plays a role in several effects of CAF-CM, including alterations in protein expression patterns, fulvestrant resistance in 3D cultures and growth inhibition. By contrast, IL-6 is unlikely to be responsible for the CAF-CM-induced activation of the PI3K/AKT pathway and fulvestrant resistance in 2D cultures.
Collapse
Affiliation(s)
- Angela Dittmer
- Clinic for Gynecology, Martin Luther University Halle‑Wittenberg, 06120 Halle/Saale, Germany
| | - Theresia Lange
- Clinic for Gynecology, Martin Luther University Halle‑Wittenberg, 06120 Halle/Saale, Germany
| | - Benjamin Leyh
- Clinic for Gynecology, Martin Luther University Halle‑Wittenberg, 06120 Halle/Saale, Germany
| | - Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle‑Wittenberg, 06120 Halle/Saale, Germany
| |
Collapse
|
23
|
Anderson AE, Maney NJ, Nair N, Lendrem DW, Skelton AJ, Diboll J, Brown PM, Smith GR, Carmody RJ, Barton A, Isaacs JD, Pratt AG. Expression of STAT3-regulated genes in circulating CD4+ T cells discriminates rheumatoid arthritis independently of clinical parameters in early arthritis. Rheumatology (Oxford) 2019; 58:1250-1258. [PMID: 30753680 PMCID: PMC6587924 DOI: 10.1093/rheumatology/kez003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 12/13/2018] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Dysregulated signal transduction and activator of transcription-3 (STAT3) signalling in CD4+ T cells has been proposed as an early pathophysiological event in RA. We sought further evidence for this observation, and to determine its clinical relevance. METHODS Microarray technology was used to measure gene expression in purified peripheral blood CD4+ T cells from treatment-naïve RA patients and disease controls newly recruited from an early arthritis clinic. Analysis focused on 12 previously proposed transcripts, and concurrent STAT3 pathway activation was determined in the same cells by flow cytometry. A pooled analysis of previous and current gene expression findings incorporated detailed clinical parameters and employed multivariate analysis. RESULTS In an independent cohort of 161 patients, expression of 11 of 12 proposed signature genes differed significantly between RA patients and controls, robustly validating the earlier findings. Differential regulation was most pronounced for the STAT3 target genes PIM1, BCL3 and SOCS3 (>1.3-fold difference; P < 0.005), each of whose expression correlated strongly with paired intracellular phospho-STAT3. In a meta-analysis of 279 patients the same three genes accounted for the majority of the signature's ability to discriminate RA patients, which was found to be independent of age, joint involvement or acute phase response. CONCLUSION The STAT3-mediated dysregulation of BCL3, SOCS3 and PIM1 in circulating CD4+ T cells is a discriminatory feature of early RA that occurs independently of acute phase response. The mechanistic and functional implications of this observation at a cellular level warrant clarification.
Collapse
Affiliation(s)
- Amy E Anderson
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Nicola J Maney
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Nisha Nair
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, and NIHR Manchester Musculoskeletal Biomedical Research Centre, Manchester NHS Foundation Trust, Manchester, UK
| | - Dennis W Lendrem
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew J Skelton
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, UK
| | - Julie Diboll
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Philip M Brown
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Graham R Smith
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, UK
| | - Ruaidhrí J Carmody
- Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Anne Barton
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, and NIHR Manchester Musculoskeletal Biomedical Research Centre, Manchester NHS Foundation Trust, Manchester, UK
| | - John D Isaacs
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
24
|
Wang Z, Zhu F. The expression profiles of immune genes in Mus musculus macrophages during Staphylococcus aureus infection. PLoS One 2018; 13:e0190490. [PMID: 29304086 PMCID: PMC5755788 DOI: 10.1371/journal.pone.0190490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 12/17/2017] [Indexed: 01/15/2023] Open
Abstract
Staphylococcus aureus is an important pathogen which is often the cause of major morbidity and mortality in both hospital and community settings. For this reason, we investigated the host cell early immune resoponse to S. aureus infection using genome-wide analysis. To do this, we infected Mus musculus RAW264.7 cells with S. aureus alone or in the presence of free peptidoglycan (PG), which appears in the S. aureus cell wall. Post infection, we performed a genome-wide analysis of RAW246.7 cells to identify significant changes in the gene expression profile. Further, we analyzed the infected RAW246.7 cells with transmission electron microscopy looking for the presence of bacterial cells inside the host cell. We also used flow cytometry to determine whether cells had induced apoptosis. The results showed that S. aureus induced apoptosis in the RAW246.7 cells but did not effectively clear away intracellular bacteria cells. However, S. aureus + PG treatment inhibited the apoptosis and activated the host cell inflammation response, possibly involving NF-κB and JAK-STAT pathways, as identified by genome-wide analysis, in RAW246.7 cells. Our study demonstrated for the first time that an independent application of free PG was capable of activating immune responses the host cells.
Collapse
Affiliation(s)
- Ziyan Wang
- College of Animal Science and Technology, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Fei Zhu
- College of Animal Science and Technology, Zhejiang Agriculture and Forestry University, Hangzhou, China
- * E-mail:
| |
Collapse
|
25
|
Tohyama M, Shirakata Y, Hanakawa Y, Dai X, Shiraishi K, Murakami M, Miyawaki S, Mori H, Utsunomiya R, Masuda K, Hashimoto K, Sayama K. Bcl-3 induced by IL-22 via STAT3 activation acts as a potentiator of psoriasis-related gene expression in epidermal keratinocytes. Eur J Immunol 2018; 48:168-179. [PMID: 28901004 DOI: 10.1002/eji.201747017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 07/26/2017] [Accepted: 09/08/2017] [Indexed: 12/25/2022]
Abstract
IL-22 induces STAT3 phosphorylation and mediates psoriasis-related gene expression. However, the signaling mechanism leading from pSTAT3 to the expression of these genes remains unclear. We focused on Bcl-3, which is induced by STAT3 activation and mediates gene expression. In cultured human epidermal keratinocytes, IL-22 increased Bcl-3, which was translocated to the nucleus with p50 via STAT3 activation. The increases in CXCL8, S100As and human β-defensin 2 mRNA expression caused by IL-22 were abolished by siRNA against Bcl-3. Although CCL20 expression was also augmented by IL-22, the knockdown of Bcl-3 increased its level. Moreover, the combination of IL-22 and IL-17A enhanced Bcl-3 production, IL-22-induced gene expression, and the expression of other psoriasis-related genes, including those encoding IL-17C, IL-19, and IL-36γ. The expression of these genes (except for CCL20) was also suppressed by the knockdown of Bcl-3. Bcl-3 overexpression induced CXCL8 and HBD2 expression but not S100As expression. We also compared Bcl-3 expression between psoriatic skin lesions and normal skin. Immunostaining revealed strong signals for Bcl-3 and p50 in the nucleus of epidermal keratinocytes from psoriatic skin. The IL-22-STAT3-Bcl-3 pathway may be important in the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Mikiko Tohyama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yuji Shirakata
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yasushi Hanakawa
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Saori Miyawaki
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryo Utsunomiya
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kana Masuda
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Hashimoto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Sayama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
26
|
Early-in-life dietary zinc deficiency and supplementation and mammary tumor development in adulthood female rats. J Nutr Biochem 2017; 44:71-79. [DOI: 10.1016/j.jnutbio.2017.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/16/2016] [Accepted: 03/02/2017] [Indexed: 11/19/2022]
|
27
|
Faridi U, Dhawan SS, Pal S, Gupta S, Shukla AK, Darokar MP, Sharma A, Shasany AK. Repurposing L-Menthol for Systems Medicine and Cancer Therapeutics? L-Menthol Induces Apoptosis through Caspase 10 and by Suppressing HSP90. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:53-64. [PMID: 26760959 DOI: 10.1089/omi.2015.0118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The objective of the present study was to repurpose L-menthol, which is frequently used in oral health and topical formulations, for cancer therapeutics. In this article, we argue that monoterpenes such as L-menthol might offer veritable potentials in systems medicine, for example, as cheaper anti-cancer compounds. Other monoterpenes such as limonene, perillyl alcohol, and geraniol have been shown to induce apoptosis in various cancer cell lines, but their mechanisms of action are yet to be completely elucidated. Earlier, we showed that L-menthol modulates tubulin polymerization and apoptosis to inhibit cancer cell proliferation. In the present report, we used an apoptosis-related gene microarray in conjunction with proteomics analyses, as well as in silico interpretations, to study gene expression modulation in human adenocarcinoma Caco-2 cell line in response to L-menthol treatment. The microarray analysis identified caspase 10 as the important initiator caspase, instead of caspase 8. The proteomics analyses showed downregulation of HSP90 protein (also corroborated by its low transcript abundance), which in turn indicated inhibition of AKT-mediated survival pathway, release of pro-apoptotic factor BAD from BAD and BCLxL complex, besides regulation of other factors related to apoptosis. Based on the combined microarray, proteomics, and in silico data, a signaling pathway for L-menthol-induced apoptosis is being presented for the first time here. These data and literature analysis have significant implications for "repurposing" L-menthol beyond oral medicine, and in understanding the mode of action of plant-derived monoterpenes towards development of cheaper anticancer drugs in future.
Collapse
Affiliation(s)
- Uzma Faridi
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Sunita S Dhawan
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Shaifali Pal
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Sanchita Gupta
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ashutosh K Shukla
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Mahendra P Darokar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ajit K Shasany
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| |
Collapse
|
28
|
Peñaloza HF, Schultz BM, Nieto PA, Salazar GA, Suazo I, Gonzalez PA, Riedel CA, Alvarez-Lobos MM, Kalergis AM, Bueno SM. Opposing roles of IL-10 in acute bacterial infection. Cytokine Growth Factor Rev 2016; 32:17-30. [PMID: 27522641 DOI: 10.1016/j.cytogfr.2016.07.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Interleukin-10 (IL-10) is recognized as an anti-inflammatory cytokine that downmodulates inflammatory immune responses at multiple levels. In innate cells, production of this cytokine is usually triggered after pathogen recognition receptor (PRR) engagement by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patters (DAMPs), as well as by other soluble factors. Importantly, IL-10 is frequently secreted during acute bacterial infections and has been described to play a key role in infection resolution, although its effects can significantly vary depending on the infecting bacterium. While the production of IL-10 might favor host survival in some cases, it may also result harmful for the host in other circumstances, as it can prevent appropriate bacterial clearance. In this review we discuss the role of IL-10 in bacterial clearance and propose that this cytokine is required to recover from infection caused by extracellular or highly pro-inflammatory bacteria. Altogether, we propose that IL-10 drives excessive suppression of the immune response upon infection with intracellular bacteria or in non-inflammatory bacterial infections, which ultimately favors bacterial persistence and dissemination within the host. Thus, the nature of the bacterium causing infection is an important factor that needs to be taken into account when considering new immunotherapies that consist on the modulation of inflammation, such as IL-10. Indeed, induction of this cytokine may significantly improve the host's immune response to certain bacteria when antibiotics are not completely effective.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Barbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pamela A Nieto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Geraldyne A Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Isidora Suazo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pablo A Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Chile
| | - Manuel M Alvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France.
| |
Collapse
|
29
|
Webb LV, Ley SC, Seddon B. TNF activation of NF-κB is essential for development of single-positive thymocytes. J Exp Med 2016; 213:1399-407. [PMID: 27432943 PMCID: PMC4986527 DOI: 10.1084/jem.20151604] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/27/2016] [Indexed: 01/19/2023] Open
Abstract
Seddon and colleagues study mice whose T cells lack both of the catalytic subunits of the IKK complex and show that impaired TNF receptor activation of NF-κB is responsible for their block in thymocyte development. NF-κB activation has been implicated at multiple stages of thymic development of T cells, during which it is thought to mediate developmental signals originating from the T cell receptor (TCR). However, the Card11–Bcl10–Malt1 (CBM) complex that is essential for TCR activation of NF-κB in peripheral T cells is not required for thymocyte development. It has remained unclear whether the TCR activates NF-κB independent of the CBM complex in thymocyte development or whether another NF-κB activating receptor is involved. In the present study, we generated mice in which T cells lacked expression of both catalytic subunits of the inhibitor of κB kinase (IKK) complex, IKK1 and IKK2, to investigate this question. Although early stages of T cell development were unperturbed, maturation of CD4 and CD8 single-positive (SP) thymocytes was blocked in mice lacking IKK1/2 in the T cell lineage. We found that IKK1/2-deficient thymocytes were specifically sensitized to TNF-induced cell death in vitro. Furthermore, the block in thymocyte development in IKK1/2-deficient mice could be rescued by blocking TNF with anti-TNF mAb or by ablation of TNFRI expression. These experiments reveal an essential role for TNF activation of NF-κB to promote the survival and development of single positive T cells in the thymus.
Collapse
Affiliation(s)
- Louise V Webb
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, England, UK
| | - Steven C Ley
- Francis Crick Institute, Mill Hill Laboratories, London NW7 1AA, England, UK
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, England, UK
| |
Collapse
|
30
|
Urban BC, Collard TJ, Eagle CJ, Southern SL, Greenhough A, Hamdollah-Zadeh M, Ghosh A, Poulsom R, Paraskeva C, Silver A, Williams AC. BCL-3 expression promotes colorectal tumorigenesis through activation of AKT signalling. Gut 2016; 65:1151-64. [PMID: 26033966 PMCID: PMC4941180 DOI: 10.1136/gutjnl-2014-308270] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/21/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Colorectal cancer remains the fourth most common cause of cancer-related mortality worldwide. Here we investigate the role of nuclear factor-κB (NF-κB) co-factor B-cell CLL/lymphoma 3 (BCL-3) in promoting colorectal tumour cell survival. DESIGN Immunohistochemistry was carried out on 47 tumour samples and normal tissue from resection margins. The role of BCL-3/NF-κB complexes on cell growth was studied in vivo and in vitro using an siRNA approach and exogenous BCL-3 expression in colorectal adenoma and carcinoma cells. The question whether BCL-3 activated the AKT/protein kinase B (PKB) pathway in colorectal tumour cells was addressed by western blotting and confocal microscopy, and the ability of 5-aminosalicylic acid (5-ASA) to suppress BCL-3 expression was also investigated. RESULTS We report increased BCL-3 expression in human colorectal cancers and demonstrate that BCL-3 expression promotes tumour cell survival in vitro and tumour growth in mouse xenografts in vivo, dependent on interaction with NF-κB p50 or p52 homodimers. We show that BCL-3 promotes cell survival under conditions relevant to the tumour microenvironment, protecting both colorectal adenoma and carcinoma cells from apoptosis via activation of the AKT survival pathway: AKT activation is mediated via both PI3K and mammalian target of rapamycin (mTOR) pathways, leading to phosphorylation of downstream targets GSK-3β and FoxO1/3a. Treatment with 5-ASA suppressed BCL-3 expression in colorectal cancer cells. CONCLUSIONS Our study helps to unravel the mechanism by which BCL-3 is linked to poor prognosis in colorectal cancer; we suggest that targeting BCL-3 activity represents an exciting therapeutic opportunity potentially increasing the sensitivity of tumour cells to conventional therapy.
Collapse
Affiliation(s)
- Bettina C Urban
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| | - Tracey J Collard
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| | - Catherine J Eagle
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| | | | | | | | - Anil Ghosh
- Centre for Digestive Diseases, National Centre for Bowel Research and Surgical Intervention, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Whitechapel, London, UK
| | - Richard Poulsom
- Centre for Digestive Diseases, National Centre for Bowel Research and Surgical Intervention, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Whitechapel, London, UK
| | - Christos Paraskeva
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| | - Andrew Silver
- Centre for Digestive Diseases, National Centre for Bowel Research and Surgical Intervention, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Whitechapel, London, UK
| | - Ann C Williams
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
31
|
Berner A, Bachmann M, Bender C, Pfeilschifter J, Christen U, Mühl H. Though Active on RINm5F Insulinoma Cells and Cultured Pancreatic Islets, Recombinant IL-22 Fails to Modulate Cytotoxicity and Disease in a Protocol of Streptozotocin-Induced Experimental Diabetes. Front Pharmacol 2016; 6:317. [PMID: 26793108 PMCID: PMC4709444 DOI: 10.3389/fphar.2015.00317] [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: 10/10/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-22 is a cytokine displaying tissue protective and pro-regenerative functions in various preclinical disease models. Anti-bacterial, pro-proliferative, and anti-apoptotic properties mediated by activation of the transcription factor signal transducer and activator of transcription (STAT)-3 are key to biological functions of this IL-10 family member. Herein, we introduce RINm5F insulinoma cells as rat β-cell line that, under the influence of IL-22, displays activation of STAT3 with induction of its downstream gene targets Socs3, Bcl3, and Reg3b. In addition, IL-22 also activates STAT1 in this cell type. To refine those observations, IL-22 biological activity was evaluated using ex vivo cultivated murine pancreatic islets. In accord with data on RINm5F cells, islet exposure to IL-22 activated STAT3 and upregulation of STAT3-inducible Socs3, Bcl3, and Steap4 was evident under those conditions. As these observations supported the hypothesis that IL-22 may exert protective functions in toxic β-cell injury, application of IL-22 was investigated in murine multiple-low-dose streptozotocin (STZ)-induced diabetes. For that purpose, recombinant IL-22 was administered thrice either immediately before and at disease onset (at d4, d6, d8) or closely thereafter (at d8, d10, d12). These two IL-22-treatment periods coincide with two early peaks of β-cell injury detectable in this model. Notably, none of the two IL-22-treatment strategies affected diabetes incidence or blood glucose levels in STZ-treated mice. Moreover, pathological changes in islet morphology analyzed 28 days after disease induction were not ameliorated by IL-22 administration. Taken together, despite being active on rat RINm5F insulinoma cells and murine pancreatic islets, recombinant IL-22 fails to protect pancreatic β-cells in the tested protocols from toxic effects of STZ and thus is unable to ameliorate disease in the widely used model of STZ-induced diabetes.
Collapse
Affiliation(s)
- Anika Berner
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt Frankfurt am Main, Germany
| | - Malte Bachmann
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt Frankfurt am Main, Germany
| | - Christine Bender
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt Frankfurt am Main, Germany
| | - Urs Christen
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt Frankfurt am Main, Germany
| | - Heiko Mühl
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt Frankfurt am Main, Germany
| |
Collapse
|
32
|
Meguro K, Suzuki K, Hosokawa J, Sanayama Y, Tanaka S, Furuta S, Ikeda K, Takatori H, Suto A, Sakamoto A, Ohara O, Nakajima H. Role of Bcl-3 in the Development of Follicular Helper T Cells and in the Pathogenesis of Rheumatoid Arthritis. Arthritis Rheumatol 2015; 67:2651-60. [DOI: 10.1002/art.39266] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 06/25/2015] [Indexed: 12/12/2022]
Affiliation(s)
| | - Kotaro Suzuki
- Chiba University Graduate School of Medicine; Chiba Japan
| | | | | | - Shigeru Tanaka
- Chiba University Graduate School of Medicine; Chiba Japan
| | | | - Kei Ikeda
- Chiba University Graduate School of Medicine; Chiba Japan
| | | | - Akira Suto
- Chiba University Graduate School of Medicine; Chiba Japan
| | - Akemi Sakamoto
- Chiba University Graduate School of Medicine; Chiba Japan
| | | | | |
Collapse
|
33
|
Chen CY, Lee DS, Yan YT, Shen CN, Hwang SM, Lee ST, Hsieh PC. Bcl3 Bridges LIF-STAT3 to Oct4 Signaling in the Maintenance of Naïve Pluripotency. Stem Cells 2015; 33:3468-80. [DOI: 10.1002/stem.2201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/25/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Chen-Yun Chen
- Program in Molecular Medicine; National Yang-Ming University and Academia Sinica; Taipei Taiwan
- Institute of Biomedical Science; Academia Sinica; Taipei Taiwan
| | - Desy S. Lee
- Institute of Clinical Medicine; National Cheng Kung University and Hospital; Tainan Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Science; Academia Sinica; Taipei Taiwan
| | - Chia-Ning Shen
- Genomics Research Center; Academia Sinica; Taipei Taiwan
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center; Food Industry Research and Development Institute; Hsinchu Taiwan
| | - Sho Tone Lee
- Institute of Biomedical Science; Academia Sinica; Taipei Taiwan
| | - Patrick C.H. Hsieh
- Program in Molecular Medicine; National Yang-Ming University and Academia Sinica; Taipei Taiwan
- Institute of Biomedical Science; Academia Sinica; Taipei Taiwan
- Institute of Clinical Medicine; National Cheng Kung University and Hospital; Tainan Taiwan
| |
Collapse
|
34
|
Popp MW, Maquat LE. Attenuation of nonsense-mediated mRNA decay facilitates the response to chemotherapeutics. Nat Commun 2015; 6:6632. [PMID: 25808464 PMCID: PMC4375787 DOI: 10.1038/ncomms7632] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 02/13/2015] [Indexed: 12/27/2022] Open
Abstract
Nonsense-mediated mRNA decay (NMD) limits the production of aberrant mRNAs containing a premature termination codon and also controls the levels of endogenous transcripts. Here we show that when human cells are treated with clinically used chemotherapeutic compounds, NMD activity declines partly as a result of the proteolytic production of a dominant-interfering form of the key NMD factor UPF1. Production of cleaved UPF1 functions to upregulate genes involved in the response to apoptotic stresses. The biological consequence is the promotion of cell death. Combined exposure of cells to a small-molecule inhibitor of NMD, NMDI-1, and the chemotherapeutic doxorubicin leads to enhanced cell death, while inhibiting UPF1 cleavage protects cells from doxorubicin challenge. We propose a model to explain why the expression levels of genes producing mRNAs of diverse structure that encode proteins of diverse function are under the purview of NMD.
Collapse
Affiliation(s)
- Maximilian W Popp
- 1] Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA [2] Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| | - Lynne E Maquat
- 1] Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA [2] Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| |
Collapse
|
35
|
Zhang H, Hu H, Greeley N, Jin J, Matthews AJ, Ohashi E, Caetano MS, Li HS, Wu X, Mandal PK, McMurray JS, Moghaddam SJ, Sun SC, Watowich SS. STAT3 restrains RANK- and TLR4-mediated signalling by suppressing expression of the E2 ubiquitin-conjugating enzyme Ubc13. Nat Commun 2014; 5:5798. [PMID: 25503582 PMCID: PMC4270087 DOI: 10.1038/ncomms6798] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/10/2014] [Indexed: 01/05/2023] Open
Abstract
The transcriptional regulator STAT3 curbs pro-inflammatory cytokine production mediated by NF-κB signaling in innate immune cells, yet the mechanism by which this occurs has been unclear. Here we identify STAT3 as a pivotal negative regulator of Ubc13, an E2 ubiquitin-conjugating enzyme that facilitates TRAF6 K63-linked ubiquitination and NF-κB activation. Ubc13 accumulates intracellularly in the absence of STAT3. Depletion of Ubc13 in Stat3-deficient macrophages subdues excessive RANKL- or LPS-dependent gene expression, indicating Ubc13 overexpression mediates enhanced transcriptional responses in the absence of STAT3. In RANKL-activated macrophages, STAT3 is stimulated by autocrine IL-6 and inhibits accrual of Ets-1, Set1 methyltransferase and trimethylation of histone H3 lysine 4 (H3K4me3) at the Ube2n (Ubc13) promoter. These results delineate a mechanism by which STAT3 operates as a transcriptional repressor on Ube2n, thus modulating NF-κB activity by regulation of Ubc13 abundance. Our data suggest this pathway plays important roles in bone homeostasis and restraint of inflammation.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Hongbo Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Nathaniel Greeley
- 1] Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA [2] The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA
| | - Jin Jin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Allison J Matthews
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Erika Ohashi
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Mauricio S Caetano
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xuefeng Wu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Pijus K Mandal
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John S McMurray
- 1] The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA [2] Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Seyed Javad Moghaddam
- 1] The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA [2] Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shao-Cong Sun
- 1] Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA [2] The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA
| | - Stephanie S Watowich
- 1] Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA [2] The University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA
| |
Collapse
|
36
|
Phesse TJ, Buchert M, Stuart E, Flanagan DJ, Faux M, Afshar-Sterle S, Walker F, Zhang HH, Nowell CJ, Jorissen R, Tan CW, Hirokawa Y, Eissmann MF, Poh AR, Malaterre J, Pearson HB, Kirsch DG, Provero P, Poli V, Ramsay RG, Sieber O, Burgess AW, Huszar D, Vincan E, Ernst M. Partial inhibition of gp130-Jak-Stat3 signaling prevents Wnt-β-catenin-mediated intestinal tumor growth and regeneration. Sci Signal 2014; 7:ra92. [PMID: 25270258 DOI: 10.1126/scisignal.2005411] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Most colon cancers arise from somatic mutations in the tumor suppressor gene APC (adenomatous polyposis coli), and these mutations cause constitutive activation of the Wnt-to-β-catenin pathway in the intestinal epithelium. Because Wnt-β-catenin signaling is required for homeostasis and regeneration of the adult intestinal epithelium, therapeutic targeting of this pathway is challenging. We found that genetic activation of the cytokine-stimulated pathway mediated by the receptor gp130, the associated Jak (Janus kinase) kinases, and the transcription factor Stat3 (signal transducer and activator of transcription 3) was required for intestinal regeneration in response to irradiation-induced damage in wild-type mice and for tumorigenesis in Apc-mutant mice. Systemic pharmacological or partial genetic inhibition of gp130-Jak-Stat3 signaling suppressed intestinal regeneration, the growth of tumors in Apc-mutant mice, and the growth of colon cancer xenografts. The growth of Apc-mutant tumors depended on gp130-Jak-Stat3 signaling for induction of the polycomb repressor Bmi-1, and the associated repression of genes encoding the cell cycle inhibitors p16 and p21. However, suppression of gp130-Jak-Stat3 signaling did not affect Wnt-β-catenin signaling or homeostasis in the intestine. Thus, these data not only suggest a molecular mechanism for how the gp130-Jak-Stat3 pathway can promote cancer but also provide a rationale for therapeutic inhibition of Jak in colon cancer.
Collapse
Affiliation(s)
- Toby J Phesse
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Michael Buchert
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Emma Stuart
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Dustin J Flanagan
- Department of Anatomy and Cell Biology, University of Melbourne, Melbourne, Victoria 3052, Australia. Victorian Infectious Diseases Reference Laboratories, North Melbourne, Victoria 3051, Australia. School of Biomedical Sciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Maree Faux
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Shoukat Afshar-Sterle
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Francesca Walker
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Hui-Hua Zhang
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Cameron J Nowell
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Robert Jorissen
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Chin Wee Tan
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Yumiko Hirokawa
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Moritz F Eissmann
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Ashleigh R Poh
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Jordane Malaterre
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne, Victoria 3052, Australia
| | - Helen B Pearson
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne, Victoria 3052, Australia
| | - David G Kirsch
- Departments of Radiation Oncology, Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Paolo Provero
- Department of Genetics, Biology and Biochemistry, University of Turin, 10126 Torino, Italy
| | - Valeria Poli
- Department of Genetics, Biology and Biochemistry, University of Turin, 10126 Torino, Italy
| | - Robert G Ramsay
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne, Victoria 3052, Australia
| | - Oliver Sieber
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Antony W Burgess
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia
| | | | - Elizabeth Vincan
- Department of Anatomy and Cell Biology, University of Melbourne, Melbourne, Victoria 3052, Australia. Victorian Infectious Diseases Reference Laboratories, North Melbourne, Victoria 3051, Australia. School of Biomedical Sciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Matthias Ernst
- Ludwig Institute for Cancer Research, Melbourne, Victoria 3052, Australia. Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia. Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia.
| |
Collapse
|
37
|
Mughal TI, Girnius S, Rosen ST, Kumar S, Wiestner A, Abdel-Wahab O, Kiladjian JJ, Wilson WH, Van Etten RA. Emerging therapeutic paradigms to target the dysregulated Janus kinase/signal transducer and activator of transcription pathway in hematological malignancies. Leuk Lymphoma 2014; 55:1968-79. [PMID: 24206094 DOI: 10.3109/10428194.2013.863307] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Over the past decade, there has been increasing biochemical evidence that the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is aberrantly activated in malignant cells from patients with a wide spectrum of cancers of the blood and immune systems. The emerging availability of small molecule inhibitors of JAK and other signaling molecules in the JAK/STAT pathway has allowed preclinical studies validating an important role of this pathway in the pathogenesis of many hematologic malignancies, and provided motivation for new strategies for treatment of these diseases. Here, a round-table panel of experts review the current preclinical and clinical landscape of the JAK/STAT pathway in acute lymphoid and myeloid leukemias, lymphomas and myeloma, and chronic myeloid neoplasms.
Collapse
|
38
|
Huang X, Meng B, Iqbal J, Ding BB, Perry AM, Cao W, Smith LM, Bi C, Jiang C, Greiner TC, Weisenburger DD, Rimsza L, Rosenwald A, Ott G, Delabie J, Campo E, Braziel RM, Gascoyne RD, Cook JR, Tubbs RR, Jaffe ES, Armitage JO, Vose JM, Staudt LM, McKeithan TW, Chan WC, Ye BH, Fu K. Activation of the STAT3 signaling pathway is associated with poor survival in diffuse large B-cell lymphoma treated with R-CHOP. J Clin Oncol 2013; 31:4520-8. [PMID: 24220563 PMCID: PMC3871513 DOI: 10.1200/jco.2012.45.6004] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE We previously reported that constitutive STAT3 activation is a prominent feature of the activated B-cell subtype of diffuse large B-cell lymphomas (ABC-DLBCL). In this study, we investigated whether STAT3 activation can risk stratify patients with DLBCL. PATIENTS AND METHODS By an immunohistochemical method, we investigated phosphotyrosine STAT3 (PY-STAT3) expression from 185 patients with DLBCL treated with R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone). Cell line-based siRNA experiments were also performed to generate an 11-gene, PY-STAT3 activation signature, which was used to study a previously published cohort of 222 patients with DLBCL. The STAT3 activation status determined by these two methods and by STAT3 mRNA levels were then correlated with survival. RESULTS PY-STAT3 was detected in 37% of DLBCL and enriched in ABC-DLBCL cases (P = .03). PY-STAT3 positivity significantly correlated with poor overall survival (OS; P = .01) and event-free survival (EFS; P = .006). Similar observations were made for high levels of STAT3 mRNA. In multivariable analysis, PY-STAT3 status (P = .02), International Prognostic Index (P = .02), and BCL2 expression (P = .046) were independent prognosticators of OS in this cohort. Among the cell-of-origin subgroups, PY-STAT3 was associated with poor EFS among non-germinal center B-cell DLBCL cases only (P = .027). Similarly, the 11-gene STAT3 activation signature correlated with poor survival in the entire DLBCL cohort (OS, P < .001; EFS, P < .001) as well as the ABC-DLBCL subgroup (OS, P = .029; EFS, P = .025). CONCLUSION STAT3 activation correlated with poor survival in patients with DLBCL treated with R-CHOP, especially those with tumors of the ABC-DLBCL subtype.
Collapse
MESH Headings
- Adult
- Aged
- Analysis of Variance
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antigens, CD20/metabolism
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cyclophosphamide/administration & dosage
- Disease-Free Survival
- Doxorubicin/administration & dosage
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Male
- Middle Aged
- Predictive Value of Tests
- Prednisone/administration & dosage
- Prognosis
- Proportional Hazards Models
- RNA, Messenger/metabolism
- Retrospective Studies
- Rituximab
- STAT3 Transcription Factor/drug effects
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Tissue Array Analysis
- Treatment Outcome
- Vincristine/administration & dosage
Collapse
Affiliation(s)
- Xin Huang
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Bin Meng
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Javeed Iqbal
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - B. Belinda Ding
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Anamarija M. Perry
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Wenfeng Cao
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Lynette M. Smith
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Chengfeng Bi
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Chunsun Jiang
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Timothy C. Greiner
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Dennis D. Weisenburger
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Lisa Rimsza
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Andreas Rosenwald
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - German Ott
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Jan Delabie
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Elias Campo
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rita M. Braziel
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Randy D. Gascoyne
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - James R. Cook
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Raymond R. Tubbs
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Elaine S. Jaffe
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - James O. Armitage
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Julie M. Vose
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Louis M. Staudt
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Timothy W. McKeithan
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Wing C. Chan
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - B. Hilda Ye
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Kai Fu
- Xin Huang, Bin Meng, Wenfeng Cao, and Kai Fu, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin; Chengfeng Bi, West China Hospital, West China School of Medicine of Sichuan University, Chengdu, China; Xin Huang, Bin Meng, Javeed Iqbal, Anamarija M. Perry, Wenfeng Cao, Lynette M. Smith, Chengfeng Bi, Chunsun Jiang, Timothy C. Greiner, Dennis D. Weisenburger, James O. Armitage, Julie M. Vose, Timothy W. McKeithan, Wing C. Chan, and Kai Fu, University of Nebraska Medical Center, Omaha, NE; B. Belinda Ding and B. Hilda Ye, Albert Einstein College of Medicine, Bronx, NY; Lisa Rimsza, University of Arizona, Tucson, AZ; Andreas Rosenwald, University of Würzburg, Würzburg, Germany; German Ott and Jan Delabie, Rikshospitalet-Radiumhospitalet Medical Center, University of Oslo, Oslo, Norway; Elias Campo, Hospital Clinic, University of Barcelona, Barcelona, Spain; Rita M. Braziel, Oregon Health and Science University, Portland, OR; Randy D. Gascoyne, British Columbia Cancer Agency, Vancouver, British Columbia, Canada; James R. Cook and Raymond R. Tubbs, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH; Elaine S. Jaffe and Louis M. Staudt, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
39
|
Zhang X, Paun A, Claudio E, Wang H, Siebenlist U. The tumor promoter and NF-κB modulator Bcl-3 regulates splenic B cell development. THE JOURNAL OF IMMUNOLOGY 2013; 191:5984-92. [PMID: 24244019 DOI: 10.4049/jimmunol.1300611] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bcl-3 is an atypical member of the family of IκB proteins. Unlike the classic members, Bcl-3 functions as a nuclear transcriptional cofactor that may, depending on context, promote or suppress genes via association with p50/NF-κB1 or p52/NF-κB2 homodimers. Bcl-3 is also an oncogene, because it is a partner in recurrent translocations in B cell tumors, resulting in deregulated expression. Bcl-3 functions, however, remain poorly understood. We have investigated the role of Bcl-3 in B cells and discovered a previously unknown involvement in the splenic development of these cells. Loss of Bcl-3 in B cells resulted in significantly more marginal zone (MZ) and fewer follicular (FO) B cells. Conversely, transgenic expression of Bcl-3 in B cells generated fewer MZ and more FO B cells. Both Bcl-3(-/-) FO and MZ B cells were more responsive to LPS stimulation compared with their wild-type counterparts, including increased proliferation. By contrast, Bcl-3(-/-) FO B cells were more prone to apoptosis upon BCR stimulation, also limiting their expansion. The data reveal Bcl-3 as a regulator of B cell fate determination, restricting the MZ path and favoring the FO pathway, at least in part, via increased signal-specific survival of the latter, a finding of relevance to its tumorigenic activity.
Collapse
Affiliation(s)
- Xiaoren Zhang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | |
Collapse
|
40
|
Hutchins AP, Diez D, Miranda-Saavedra D. Genomic and computational approaches to dissect the mechanisms of STAT3's universal and cell type-specific functions. JAKSTAT 2013; 2:e25097. [PMID: 24416643 PMCID: PMC3876425 DOI: 10.4161/jkst.25097] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/20/2013] [Accepted: 05/20/2013] [Indexed: 12/11/2022] Open
Abstract
STAT3 is the quintessential pleiotropic transcription factor with many biological roles throughout development as well as in multiple adult tissues. Its functional heterogeneity is encoded in the range of genome-wide binding patterns that specify different regulatory networks in distinct cell types. However, STAT3 does not display remarkable DNA binding preferences that may help correlate specific motifs with individual biological functions or cell types. Therefore, achieving a detailed understanding of the regulatory mechanisms that endow STAT3 (or any other pleiotropic transcription factor) with such a rainbow of functions is not only a central problem in biology but also a fiendishly difficult one. Here we describe key genomic and computational approaches that have shed light into this question, and present the two current models of STAT3 binding (universal and cell type-specific). We also discuss the role that the local epigenetic environment plays in the selection of STAT3 binding sites.
Collapse
Affiliation(s)
- Andrew Paul Hutchins
- Bioinformatics and Genomics Laboratory; World Premier International (WPI) Immunology Frontier Research Center (IFReC); Osaka University; Suita, Osaka Japan
| | - Diego Diez
- Bioinformatics and Genomics Laboratory; World Premier International (WPI) Immunology Frontier Research Center (IFReC); Osaka University; Suita, Osaka Japan
| | - Diego Miranda-Saavedra
- Bioinformatics and Genomics Laboratory; World Premier International (WPI) Immunology Frontier Research Center (IFReC); Osaka University; Suita, Osaka Japan
| |
Collapse
|
41
|
Sharma M, Sharma S, Arora M, Kaul D. Regulation of cellular Cyclin D1 gene by arsenic is mediated through miR-2909. Gene 2013; 522:60-4. [PMID: 23562784 DOI: 10.1016/j.gene.2013.03.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/21/2013] [Accepted: 03/21/2013] [Indexed: 10/27/2022]
Abstract
Arsenic through its ability to regulate genes that link cell cycle control with apoptosis has been widely recognized to play a crucial role in oncogenomics. However, the molecular event by which arsenic affects such genes is far from clear. Here we provide reasonably good evidence to support the view that arsenic exposure to human PBMCs (peripheral blood mononuclear cells) at low concentrations results in the over-expression of miR-2909 within these cells. This over-expressed miR-2909 was found to regulate CCND1 (Cyclin D1) gene expression, within these cells by inducing splice-switching of tumor suppresser CYLD (Cylindromatosis) gene as well as modulation of SP1 (Specificity Protein 1) activity through the repression of KLF4 (Kruppel-like factor4) expression at the translational level. Arsenic dependent regulation of AATF (Apoptosis Antagonizing Transcription factor) and BCL3 (B-cell Lymphoma 3) were also found to be modulated through its capacity to induce miR-2909 expression. Based upon these observations, a novel epigenomic pathway was proposed which may not only be useful in understanding the paradoxical role of arsenic in oncogenomics but also may even be useful in devising various strategies for the treatment/prevention of tumors induced by arsenic.
Collapse
Affiliation(s)
- M Sharma
- Experimental Medicine & Biotechnology Department, Postgraduate Institute of Medical Education & Research, Chandigarh-160012, India
| | | | | | | |
Collapse
|
42
|
Lee HS, Song MK, Choi HS, Shin CY, Lee EI, Ryu JC. Analysis of mRNA expression profiles highlights alterations in modulation of the DNA damage-related genes under butanal exposure in A549 human alveolar epithelial cells. Mol Cell Toxicol 2013. [DOI: 10.1007/s13273-013-0012-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
43
|
Bcl-3 suppresses Tax-induced NF-κB activation through p65 nuclear translocation blockage in HTLV-1-infected cells. Int J Oncol 2012; 42:269-76. [PMID: 23135533 DOI: 10.3892/ijo.2012.1685] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/16/2012] [Indexed: 11/05/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) Tax-induced persistent activation of the NF-κB pathway is perceived as the primary cause of adult T cell leukemia (ATL), an aggressive leukemia caused by HTLV-1. Although elevated oncoprotein Bcl-3 levels are found in many HTLV-1-infected T cell lines and ATL cells, the role of Bcl-3 in the malignant progression caused by HTLV-1 retrovirus remains poorly understood. We confirmed, in the present study, that the Tax-induced NF-κB activation involves the regulation of Bcl-3. Both knockdown and overexpression of Bcl-3 inhibit the Tax-induced NF-κB activation. Similarly, excessive Bcl-3 inhibits the NF-κB/DNA binding activity and significantly decreases Tax-induced p65 nuclear translocation. The present results demonstrate the pleiotropic roles of Bcl-3 in Tax-induced NF-κB activation and indicate that a balance in the aberrant Bcl-3 expression may be established to play an important role in the maintenance of proliferation and inhibition of apoptosis in HTLV-1-infected and ATL cells.
Collapse
|
44
|
Pratt AG, Swan DC, Richardson S, Wilson G, Hilkens CMU, Young DA, Isaacs JD. A CD4 T cell gene signature for early rheumatoid arthritis implicates interleukin 6-mediated STAT3 signalling, particularly in anti-citrullinated peptide antibody-negative disease. Ann Rheum Dis 2012; 71:1374-81. [PMID: 22532634 PMCID: PMC3396452 DOI: 10.1136/annrheumdis-2011-200968] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2012] [Indexed: 11/04/2022]
Abstract
OBJECTIVE We sought clinically relevant predictive biomarkers present in CD4 T-cells, or in serum, that identified those patients with undifferentiated arthritis (UA) who subsequently develop rheumatoid arthritis (RA). METHODS Total RNA was isolated from highly purified peripheral blood CD4 T cells of 173 early arthritis clinic patients. Paired serum samples were also stored. Microarray analysis of RNA samples was performed and differential transcript expression among 111 'training cohort' patients confirmed using real-time quantitative PCR. Machine learning approaches tested the utility of a classification model among an independent validation cohort presenting with UA (62 patients). Cytokine measurements were performed using a highly sensitive electrochemiluminescence detection system. RESULTS A 12-gene transcriptional 'signature' identified RA patients in the training cohort and predicted the subsequent development of RA among UA patients in the validation cohort (sensitivity 68%, specificity 70%). STAT3-inducible genes were over-represented in the signature, particularly in anti-citrullinated peptide antibody-negative disease, providing a risk metric of similar predictive value to the Leiden score in seronegative UA (sensitivity 85%, specificity 75%). Baseline levels of serum interleukin 6 (IL-6) (which signals via STAT3) were highest in anti-citrullinated peptide antibodies-negative RA and distinguished this subgroup from non-RA inflammatory synovitis (corrected p<0.05).Paired serum IL-6 measurements correlated strongly with STAT3-inducible gene expression. CONCLUSION The authors have identified IL-6-mediated STAT-3 signalling in CD4 T cells during the earliest clinical phase of RA, which is most prominent in seronegative disease. While highlighting potential biomarker(s) for early RA, the role of this pathway in disease pathogenesis awaits clarification.
Collapse
Affiliation(s)
- Arthur G Pratt
- Institute of Cellular Medicine (Musculoskeletal Research Group), Newcastle University, Newcastle upon Tyne, UK
- Musculoskeletal Directorate, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Daniel C Swan
- Institute of Cell and Molecular Biology (Bioinformatics Support Unit), Newcastle University, Newcastle upon Tyne, UK
| | - Sarah Richardson
- Institute of Cellular Medicine (Musculoskeletal Research Group), Newcastle University, Newcastle upon Tyne, UK
| | - Gillian Wilson
- Musculoskeletal Directorate, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Catharien M U Hilkens
- Institute of Cellular Medicine (Musculoskeletal Research Group), Newcastle University, Newcastle upon Tyne, UK
| | - David A Young
- Institute of Cellular Medicine (Musculoskeletal Research Group), Newcastle University, Newcastle upon Tyne, UK
| | - John D Isaacs
- Institute of Cellular Medicine (Musculoskeletal Research Group), Newcastle University, Newcastle upon Tyne, UK
- Musculoskeletal Directorate, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
45
|
Bala S, Tang A, Catalano D, Petrasek J, Taha O, Kodys K, Szabo G. Induction of Bcl-3 by acute binge alcohol results in toll-like receptor 4/LPS tolerance. J Leukoc Biol 2012; 92:611-20. [PMID: 22782967 DOI: 10.1189/jlb.0112050] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Acute alcohol binge results in immunosuppression and impaired production of proinflammatory cytokines, including TNF-α. TNF-α production is induced by LPS, a TLR4 ligand, and is tightly regulated at various levels of the signaling cascade, including the NF-κB transcription factor. Here, we hypothesized that acute alcohol induces TLR4/LPS tolerance via Bcl-3, a nuclear protein and member of the NF-κB family. We found that acute alcohol pretreatment resulted in the same attenuating effect as LPS pretreatment on TLR4-induced TNF-α production in human monocytes and murine RAW 264.7 macrophages. Acute alcohol-induced Bcl-3 expression and IP studies revealed increased association of Bcl-3 with NF-κB p50 homodimers in alcohol-treated macrophages and in mice. ChIP assays revealed increased occupancy of Bcl-3 and p50 at the promoter region of TNF-α in alcohol-pretreated cells. To confirm that the Bcl-3-p50 complex regulates transcription/production of TNF-α during acute alcohol exposure, we inhibited Bcl-3 expression using a targeted siRNA. Bcl-3 knockdown prevented the alcohol-induced inhibition of TNF-α mRNA and protein production. In a mouse model of binge alcohol, an increase in Bcl-3 and a concomitant decrease in TNF-α but no change in IL-10 production were found in mice that received alcohol followed by LPS challenge. In summary, our novel data suggest that acute alcohol treatment in vitro and in vivo induces molecular signatures of TLR4/LPS tolerance through the induction of Bcl-3, a negative regulator of TNF-α transcription via its association with NF-κB p50/p50 dimers.
Collapse
Affiliation(s)
- Shashi Bala
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Ahlqvist K, Saamarthy K, Syed Khaja AS, Bjartell A, Massoumi R. Expression of Id proteins is regulated by the Bcl-3 proto-oncogene in prostate cancer. Oncogene 2012; 32:1601-8. [PMID: 22580608 DOI: 10.1038/onc.2012.175] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
B-cell leukemia 3 (Bcl-3) is a member of the inhibitor of κB family, which regulates a wide range of biological processes by functioning as a transcriptional activator or as a repressor of target genes. As high levels of Bcl-3 expression and activation have been detected in different types of human cancer, Bcl-3 has been labeled a proto-oncogene. Our study uncovered a markedly upregulated Bcl-3 expression in human prostate cancer (PCa), where inflammatory cell infiltration was observed. Elevated Bcl-3 expression in PCa was dependent on the proinflammatory cytokine interleukin-6-mediated STAT3 activation. Microarray analyses, using Bcl-3 knockdown in PCa cells, identified the inhibitor of DNA-binding (Id) family of helix-loop-helix proteins as potential Bcl-3-regulated genes. Bcl-3 knockdown reduced the abundance of Id-1 and Id-2 proteins and boosted PCa cells to be more receptive to undergoing apoptosis following treatment with anticancer drug. Our data imply that inactivation of Bcl-3 may lead to sensitization of cancer cells to chemotherapeutic drug-induced apoptosis, thus suggesting a potential therapeutic strategy in PCa treatment.
Collapse
Affiliation(s)
- K Ahlqvist
- Center for Molecular Tumor Pathology, Department of Laboratory Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | | | | | | |
Collapse
|
47
|
Li Y, Kundu P, Seow SW, de Matos CT, Aronsson L, Chin KC, Kärre K, Pettersson S, Greicius G. Gut microbiota accelerate tumor growth via c-jun and STAT3 phosphorylation in APCMin/+ mice. Carcinogenesis 2012; 33:1231-8. [PMID: 22461519 DOI: 10.1093/carcin/bgs137] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is increasingly recognized as a major contributor of human colorectal cancer (CRC). While gut microbiota can trigger inflammation in the intestinal tract, the precise signaling pathways through which host cells respond to inflammatory bacterial stimulation are unclear. Here, we show that gut microbiota enhances intestinal tumor load in the APC(Min/+) mouse model of CRC. Furthermore, systemic anemia occurs coincident with rapid tumor growth, suggesting a role for intestinal barrier damage and erythropoiesis-stimulating mitogens. Short-term stimulation assays of murine colonic tumor cells reveal that lipopolysaccharide, a microbial cell wall component, can accelerate cell growth via a c-Jun/JNK activation pathway. Colonic tumors are also infiltrated by CD11b+ myeloid cells expressing high levels of phospho-STAT3 (p-Tyr705). Our results implicate the role of gut microbiota, through triggering the c-Jun/JNK and STAT3 signaling pathways in combination with anemia, in the acceleration of tumor growth in APC(Min/+) mice.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hayden MS, Ghosh S. NF-κB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev 2012; 26:203-34. [PMID: 22302935 DOI: 10.1101/gad.183434.111] [Citation(s) in RCA: 1348] [Impact Index Per Article: 103.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ability to sense and adjust to the environment is crucial to life. For multicellular organisms, the ability to respond to external changes is essential not only for survival but also for normal development and physiology. Although signaling events can directly modify cellular function, typically signaling acts to alter transcriptional responses to generate both transient and sustained changes. Rapid, but transient, changes in gene expression are mediated by inducible transcription factors such as NF-κB. For the past 25 years, NF-κB has served as a paradigm for inducible transcription factors and has provided numerous insights into how signaling events influence gene expression and physiology. Since its discovery as a regulator of expression of the κ light chain gene in B cells, research on NF-κB continues to yield new insights into fundamental cellular processes. Advances in understanding the mechanisms that regulate NF-κB have been accompanied by progress in elucidating the biological significance of this transcription factor in various physiological processes. NF-κB likely plays the most prominent role in the development and function of the immune system and, not surprisingly, when dysregulated, contributes to the pathophysiology of inflammatory disease. As our appreciation of the fundamental role of inflammation in disease pathogenesis has increased, so too has the importance of NF-κB as a key regulatory molecule gained progressively greater significance. However, despite the tremendous progress that has been made in understanding the regulation of NF-κB, there is much that remains to be understood. In this review, we highlight both the progress that has been made and the fundamental questions that remain unanswered after 25 years of study.
Collapse
Affiliation(s)
- Matthew S Hayden
- Department of Microbiology and Immunology, College of Physicians and Surgeons, New York, New York 10032, USA
| | | |
Collapse
|
49
|
Maldonado V, Melendez-Zajgla J. Role of Bcl-3 in solid tumors. Mol Cancer 2011; 10:152. [PMID: 22195643 PMCID: PMC3258214 DOI: 10.1186/1476-4598-10-152] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 12/23/2011] [Indexed: 12/19/2022] Open
Abstract
Bcl-3 is an established oncogene in hematologic malignancies, such as B-cell chronic lymphocytic leukemias. Nevertheless, recent research has shown that it also participates in progression of diverse solid tumors. The present review summarizes the current knowledge of Bcl3 role in solid tumors progression, including some new insights in its possible molecular mechanisms of action.
Collapse
|
50
|
Human lactoferrin upregulates BCL-3 in the K562 erythroleukemia cell. BIOCHIP JOURNAL 2011. [DOI: 10.1007/s13206-011-5410-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|