1
|
de Mattos K, Scott-Boyer MP, Droit A, Viger RS, Tremblay JJ. Identification of MEF2A, MEF2C, and MEF2D interactomes in basal and Fsk-stimulated mouse MA-10 Leydig cells. Andrology 2025. [PMID: 40277654 DOI: 10.1111/andr.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/06/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Myocyte enhancer factor 2 transcription factors regulate essential transcriptional programs in various cell types. The activity of myocyte enhancer factor 2 factors is modulated through interactions with cofactors, chromatin remodelers, and other regulatory proteins, which are dependent on cell context and physiological state. In steroidogenic Leydig cells, MEF2A, MEF2C, and MEF2D are key regulators of genes involved in steroid hormone synthesis, reproductive function, and oxidative stress defense. However, the specific network of myocyte enhancer factor 2-interacting proteins in Leydig cells remains unknown. OBJECTIVE To identify the interactome of each MEF2 factor present in Leydig cells. MATERIALS AND METHODS TurboID proximity-mediated biotinylation combined with mass spectrometry and bioinformatic analyses were used to identify the protein‒protein interaction networks of MEF2A, MEF2C, and MEF2D in MA-10 Leydig cells under basal and stimulated conditions. RESULTS We identified 109 potential myocyte enhancer factor 2-interacting proteins, including some previously known myocyte enhancer factor 2 partners. The interactome for each myocyte enhancer factor 2 factor is dynamic and exhibits unique and shared interaction networks between basal and stimulated conditions. Further analysis through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment categorized these interactions, revealing involvement in pathways related to cellular metabolism, transcriptional regulation, and steroidogenesis. DISCUSSION AND CONCLUSION These findings suggest that myocyte enhancer factor 2 factors can participate in diverse transcriptional activities, capable of gene activation or repression, depending on different protein‒protein interactions. In addition, the differential interactome for each myocyte enhancer factor 2 factor suggests unique regulatory roles for each factor in modulating Leydig cell function. Overall, this study provides new mechanistic insights into myocyte enhancer factor 2 action in Leydig cells by identifying interacting partners that likely influence their functions.
Collapse
Affiliation(s)
- Karine de Mattos
- Reproduction, Santé de la Mère et de l'enfant, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Canada
| | - Marie-Pier Scott-Boyer
- Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Canada
| | - Arnaud Droit
- Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Robert S Viger
- Reproduction, Santé de la Mère et de l'enfant, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Canada
- Centre for Research in Reproduction, Development and Intergenerational Health, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Jacques J Tremblay
- Reproduction, Santé de la Mère et de l'enfant, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Canada
- Centre for Research in Reproduction, Development and Intergenerational Health, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
2
|
Udemgba C, Pillay B, Shafer S, Alberstadt A, Abers M, Gilliaux O, Chen K, Rae W, Hanitsch L, Von Bernuth H, Neves JF, Raje N, Moens L, van Hagen PM, Bergerson J, Hartog N, Niehues T, Dückers G, Falcone E, Keller M, Hsu A, Meyts I, Holland SM. IRF2BP2 deficiency: An important form of common variable immunodeficiency with inflammation. J Allergy Clin Immunol 2025:S0091-6749(25)00275-1. [PMID: 40090425 DOI: 10.1016/j.jaci.2025.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND IRF2BP2 is a transcription factor that plays an important role in regulating immune pathways, angiogenesis, apoptosis, and cell differentiation. Defects in this gene have been implicated in immunodeficiency. OBJECTIVES To deepen the understanding of the clinical implications of IRF2BP2 variants, we sought to clinically characterize and functionally test 34 individuals with IRF2BP2 variants. METHODS We collected 34 subjects across 18 families with mutations in IRF2BP2. Records were abstracted for clinical phenotypes. Functional testing was performed on PBMCs. NFAT luciferase gene reporter constructs and quantitative cDNA determinations were used to evaluate repressor activity associated with ectopic expression of various IRF2BP2 mutant constructs in Jurkat cells. RESULTS Most subjects had immunodeficiency (91%, n = 30 of 33) with variable gastrointestinal (65%, n = 20 of 31) and inflammatory or autoimmune features (57%, n = 17 of 30), including chronic abdominal pain, colitis, diarrhea, constipation, vitiligo, alopecia, and migratory rashes. There was a reduced frequency of memory B cells with poor immunoglobulin production and reduced calcium flux in response to B-cell receptor stimuli. PBMCs had increased apoptosis in vitro compared to healthy controls. Impaired IRF2BP2 repression of NFAT activation was observed using patient-derived mutant IRF2BP2 constructs compared to wild-type constructs. Similarly, TNF-α transcript levels were higher using patient-derived mutations compared to wild-type IRF2BP2 constructs. CONCLUSIONS IRF2BP2 deficiency causes a complex immunodeficiency including gastrointestinal and inflammatory disorders as well as impaired B-cell maturation. Impaired repression of the NFAT pathway appears to enhance proinflammatory signaling through proinflammatory cytokine expression.
Collapse
Affiliation(s)
- Chioma Udemgba
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Bethany Pillay
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Samantha Shafer
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Angelika Alberstadt
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael Abers
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Olivier Gilliaux
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Karin Chen
- Division of Immunology, Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Wash
| | - William Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Leif Hanitsch
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin Institute of Health at Charité, Berlin, Germany
| | - Horst Von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joao Farela Neves
- Primary Immunodeficiencies Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Nikita Raje
- Division of Allergy, Immunology, Pulmonology, and Sleep Medicine, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri Kansas City, Washington, DC
| | - Leen Moens
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - P Martin van Hagen
- Division of Clinical Immunology, Department of Internal Medicine, Rotterdam, The Netherlands; Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jenna Bergerson
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Nicholas Hartog
- Michigan State University College of Human Medicine Corewell Health and Helen DeVos Children's Hospital, Grand Rapids, Mich
| | - Tim Niehues
- Centre for Child and Adolescent Health, Helios Klinikum Krefeld, affiliated with Rheinisch-Westfälische Technische Hochschule University Aachen, Aachen, Germany
| | - Gregor Dückers
- Centre for Child and Adolescent Health, Helios Klinikum Krefeld, affiliated with Rheinisch-Westfälische Technische Hochschule University Aachen, Aachen, Germany
| | - Emilia Falcone
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael Keller
- Division of Allergy and Immunology, Children's National Hospital, Washington, DC
| | - Amy Hsu
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Steven M Holland
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
3
|
Chen YJN, Shi RC, Xiang YC, Fan L, Tang H, He G, Zhou M, Feng XZ, Tan JD, Huang P, Ye X, Zhao K, Fu WY, Li LL, Bian XT, Chen H, Wang F, Wang T, Zhang CK, Zhou BH, Chen W, Liang TT, Lv JT, Kang X, Shi YX, Kim E, Qin YH, Hettinghouse A, Wang KD, Zhao XL, Yang MY, Tang YZ, Piao HL, Guo L, Liu CJ, Miao HM, Tang KL. Malate initiates a proton-sensing pathway essential for pH regulation of inflammation. Signal Transduct Target Ther 2024; 9:367. [PMID: 39737965 PMCID: PMC11683149 DOI: 10.1038/s41392-024-02076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025] Open
Abstract
Metabolites can double as a signaling modality that initiates physiological adaptations. Metabolism, a chemical language encoding biological information, has been recognized as a powerful principle directing inflammatory responses. Cytosolic pH is a regulator of inflammatory response in macrophages. Here, we found that L-malate exerts anti-inflammatory effect via BiP-IRF2BP2 signaling, which is a sensor of cytosolic pH in macrophages. First, L-malate, a TCA intermediate upregulated in pro-inflammatory macrophages, was identified as a potent anti-inflammatory metabolite through initial screening. Subsequent screening with DARTS and MS led to the isolation of L-malate-BiP binding. Further screening through protein‒protein interaction microarrays identified a L-malate-restrained coupling of BiP with IRF2BP2, a known anti-inflammatory protein. Interestingly, pH reduction, which promotes carboxyl protonation of L-malate, facilitates L-malate and carboxylate analogues such as succinate to bind BiP, and disrupt BiP-IRF2BP2 interaction in a carboxyl-dependent manner. Both L-malate and acidification inhibit BiP-IRF2BP2 interaction, and protect IRF2BP2 from BiP-driven degradation in macrophages. Furthermore, both in vitro and in vivo, BiP-IRF2BP2 signal is required for effects of both L-malate and pH on inflammatory responses. These findings reveal a previously unrecognized, proton/carboxylate dual sensing pathway wherein pH and L-malate regulate inflammatory responses, indicating the role of certain carboxylate metabolites as adaptors in the proton biosensing by interactions between macromolecules.
Collapse
Affiliation(s)
- Yu-Jia-Nan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China.
| | - Rong-Chen Shi
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Yuan-Cai Xiang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Fan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases & Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Hong Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Gang He
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Mei Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin-Zhe Feng
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Jin-Dong Tan
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pan Huang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xiao Ye
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Wen-Yu Fu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Liu-Li Li
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
| | - Xu-Ting Bian
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Feng Wang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Teng Wang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - Chen-Ke Zhang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Bing-Hua Zhou
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Wan Chen
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Tao-Tao Liang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jing-Tong Lv
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, 400038, China
| | - You-Xing Shi
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Ellen Kim
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Yin-Hua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Army Medical University, Chongqing, 400038, China
| | - Aubryanna Hettinghouse
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
| | - Kai-di Wang
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266000, China
| | - Xiang-Li Zhao
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Ming-Yu Yang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yu-Zhen Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lin Guo
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY, 10003, USA.
- Department of Orthopedics and Rehabilitations, Yale University School of Medicine, New Haven, CT, 06519, USA.
| | - Hong-Ming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, 400038, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Kang-Lai Tang
- Department of Orthopedic Surgery/Sports Medicine Center, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
4
|
Chen L, Liu C, Xiang X, Qiu W, Guo K. miR‑155 promotes an inflammatory response in HaCaT cells via the IRF2BP2/KLF2/NF‑κB pathway in psoriasis. Int J Mol Med 2024; 54:91. [PMID: 39219281 PMCID: PMC11374146 DOI: 10.3892/ijmm.2024.5415] [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: 02/01/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Psoriasis is a chronic inflammatory skin condition with numerous causes, including genetic, immunological and infectious factors. The course of psoriasis is long and recurrence is common; pathogenesis is not completely understood. However, there is an association between advancement of psoriasis and aberrant microRNA (miR or miRNA)‑155 expression. Through bioinformatics, the present study aimed to analyze the differentially expressed genes and miRNAs in psoriasis and its biological mechanism and function psoriatic inflammation. First of all, differentially expressed genes (DEGs) and miRNAs (DEMs) in patients with psoriasis were identified using GEO2R interactive web application. A psoriasis inflammatory model was established using lipopolysaccharide (LPS)‑treated HaCaT keratinocytes, which were transfected with miR‑155 mimic or inhibitor. Cell Counting Kit‑8 was used for the assessment of cell viability and proliferation, and changes in the cell cycle were examined using flow cytometry. ELISA and reverse transcription‑quantitative PCR (RT‑qPCR) were used to detect the expression levels of the inflammatory factors IL‑1β and IL‑6. The dual‑luciferase reporter assay was used to verify the targeting association between miR‑155‑5p and IFN regulatory factor 2 binding protein 2 (IRF2BP2). To verify the targeting association of miR‑155 and the IRF2BP2/kruppel‑like factor 2 (KLF2)/NF‑κB signaling pathway, expression levels of IRF2BP2, KLF2 and p65 were identified by RT‑qPCR and western blotting. IRF2BP2 levels were also confirmed by immunofluorescence, in conjunction with bioinformatics database analysis. Overexpression of miR‑155 inhibited proliferation of HaCaT cells and increased the number of cells in S phase and decreasing number of cells in G1 and G2 phase. In the LPS‑induced inflammatory state, miR‑155 overexpression heightened the inflammatory response of HaCaT cells while inhibition of miR‑155 lessened it. Suppression of inflammatory cytokine expression by miR‑155‑5p inhibitor was reversed by knockdown of IRF2BP2. miR‑155 was shown to interact with IRF2BP2 to negatively regulate its expression, leading to decreased KLF2 expression and increased p65 expression and secretion of inflammatory factors, intensifying the inflammatory response of HaCaT cells. Therefore, miR‑155 may contribute to development of psoriasis by inducing tissue and cell damage by increasing the inflammatory response of HaCaT cells via the IRF2BP2/KLF2/NF‑κB pathway. In conclusion, the results of the present study offer novel perspectives on the role of miR‑155 in the onset and progression of psoriasis.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Chang Liu
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Xuesong Xiang
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Wenhong Qiu
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Kaiwen Guo
- Department of Pathogenic Biology, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei 430065, P.R. China
| |
Collapse
|
5
|
Chen Y, Zhuo R, Sun L, Tao Y, Li G, Zhu F, Xu Y, Wang J, Li Z, Yu J, Yin H, Wu D, Li X, Fang F, Xie Y, Hu Y, Wang H, Yang C, Shi L, Wang X, Zhang Z, Pan J. Super-enhancer-driven IRF2BP2 enhances ALK activity and promotes neuroblastoma cell proliferation. Neuro Oncol 2024; 26:1878-1894. [PMID: 38864832 PMCID: PMC11449008 DOI: 10.1093/neuonc/noae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Super-enhancers (SEs) typically govern the expression of critical oncogenes and play a fundamental role in the initiation and progression of cancer. Focusing on genes that are abnormally regulated by SE in cancer may be a new strategy for understanding pathogenesis. In the context of this investigation, we have identified a previously unreported SE-driven gene IRF2BP2 in neuroblastoma (NB). METHODS The expression and prognostic value of IRF2BP2 were detected in public databases and clinical samples. The effect of IRF2BP2 on NB cell growth and apoptosis was evaluated through in vivo and in vitro functional loss experiments. The molecular mechanism of IRF2BP2 was investigated by the study of chromatin regulatory regions and transcriptome sequencing. RESULTS The sustained high expression of IRF2BP2 results from the activation of a novel SE established by NB master transcription factors MYCN, MEIS2, and HAND2, and they form a new complex that regulates the gene network associated with the proliferation of NB cell populations. We also observed a significant enrichment of the AP-1 family at the binding sites of IRF2BP2. Remarkably, within NB cells, AP-1 plays a pivotal role in shaping the chromatin accessibility landscape, thereby exposing the binding site for IRF2BP2. This orchestrated action enables AP-1 and IRF2BP2 to collaboratively stimulate the expression of the NB susceptibility gene ALK, thereby upholding the highly proliferative phenotype characteristic of NB. CONCLUSIONS Our findings indicate that SE-driven IRF2BP2 can bind to AP-1 to maintain the survival of tumor cells via regulating chromatin accessibility of the NB susceptibility gene ALK.
Collapse
Affiliation(s)
- Yanling Chen
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Ran Zhuo
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Lichao Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
| | - Yanfang Tao
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Gen Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Frank Zhu
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yunyun Xu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Jianwei Wang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Zhiheng Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Juanjuan Yu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Hongli Yin
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Di Wu
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Fang Fang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Yi Xie
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Yizhou Hu
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Hairong Wang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Chun Yang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Lei Shi
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodong Wang
- Department of Orthopedics, Children’s Hospital of Soochow University, Suzhou, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Panir K, Schjenken JE, Breen J, Chan HY, Greaves E, Robertson SA, Hull ML. RNA sequencing reveals molecular mechanisms of endometriosis lesion development in mice. Dis Model Mech 2024; 17:dmm050566. [PMID: 39385609 PMCID: PMC11524442 DOI: 10.1242/dmm.050566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Understanding of molecular mechanisms contributing to the pathophysiology of endometriosis, and upstream drivers of lesion formation, remains limited. Using a C57Bl/6 mouse model in which decidualized endometrial tissue is injected subcutaneously in the abdomen of recipient mice, we generated a comprehensive profile of gene expression in decidualized endometrial tissue (n=4), and in endometriosis-like lesions at Day 7 (n=4) and Day 14 (n=4) of formation. High-throughput mRNA sequencing allowed identification of genes and pathways involved in the initiation and progression of endometriosis-like lesions. We observed distinct patterns of gene expression with substantial differences between the lesions and the decidualized endometrium that remained stable across the two lesion timepoints, and showed similarity to transcriptional changes implicated in human endometriosis lesion formation. Pathway enrichment analysis revealed several immune and inflammatory response-associated canonical pathways, multiple potential upstream regulators, and involvement of genes not previously implicated in endometriosis pathogenesis, including IRF2BP2 and ZBTB10, suggesting novel roles in disease progression. Collectively, the provided data will be a useful resource to inform research on the molecular mechanisms contributing to endometriosis-like lesion development in this mouse model.
Collapse
Affiliation(s)
- Kavita Panir
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - John E. Schjenken
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - James Breen
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- South Australian Genomics Centre (SAGC), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Computational and Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Hon Yeung Chan
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Erin Greaves
- Centre for Early Life, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Sarah A. Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - M. Louise Hull
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA 5006, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Department of Obstetrics and Gynaecology, Women's and Children's Hospital, Adelaide, SA 5006, Australia
| |
Collapse
|
7
|
Anim M, Sogkas G, Camacho-Ordonez N, Schmidt G, Elsayed A, Proietti M, Witte T, Grimbacher B, Atschekzei F. Novel hypermorphic variants in IRF2BP2 identified in patients with common variable immunodeficiency and autoimmunity. Clin Immunol 2024; 266:110326. [PMID: 39059757 DOI: 10.1016/j.clim.2024.110326] [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: 06/03/2024] [Revised: 07/12/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
The interferon regulatory factor 2 binding protein 2 (IRF2BP2) is a transcriptional regulator, functioning a transcriptional corepressor by interacting with the interferon regulatory factor-2. The ubiquitous expression of IRF2BP2 by diverse cell types and tissues suggests its potential involvement in different cell signalling pathways. Variants inIRF2BP2have been recently identified to cause familial common variable immunodeficiency (CVID) characterized by immune dysregulation. This study investigated three rare novel variants inIRF2BP2, identified in patients with primary antibody deficiency and autoimmunity by whole exome-sequencing (WES). Following transient overexpression of EGFP-fused mutants in HEK293 cells and transfection in Jurkat cell lines, we used fluorescence microscopy, real-time PCR and Western blotting to analyze their effects on IRF2BP2 expression, subcellular localization, nuclear translocation of IRF2, and the transcriptional activation of NFκB1(p50). We found altered IRF2BP2 mRNA and protein expression levels in the mutants compared to the wild type after IRF2BP2 overexpression. In confocal fluorescence microscopy, variants in the C-terminal RING finger domain showed an irregular aggregate formation and distribution instead of the expected nuclear localization compared to the variants in the N-terminal zinc finger domain and their wildtype counterpart. Immunoblotting revealed an impaired IRF2 and NFκB1 (p50) nuclear localization in the mutants compared to the IRF2BP2 wildtype counterpart. LPS stimulation reduced IRF2BP2 mRNA expression in the variants compared to the wild type. Our findings significantly contribute to understanding the clinical significance of IRF2BP2 mutations in the pathogenesis of immunodeficiency and immune dysregulation. We observed impairment of the nuclear translocation of IRF2 and NFκB1 (p50) due to the upregulation of IRF2BP2, potentially affecting specific gene expressions involved in immune regulation.
Collapse
Affiliation(s)
- Manfred Anim
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Hannover Biomedical Research School (HBRS), Hannover Medical School, Hanover, Germany
| | - Georgios Sogkas
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Hanover, Germany
| | - Nadezhda Camacho-Ordonez
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany; Clinic of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany
| | - Gunnar Schmidt
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Abdulwahab Elsayed
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Michele Proietti
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Hanover, Germany.; Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Hanover, Germany
| | - Bodo Grimbacher
- RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Hanover, Germany.; Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Clinic of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany; DZIF - German Center for Infection Research, Satellite Center Freiburg, Germany
| | - Faranaz Atschekzei
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Hanover, Germany..
| |
Collapse
|
8
|
Pfeuffer L, Siegert V, Frede J, Rieger L, Trozzo R, de Andrade Krätzig N, Ring S, Sarhadi S, Beck N, Niedermeier S, Abril-Gil M, Elbahloul M, Remke M, Steiger K, Eichner R, Jellusova J, Rad R, Bassermann F, Winter C, Ruland J, Buchner M. B-cell intrinsic RANK signaling cooperates with TCL1 to induce lineage-dependent B-cell transformation. Blood Cancer J 2024; 14:151. [PMID: 39198400 PMCID: PMC11358282 DOI: 10.1038/s41408-024-01123-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
B-cell malignancies, such as chronic lymphocytic leukemia (CLL) and multiple myeloma (MM), remain incurable, with MM particularly prone to relapse. Our study introduces a novel mouse model with active RANK signaling and the TCL1 oncogene, displaying both CLL and MM phenotypes. In younger mice, TCL1 and RANK expression expands CLL-like B1-lymphocytes, while MM originates from B2-cells, becoming predominant in later stages and leading to severe disease progression and mortality. The induced MM mimics human disease, exhibiting features like clonal plasma cell expansion, paraproteinemia, anemia, and kidney and bone failure, as well as critical immunosurveillance strategies that promote a tumor-supportive microenvironment. This research elucidates the differential impacts of RANK activation in B1- and B2-cells and underscores the distinct roles of single versus combined oncogenes in B-cell malignancies. We also demonstrate that human MM cells express RANK and that inhibiting RANK signaling can reduce MM progression in a xenotransplantation model. Our study provides a rationale for further investigating the effects of RANK signaling in B-cell transformation and the shaping of a tumor-promoting microenvironment.
Collapse
Affiliation(s)
- Lisa Pfeuffer
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Viola Siegert
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Julia Frede
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Leonie Rieger
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Medicine III, TUM School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Riccardo Trozzo
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Health, Technical University of Munich, 81675, Munich, Germany
| | - Niklas de Andrade Krätzig
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Health, Technical University of Munich, 81675, Munich, Germany
| | - Sandra Ring
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Shamim Sarhadi
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Nicole Beck
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Stefan Niedermeier
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Mar Abril-Gil
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Mohamed Elbahloul
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Medicine III, TUM School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marianne Remke
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Katja Steiger
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Ruth Eichner
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Medicine III, TUM School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Julia Jellusova
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Roland Rad
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and Health, Technical University of Munich, 81675, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Florian Bassermann
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- Department of Medicine III, TUM School of Medicine and Health, Technical University Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Bavarian Center for Cancer Research (BZKF), Munich, Germany
| | - Christof Winter
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Munich, 81675, Munich, Germany
| | - Maike Buchner
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany.
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany.
| |
Collapse
|
9
|
Fischer S, Weber LM, Stielow B, Frech M, Simon C, Geller M, Könnecke J, Finkernagel F, Forné I, Nist A, Bauer UM, Stiewe T, Neubauer A, Liefke R. IRF2BP2 counteracts the ATF7/JDP2 AP-1 heterodimer to prevent inflammatory overactivation in acute myeloid leukemia (AML) cells. Nucleic Acids Res 2024; 52:7590-7609. [PMID: 38801077 PMCID: PMC11260449 DOI: 10.1093/nar/gkae437] [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: 07/01/2023] [Revised: 04/16/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by abnormal proliferation and accumulation of immature myeloid cells in the bone marrow. Inflammation plays a crucial role in AML progression, but excessive activation of cell-intrinsic inflammatory pathways can also trigger cell death. IRF2BP2 is a chromatin regulator implicated in AML pathogenesis, although its precise role in this disease is not fully understood. In this study, we demonstrate that IRF2BP2 interacts with the AP-1 heterodimer ATF7/JDP2, which is involved in activating inflammatory pathways in AML cells. We show that IRF2BP2 is recruited by the ATF7/JDP2 dimer to chromatin and counteracts its gene-activating function. Loss of IRF2BP2 leads to overactivation of inflammatory pathways, resulting in strongly reduced proliferation. Our research indicates that a precise equilibrium between activating and repressive transcriptional mechanisms creates a pro-oncogenic inflammatory environment in AML cells. The ATF7/JDP2-IRF2BP2 regulatory axis is likely a key regulator of this process and may, therefore, represent a promising therapeutic vulnerability for AML. Thus, our study provides new insights into the molecular mechanisms underlying AML pathogenesis and identifies a potential therapeutic target for AML treatment.
Collapse
Affiliation(s)
- Sabrina Fischer
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Lisa Marie Weber
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Bastian Stielow
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Miriam Frech
- Department of Hematology, Oncology, and Immunology, University Hospital Giessen and Marburg, Marburg 35043, Germany
| | - Clara Simon
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Merle Geller
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Julie Könnecke
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Florian Finkernagel
- Translational Oncology Group, Center for Tumor Biology and Immunology (ZTI), Philipps University of Marburg, Marburg 35043, Germany
| | - Ignasi Forné
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Martinsried 82152, Germany
| | - Andrea Nist
- Genomics Core Facility, Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University of Marburg, Marburg 35043, Germany
| | - Uta-Maria Bauer
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University of Marburg, Marburg 35043, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology, and Immunology, University Hospital Giessen and Marburg, Marburg 35043, Germany
| | - Robert Liefke
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University of Marburg, Marburg 35043, Germany
- Department of Hematology, Oncology, and Immunology, University Hospital Giessen and Marburg, Marburg 35043, Germany
| |
Collapse
|
10
|
Jiang M, Wang X, Yu M, Jiang S, Hong M, Zhou Y, Li F, Liu H, Zhang Z. Report of IRF2BP1 as a novel partner of RARA in variant acute promyelocytic leukemia. Am J Hematol 2024; 99:1005-1007. [PMID: 38410879 DOI: 10.1002/ajh.27272] [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: 12/04/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 02/28/2024]
Abstract
IRF2BP1 breaked in the middle of exon 1 at the c.322 position and fused with RARA intron 2 which is located at 3717 bp upstream of its exon 3. The fusion produced a new intron by forming a paired splicing donor GT at 9 bp downstream of RARA breakpoint and acceptor AG at the 5' end of RARA exon 3. The IRF2BP1::RARA fusion gene leads a fusion transcript involving IRF2BP1 exon 1 and RARA exon 3, linked by a 9-bp fragment derived from RARA intron 2. The patient with IRF2BP1::RARA has same clinical features of APL.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Clinical Laboratory, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuemei Wang
- Department of Blood Transfusion, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Min Yu
- Department of Hematology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shuling Jiang
- Nursing School of Nanchang University, Nanchang, China
| | - Miao Hong
- Department of Blood Transfusion, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuru Zhou
- Department of Blood Transfusion, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fei Li
- Department of Hematology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongxing Liu
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Zhanglin Zhang
- Department of Blood Transfusion, Institute of transfusion, Jiangxi Key Laboratory of transfusion, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Pang X, Li TJ, Shi RJ, Wan ZX, Tang YY, Tang YL, Liang XH. IRF2BP2 drives lymphatic metastasis in OSCC cells by elevating mitochondrial fission-dependent fatty acid oxidation. Mol Carcinog 2024; 63:45-60. [PMID: 37737489 DOI: 10.1002/mc.23635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
Lymph node metastasis (LNM) is a major determinant for the poor outcome of oral squamous cell carcinoma (OSCC). Interferon regulatory factor 2 binding protein 2 (IRF2BP2) has been reported to modulate the development and progression of several types of cancers, while its role in OSCC with LNM has not been reported yet. The expression of IRF2BP2 and its association with LNM were evaluated by immunohistochemistry and qualitative reverse transcription polymerase chain reaction in clinically collected OSCC tissues. Then, loss-of-function and rescue assays were conducted to identify the role of IRF2BP2-mediated fatty acid oxidation (FAO) in the invasion, lymphoinvasion, and epithelial-mesenchymal transition (EMT) in OSCC cells. Importantly, confocal microscope, transmission electron microscope, immunofluorescence, and Western blot were applied to identify the involvement of mitochondrial fission in IRF2BP2-regulated FAO. Lastly, the in vivo models were established to evaluate the role of IRF2BP2 in OSCC. IRF2BP2 overexpression has been associated with LNM in OSCC, whose knockdown inhibited invasion, lymphoinvasion, and EMT of OSCC cells, as well as retarded FAO rate with CPT1A downregulation. And CPT1A overexpression rescued invasion, lymphoinvasion, and induced EMT in IRF2BP2-silenced OSCC cells. Mechanically, IRF2BP2 accelerated mitochondrial fission by contributing to Drp1 S616 phosphorylation and mitochondrial localization, resulting in the upregulation of CPT1A. In addition, IRF2BP2 knockdown significantly inhibited tumor growth and LNM in vivo. The highly expressed IRF2BP2 may induce the phosphorylation and mitochondrial translocation of Drp1 to activate mitochondrial fission, which upregulated CPT1A expression and FAO rate, resulting in LNM in OSCC. This highlighted a potential therapeutic vulnerability for the treatment of LNM+ OSCC via targeting IRF2BP2-FAO.
Collapse
Affiliation(s)
- Xin Pang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tian-Jiao Li
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rong-Jia Shi
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zi-Xin Wan
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Yang Tang
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Ling Tang
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin-Hua Liang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Panagopoulos I, Gorunova L, Lobmaier I, Heim S. Fusion of the Genes for Interferon Regulatory Factor 2 Binding Protein 2 ( IRF2BP2) and Caudal Type Homeobox 1 ( CDX1) in a Chondrogenic Tumor. In Vivo 2023; 37:2459-2463. [PMID: 37905608 PMCID: PMC10621452 DOI: 10.21873/invivo.13352] [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: 06/27/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND/AIM Chondrogenic tumors are benign, intermediate or malignant neoplasms showing cartilaginous differentiation. In 2012, we reported a mesenchymal chondrosarcoma carrying a t(1;5)(q42;q32) leading to an IRF2BP2::CDX1 fusion gene. Here, we report a second chondrogenic tumor carrying an IRF2BP2::CDX1 chimera. CASE REPORT Radiological examination of a 41 years old woman showed an osteolytic lesion in the os pubis with a large soft tissue component. Examination of a core needle biopsy led to the diagnosis chondromyxoid fibroma, and the patient was treated with curettage. Microscopic examination of the specimen showed a tumor tissue in which a pink-bluish background matrix was studded with small spindled to stellate cells without atypia, fitting well the chondromyxoid fibroma diagnosis. Focally, a more cartilage-like appearance was observed with cells lying in lacunae and areas with calcification. G-banding analysis of short-term cultured tumor cells yielded the karyotype 46,XX,der(1)inv(1)(p33~34q42) add(1)(p32)?ins(1;?)(q42;?),del(5)(q31),der(5)t(1;5)(q42;q35)[12]/46,XX[3]. RT-PCR together with Sanger sequencing showed the presence of two IRF2BP2::CDX1 chimeric transcripts in which exon 1 of the IRF2BP2 reference sequence NM_182972.3 or NM_001077397.1 was fused to exon 2 of CDX1. Both chimeras were predicted to code for proteins containing the zinc finger domain of IRF2BP2 and homeobox domain of CDX1. CONCLUSION IRF2BP2::CDX1 chimera is recurrent in chondrogenic tumors. The data are still too sparse to conclude whether it is a hallmark of benign or malignant tumors.
Collapse
Affiliation(s)
- Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway;
| | - Ludmila Gorunova
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ingvild Lobmaier
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
García-Aznar JM, Maneiro Pampín E, García Ramos M, Acuña Pérez MJ, Paz Gandiaga N, Minguell Domingo L, Calavia O, Soler-Palacin P, Colobran R, Novoa Bolívar EM, Ocejo Vinyals JG. Novel frameshift variants expand the map of the genetic defects in IRF2BP2. Front Immunol 2023; 14:1279171. [PMID: 37876937 PMCID: PMC10593445 DOI: 10.3389/fimmu.2023.1279171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Background At present, the knowledge about disease-causing mutations in IRF2BP2 is very limited because only a few patients affected by this condition have been reported. As previous studies have described, the haploinsufficiency of this interferon transcriptional corepressors leads to the development of CVID. Very recently, a more accurate phenotype produced by truncating variants in this gene has been defined, manifesting CVID with gastrointestinal inflammatory symptoms and autoimmune manifestations. Methods We analyzed 5 index cases with suspected primary immunodeficiency by high throughput sequencing. They were submitted for a genetic test with a panel of genes associated with immune system diseases, including IRF2BP2. The screening of SNVs, indels and CNVs fulfilling the criteria with very low allelic frequency and high protein impact, revealed five novel variants in IRF2BP2. In addition, we isolated both wild-type and mutated allele of the cDNA from one of the families. Results In this study, we report five novel loss-of-function (LoF) mutations in IRF2BP2 that likely cause primary immunodeficiency, with CVID as more frequent phenotype, variable expression of inflammatory gastrointestinal features, and one patient with predisposition of viral infection. All identified variants were frameshift changes, and one of them was a large deletion located on chromosome 1q42, which includes the whole sequence of IRF2BP2, among other genes. Both de novo and dominant modes of inheritance were observed in the families here presented, as well as incomplete penetrance. Conclusions We describe novel variants in a delimited low-complex region, which may be considered a hotspot in IRF2BP2. Moreover, this is the first time that a large CNV in IRF2BP2 has been reported to cause CVID. The distinct mechanisms than LoF in IRF2BP2 could cause different phenotype compared with the mainly described. Further investigations are necessary to comprehend the regulatory mechanisms of IRF2BP2, which could be under variable expression of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Nerea Paz Gandiaga
- Genetics Division, Universitary Hospital Marqués de Valdecilla, Santander, Canatabria, Spain
| | | | - Olga Calavia
- Pediatrics Division, Hospital Joan XXIII, Tarragona, Catalonia, Spain
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Children’s Hospital, Barcelona, Catalonia, Spain
- Infection and Immunity in Pediatric Patients Research Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
| | - Roger Colobran
- Immunology Division, Vall d’Hebron University Hospital (HUVH), Barcelona, Catalonia, Spain
- Translational Immunology Research Group, Vall d’Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
- Department of Clinical and Molecular Genetics, Vall d’Hebron University Hospital (HUVH), Barcelona, Catalonia, Spain
- Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona (UAB), Barcelona, Catalonia, Spain
| | | | | |
Collapse
|
14
|
Pan Y, Shang G, Li J, Zhang Y, Liu J, Ji Y, Ding J, Wang X. Case Report: A novel IRF2BP2 mutation in an IEI patient with recurrent infections and autoimmune disorders. Front Immunol 2023; 14:967345. [PMID: 37350971 PMCID: PMC10282741 DOI: 10.3389/fimmu.2023.967345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 04/20/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Inborn errors of immunity (IEI) are a heterogeneous group of disorders characterized by increased risk of infections, autoimmunity, autoinflammatory diseases, malignancy and allergy. Next-generation sequencing has revolutionized the identification of genetic background of these patients and assists in diagnosis and treatment. In this study, we identified a probable unique monogenic cause of IEI, and evaluated the immunological methods and pathogenic detections. Methods A family with a member with a clinical diagnosis of IEI was screened by whole genomic sequencing (WGS). Demographic data, clinical manifestations, medical history, physical examination, laboratory findings and imaging features of the patient were extracted from medical records. Comprehensive immune monitoring methods include a complete blood count with differential, serum levels of cytokines and autoantibodies, T-cell and B-cell subsets analysis and measurement of serum immunoglobulins. In addition, metagenomic sequencing (mNGS) of blood, cerebrospinal fluid and biopsy from small intestine were used to detect potential pathogens. Results The patient manifested with recurrent infections and autoimmune disorders, who was eventually diagnosed with IEI. Repetitive mNGS tests of blood, cerebrospinal fluid and biopsy from small intestine didn't detect pathogenic microorganism. Immunological tests showed a slightly decreased level of IgG than normal, elevated levels of tumor necrosis factor and interleukin-6. Lymphocyte flow cytometry showed elevated total B cells and natural killer cells, decreased total T cells and B-cell plasmablasts. WGS of the patient identified a novel heterozygous mutation in IRF2BP2 (c.439_450dup p. Thr147_Pro150dup), which was also confirmed in his father. The mutation was classified as variant of uncertain significance (VUS) according to the American College of Medical Genetics and Genomics guidelines. Conclusion We identified a novel IRF2BP2 mutation in a family with a member diagnosed with IEI. Immune monitoring and WGS as auxiliary tests are helpful in identifying genetic defects and assisting diagnosis in patients with clinically highly suspected immune abnormalities and deficiencies in inflammation regulation. In addition, mNGS techniques allow a more comprehensive assessment of the pathogenic characteristics of these patients. This report further validates the association of IRF2BP2 deficiency and IEI, and expands IEI phenotypes.
Collapse
Affiliation(s)
- Yiwen Pan
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoguo Shang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Li
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuwen Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianying Liu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- The State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Sutcliffe M, Nair N, Oliver J, Morgan AW, Isaacs JD, Wilson AG, Verstappen SMM, Viatte S, Hyrich KL, Morris AP, Barton A, Plant D. Pre-defined gene co-expression modules in rheumatoid arthritis transition towards molecular health following anti-TNF therapy. Rheumatology (Oxford) 2022; 61:4935-4944. [PMID: 35377444 PMCID: PMC9707314 DOI: 10.1093/rheumatology/keac204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/31/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND No reliable biomarkers to predict response to TNF inhibitors (TNFi) in RA patients currently exist. The aims of this study were to replicate changes in gene co-expression modules that were previously reported in response to TNFi therapy in RA; to test if changes in module expression are specific to TNFi therapy; and to determine whether module expression transitions towards a disease-free state in responding patients. METHOD Published transcriptomic data from the whole blood of disease-free controls (n = 10) and RA patients, treated with the TNFi adalimumab (n = 70) or methotrexate (n = 85), were studied. Treatment response was assessed using the EULAR response criteria following 3 or 6 months of treatment. Change in transcript expression between pre- and post-treatment was recorded for previously defined modules. Linear mixed models tested whether modular expression after treatment transitioned towards a disease-free state. RESULTS For 25 of the 27 modules, change in expression between pre- and post-treatment in the adalimumab cohort replicated published findings. Of these 25 modules, six transitioned towards a disease-free state by 3 months (P < 0.05), irrespective of clinical response. One module (M3.2), related to inflammation and TNF biology, significantly correlated with response to adalimumab. Similar patterns of modular expression, with reduced magnitude, were observed in the methotrexate cohort. CONCLUSION This study provides independent validation of changes in module expression in response to therapy in RA. However, these effects are not specific to TNFi. Further studies are required to determine whether specific modules could assist molecular classification of therapeutic response.
Collapse
Affiliation(s)
- Megan Sutcliffe
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester
| | - Nisha Nair
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester
| | - James Oliver
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester
| | - Ann W Morgan
- School of Medicine, University of Leeds & NIHR Leeds Biomedical Research Centre and NIHR In Vitro Diagnostic Co-operative, Leeds Teaching Hospitals NHS Trust, University of Leeds, Leeds
| | - John D Isaacs
- Translational & Clinical Research Institution, Newcastle University & Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle University, Newcastle upon Tyne, UK
| | - Anthony G Wilson
- School of Medicine & Medical Science, Conway Institute, University College Dublin, Bellfield, Dublin 4, Ireland
| | - Suzanne M M Verstappen
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester.,Versus Arthritis Centre for Epidemiology, Centre for Musculoskeletal Research
| | - Sebastien Viatte
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester.,Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Kimme L Hyrich
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester.,Versus Arthritis Centre for Epidemiology, Centre for Musculoskeletal Research
| | - Andrew P Morris
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester
| | - Anne Barton
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester
| | - Darren Plant
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal Sciences, The University of Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester
| |
Collapse
|
16
|
Pouliquen DL, Malloci M, Boissard A, Henry C, Guette C. Proteomes of Residual Tumors in Curcumin-Treated Rats Reveal Changes in Microenvironment/Malignant Cell Crosstalk in a Highly Invasive Model of Mesothelioma. Int J Mol Sci 2022; 23:ijms232213732. [PMID: 36430209 PMCID: PMC9691155 DOI: 10.3390/ijms232213732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Curcumin exhibits both immunomodulatory properties and anticarcinogenic effects which have been investigated in different experimental tumor models and cancer types. Its interactions with multiple signaling pathways have been documented through proteomic studies on malignant cells in culture; however, in vivo approaches are scarce. In this study, we used a rat model of highly invasive peritoneal mesothelioma to analyze the residual tumor proteomes of curcumin-treated rats in comparison with untreated tumor-bearing rats (G1) and provide insights into the modifications in the tumor microenvironment/malignant cell crosstalk. The cross-comparing analyses of the histological sections of residual tumors from two groups of rats given curcumin twice on days 21 and 26 after the tumor challenge (G2) or four times on days 7, 9, 11 and 14 (G3), in comparison with G1, identified a common increase in caveolin-1 which linked with significant abundance changes affecting 115 other proteins. The comparison of G3 vs. G2 revealed additional features for 65 main proteins, including an increase in histidine-rich glycoprotein and highly significant abundance changes for 22 other proteins regulating the tumor microenvironment, linked with the presence of numerous activated T cells. These results highlight new features in the multiple actions of curcumin on tumor microenvironment components and cancer cell invasiveness.
Collapse
Affiliation(s)
- Daniel L. Pouliquen
- Université d’Angers, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
- Correspondence: ; Tel.: +33-2-41352854
| | - Marine Malloci
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, F-44000 Nantes, France
| | - Alice Boissard
- Université d’Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Cécile Henry
- Université d’Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Catherine Guette
- Université d’Angers, ICO, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| |
Collapse
|
17
|
Davies CJ, Fan Z, Morgado KP, Liu Y, Regouski M, Meng Q, Thomas AJ, Yun SI, Song BH, Frank JC, Perisse IV, Van Wettere A, Lee YM, Polejaeva IA. Development and characterization of type I interferon receptor knockout sheep: A model for viral immunology and reproductive signaling. Front Genet 2022; 13:986316. [PMID: 36246651 PMCID: PMC9556006 DOI: 10.3389/fgene.2022.986316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/17/2022] [Indexed: 11/21/2022] Open
Abstract
Type I interferons (IFNs) initiate immune responses to viral infections. Their effects are mediated by the type I IFN receptor, IFNAR, comprised of two subunits: IFNAR1 and IFNAR2. One or both chains of the sheep IFNAR were disrupted in fetal fibroblast lines using CRISPR/Cas9 and 12 lambs were produced by somatic cell nuclear transfer (SCNT). Quantitative reverse transcription-polymerase chain reaction for IFN-stimulated gene expression showed that IFNAR deficient sheep fail to respond to IFN-alpha. Furthermore, fibroblast cells from an IFNAR2−/− fetus supported significantly higher levels of Zika virus (ZIKV) replication than wild-type fetal fibroblast cells. Although many lambs have died from SCNT related problems or infections, one fertile IFNAR2−/− ram lived to over 4 years of age, remained healthy, and produced more than 80 offspring. Interestingly, ZIKV infection studies failed to demonstrate a high level of susceptibility. Presumably, these sheep compensated for a lack of type I IFN signaling using the type II, IFN-gamma and type III, IFN-lambda pathways. These sheep constitute a unique model for studying the pathogenesis of viral infection. Historical data supports the concept that ruminants utilize a novel type I IFN, IFN-tau, for pregnancy recognition. Consequently, IFNAR deficient ewes are likely to be infertile, making IFNAR knockout sheep a valuable model for studying pregnancy recognition. A breeding herd of 32 IFNAR2+/− ewes, which are fertile, has been developed for production of IFNAR2−/− sheep for both infection and reproduction studies.
Collapse
Affiliation(s)
- Christopher J. Davies
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
- Center for Integrated BioSystems, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
- *Correspondence: Christopher J. Davies, ; Irina A. Polejaeva,
| | - Zhiqiang Fan
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Kira P. Morgado
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
- Center for Integrated BioSystems, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Ying Liu
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Misha Regouski
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Qinggang Meng
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Aaron J. Thomas
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
- Center for Integrated BioSystems, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Sang-Im Yun
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Byung-Hak Song
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Jordan C. Frank
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Iuri V. Perisse
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Arnaud Van Wettere
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Young-Min Lee
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
| | - Irina A. Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, United States
- *Correspondence: Christopher J. Davies, ; Irina A. Polejaeva,
| |
Collapse
|