1
|
Tajeri S, Shiels B, Langsley G, Nijhof AM. Upregulation of haematopoetic cell kinase (Hck) activity by a secreted parasite effector protein (Ta9) drives proliferation of Theileria annulata-transformed leukocytes. Microb Pathog 2025; 199:107252. [PMID: 39730099 DOI: 10.1016/j.micpath.2024.107252] [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: 08/07/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 12/29/2024]
Abstract
Reversible transformation of bovine leukocytes by the intracellular parasites Theileria annulata and Theileria parva is central to pathogenesis of the diseases they cause, tropical theileriosis and East Coast Fever, respectively. Parasite-dependent constitutive activation of major host transcription factors such as AP-1 (Activating Protein 1) and NF-κB (Nuclear Factor-Kappa B) sustains the transformed state. Although parasite interaction with host cell signaling pathways upstream of AP-1 have been studied, the precise contribution of Theileria encoded factors capable of modulating AP-1 transcriptional activity, and other infection-altered signaling pathways is not fully understood. We previously showed that the Ta9 protein from T. annulata (TA15705) is secreted into the host cell cytoplasm and contributes to infection-induced AP-1 transcriptional activity. The current study employed RNA-seq to investigate the ability of ectopically expressed Ta9 to modulate the gene transcription profile of a bovine macrophage cell line, BoMac. RNA-seq identified 560 (400 upregulated and 160 downregulated) differentially expressed genes. KEGG analysis predicted a high number of upregulated genes associated with carcinogenesis such as CCND1, CDKN1A, ETV4, ETV5, FLI1, FRA1, GLI2, GRO1, HCK, IL7R, MYBL1, MYCN, PIM1 and TAL1. Ta9 introduction also affected genes associated with proinflammatory processes such as cytokines, chemokines, growth factors and metalloproteinases. Enrichment analysis of differentially expressed genes revealed that Ta9 is potentially involved in activating other host cell signaling pathways in addition to those that lead to induction of AP-1. Comparing our data with data on differentially expressed BoMac genes modulated by the secreted TashAT2 factor of T. annulata identified the gene encoding the tyrosine protein kinase hematopoietic cell kinase (HCK) as common to both data sets. HCK is essential for the proliferation of T. parva-transformed B cells and herein, we demonstrate that enzymatic activity of HCK is also essential for T. annulata- and T. lestoquardi-transformed macrophage proliferation.
Collapse
Affiliation(s)
- Shahin Tajeri
- Freie Universität Berlin, Institute for Parasitology and Tropical Veterinary Medicine, Berlin, Germany; Freie Universität Berlin, Veterinary Centre for Resistance Research, Berlin, Germany.
| | - Brian Shiels
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gordon Langsley
- Inserm U1016-CNRS UMR8104, Institut Cochin, Paris, France; Laboratoire de Biologie Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France
| | - Ard Menzo Nijhof
- Freie Universität Berlin, Institute for Parasitology and Tropical Veterinary Medicine, Berlin, Germany; Freie Universität Berlin, Veterinary Centre for Resistance Research, Berlin, Germany.
| |
Collapse
|
2
|
Shimba A, Tani-Ichi S, Masuda K, Cui G, Munakata S, Abe S, Kitano S, Miyachi H, Kawamoto H, Ikuta K. A Chimeric IL-7Rα/IL-2Rβ Receptor Promotes the Differentiation of T Cell Progenitors into B Cells and Type 2 Innate Lymphoid Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:952-964. [PMID: 39140896 DOI: 10.4049/jimmunol.2300483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 07/29/2024] [Indexed: 08/15/2024]
Abstract
IL-7 and IL-2 are evolutionarily related cytokines that play critical roles in the development and expansion of immune cells. Although both IL-7R and IL-2R activate similar signaling molecules, whether their signals have specific or overlapping functions during lymphocyte differentiation remains unclear. To address this question, we generated IL-7R α-chain (IL-7Rα)/IL-2R β-chain (IL-24β) (72R) knock-in mice expressing a chimeric receptor consisting of the extracellular domain of IL-7Rα and the intracellular domain of IL-2Rβ under the control of the endogenous IL-7Rα promoter. Notably, this 72R receptor induced higher levels of STAT5 and Akt phosphorylation in T cells. In the periphery of 72R mice, the number of T cells, B cells, and type 2 innate lymphoid cells (ILC2s) was increased, whereas early T cell progenitors and double-negative 2 thymocytes were reduced in the thymus. In addition, cell proliferation and Notch signaling were impaired in the early thymocytes of 72R mice, leading to their differentiation into thymic B cells. Interestingly, ILC2s were increased in the thymus of 72R mice. Early T cell progenitors from 72R mice, but not from wild-type mice, differentiated into NK cells and ILC2-like cells when cocultured with a thymic stromal cell line. Thus, this study indicates that the chimeric 72R receptor transduces more robust signals than the authentic IL-7Rα, thereby inducing the alternative differentiation of T cell progenitors into other cell lineages. This suggests that cytokine receptors may provide instructive signals for cell fate decisions.
Collapse
Affiliation(s)
- Akihiro Shimba
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shizue Tani-Ichi
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kyoko Masuda
- Laboratory of Immunology, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Guangwei Cui
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Satoru Munakata
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shinya Abe
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Satsuki Kitano
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hitoshi Miyachi
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Kawamoto
- Laboratory of Immunology, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
Ha TC, Morgan MA, Thrasher AJ, Schambach A. Alpharetroviral Vector-Mediated Gene Therapy for IL7RA-Deficient Severe Combined Immunodeficiency. Hum Gene Ther 2024; 35:669-679. [PMID: 39150017 DOI: 10.1089/hum.2024.103] [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] [Indexed: 08/17/2024] Open
Abstract
Severe combined immunodeficiency (SCID) encompasses rare primary immunodeficiency disorders characterized by deficient T-cell development, which leads to a severely compromised immune system and susceptibility to life-threatening infections. Among SCID subtypes, IL7RA-SCID is caused by mutations in the interleukin 7 receptor alpha chain (IL7RA) and represents a significant subset of patients with limited treatment options. This study investigated the efficacy of a self-inactivating (SIN) alpharetroviral vector (ARV) engineered to deliver a codon-optimized IL7RA cDNA to restore T-cell development in Il7r-knockout mice. We compared the elongation factor 1 alpha short (EFS) promoter and the lymphoid-restricted Lck promoter for their ability to drive IL7RA expression and found that the EFS promoter enabled robust and sustained IL7RA expression that led to the functional rescue of T-lymphopoiesis in vitro and in vivo. Conversely, though effective in vitro, the Lck promoter failed to produce viable T-cell populations in vivo. Our results highlight the potential of using SIN-ARVs as a gene therapy (GT) strategy for treating IL7RA-SCID. Importantly, sustained production of T-lymphocytes was found in both primary and secondary transplant recipient animals with no adverse effects, supporting the safety and feasibility of this approach. Overall, this study provides valuable insights into the development of GT for IL7RA-SCID and underscores the clinical potential of an EFS-driven SIN-ARV to restore IL7RA-deficient immune function.
Collapse
Affiliation(s)
- Teng-Cheong Ha
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH, Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Michael A Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH, Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Adrian J Thrasher
- Infection, Immunity, and Inflammation Teaching and Research Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Center, London, United Kingdom
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH, Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Xu X, Huang Z, Ding C, Deng S, Ou J, Cai Z, Zhou Y, Liang H, Chen J, Wang Z, Liu X, Xuan L, Liu Q, Zheng Z, Li Z, Zhou H. STAT5 phosphorylation plus minimal residual disease defines a novel risk classification in adult B-cell acute lymphoblastic leukaemia. Br J Haematol 2024; 205:517-528. [PMID: 38639167 DOI: 10.1111/bjh.19467] [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: 12/20/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
The dysregulation of the Janus family tyrosine kinase-signal transducer and activator of transcription (JAK-STAT) is closely related to acute lymphoblastic leukaemia (ALL), whereas the clinical value of phosphorylated STAT5 (pSTAT5) remains elusive. Herein we performed a prospective study on clinical significance of flow cytometry-based pSTAT5 in adult B-ALL patients. A total of 184 patients were enrolled in the Precision-Classification-Directed-Target-Total-Therapy (PDT)-ALL-2016 cohort between January 2018 and December 2021, and STAT5 phosphorylation was detected by flow cytometry at diagnosis. Based on flow-pSTAT5, the population was classified into pSTAT5low (113/184, 61.1%) and pSTAT5high (71/184, 38.9%). Overall survival (OS) and event-free survival (EFS) were inferior in pSTAT5high patients than in those with pSTAT5low (OS, 44.8% vs. 65.2%, p = 0.004; EFS, 23.5% vs. 52.1%, p < 0.001), which was further confirmed in an external validation cohort. Furthermore, pSTAT5 plus flow-based minimal residual disease (MRD) postinduction defines a novel risk classification as being high risk (HR, pSTAT5high + MRD+), standard risk (SR, pSTAT5low + MRD-) and others as moderate-risk group. Three identified patient subgroups are distinguishable with disparate survival curves (3-year OS rates, 36.5%, 56.7% and 76.3%, p < 0.001), which was confirmed on multivariate analysis (hazard ratio 3.53, p = 0.003). Collectively, our study proposed a novel, simple and flow-based risk classification by integrating pSTAT5 and MRD in favour of risk-guided treatment for B-ALL.
Collapse
Affiliation(s)
- Xiuli Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Hematology, Ganzhou People's Hospital (Nanfang Hospital Ganzhou Hospital), Ganzhou, China
| | - Zicong Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chenhao Ding
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shiyu Deng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawang Ou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihong Cai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haimei Liang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - ZhiXiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Hematology, Ganzhou People's Hospital (Nanfang Hospital Ganzhou Hospital), Ganzhou, China
| | - Xiaoli Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Hematology, Ganzhou People's Hospital (Nanfang Hospital Ganzhou Hospital), Ganzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research, Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research, Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Zhongxin Zheng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Hematology, Ganzhou People's Hospital (Nanfang Hospital Ganzhou Hospital), Ganzhou, China
- Clinical Medical Research, Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| |
Collapse
|
5
|
Fernandes MB, Gomes AM, Oliveira ML, Caldas J, Lúcio P, Kim R, Caye-Eude A, Pereira F, De Sousa AB, De Stefano A, Follo MY, Soares MV, Lacerda JF, Desterro J, Cavé H, Clappier E, Duarte X, Ribeiro P, Barata JT. Differential activation of basal and IL-7-induced PI3K/Akt/ mTOR and JAK/STAT5 signaling distinguishes pediatric from adult acute lymphoblastic leukemia. Haematologica 2024; 109:2671-2675. [PMID: 38654666 PMCID: PMC11290547 DOI: 10.3324/haematol.2023.284102] [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: 08/18/2023] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Marta B Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - A Margarida Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - Mariana L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | | | - Paulo Lúcio
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal; Champalimaud Centre for the Unknown, Lisboa
| | - Rathana Kim
- Hematology Laboratory, Saint-Louis Hospital, AP-HP, Paris, France; Saint-Louis Research Institute, Université de Paris, INSERM U944/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7212, Paris
| | - Aurélie Caye-Eude
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Filomena Pereira
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa
| | | | - Alessia De Stefano
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Portugal; University of Bologna, Department of Biomedical and Neuromotor Sciences
| | - Matilde Y Follo
- University of Bologna, Department of Biomedical and Neuromotor Sciences
| | - Maria V Soares
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - João F Lacerda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa
| | - Joana Desterro
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa
| | - Hélène Cavé
- Saint-Louis Research Institute, Université de Paris, INSERM U944/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7212, Paris, France; Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris
| | - Emmanuelle Clappier
- Champalimaud Centre for the Unknown, Lisboa, Portugal; Hematology Laboratory, Saint-Louis Hospital, AP-HP, Paris
| | - Ximo Duarte
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa
| | | | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa.
| |
Collapse
|
6
|
Baumgärtner LAF, Ettich J, Balles H, Lapp DJ, Mossner S, Bassenge C, Ouzin M, Hanenberg H, Scheller J, Floss DM. Unpaired cysteine insertions favor transmembrane dimerization and induce ligand-independent constitutive cytokine receptor signaling. Biol Chem 2024; 405:531-544. [PMID: 38695485 DOI: 10.1515/hsz-2023-0344] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/25/2024] [Indexed: 07/14/2024]
Abstract
Naturally occurring gain-of-function (GOF) mutants have been identified in patients for a variety of cytokine receptors. Although this constitutive activation of cytokine receptors is strongly associated with malignant disorders, ligand-independent receptor activation is also a useful tool in synthetic biology e.g. to improve adoptive cellular therapies with genetically modified T-cells. Balanced Interleukin (IL-)7 signaling via a heterodimer of IL-7 receptor (IL-7Rα) and the common γ-chain (γc) controls T- and B-cell development and expansion, whereas uncontrolled IL-7 signaling can drive acute lymphoid leukemia (ALL) development. The ALL-driver mutation PPCL in the transmembrane domain of IL-7Rα is a mutational insertion of the four amino acids proline-proline-cysteine-leucine and leads to ligand-independent receptor dimerization and constitutive activation. We showed here in the cytokine-dependent pre-B-cell line Ba/F3 that the PPCL-insertion in a synthetic version of the IL-7Rα induced γc-independent STAT5 and ERK phosphorylation and also proliferation of the cells and that booster-stimulation by arteficial ligands additionally generated non-canonical STAT3 phosphorylation via the synthetic IL-7Rα-PPCL-receptors. Transfer of the IL-7Rα transmembrane domain with the PPCL insertion into natural and synthetic cytokine receptor chains of the IL-6, IL-12 and Interferon families also resulted in constitutive receptor signaling. In conclusion, our data suggested that the insertion of the mutated PPCL IL-7Rα transmembrane domain is an universal approach to generate ligand-independent, constitutively active cytokine receptors.
Collapse
Affiliation(s)
- Lynn Affrica Felicitas Baumgärtner
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Helene Balles
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Dorothee Johanna Lapp
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Sofie Mossner
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Christin Bassenge
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Meryem Ouzin
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Doreen Manuela Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| |
Collapse
|
7
|
Garcia C, Miller-Awe MD, Witkowski MT. Concepts in B cell acute lymphoblastic leukemia pathogenesis. J Leukoc Biol 2024; 116:18-32. [PMID: 38243586 DOI: 10.1093/jleuko/qiae015] [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: 10/26/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
B cell acute lymphoblastic leukemia (B-ALL) arises from genetic alterations impacting B cell progenitors, ultimately leading to clinically overt disease. Extensive collaborative efforts in basic and clinical research have significantly improved patient prognoses. Nevertheless, a subset of patients demonstrate resistance to conventional chemotherapeutic approaches and emerging immunotherapeutic interventions. This review highlights the mechanistic underpinnings governing B-ALL transformation. Beginning with exploring normative B cell lymphopoiesis, we delineate the influence of recurrent germline and somatic genetic aberrations on the perturbation of B cell progenitor differentiation and protumorigenic signaling, thereby facilitating the neoplastic transformation underlying B-ALL progression. Additionally, we highlight recent advances in the multifaceted landscape of B-ALL, encompassing metabolic reprogramming, microbiome influences, inflammation, and the discernible impact of socioeconomic and racial disparities on B-ALL transformation and patient survival.
Collapse
Affiliation(s)
- Clarissa Garcia
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| | - Megan D Miller-Awe
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| | - Matthew T Witkowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| |
Collapse
|
8
|
Lenk L, Baccelli I, Laqua A, Heymann J, Reimer C, Dietterle A, Winterberg D, Mary C, Corallo F, Taurelle J, Narbeburu E, Neyton S, Déramé M, Pengam S, Vogiatzi F, Bornhauser B, Bourquin JP, Raffel S, Dovhan V, Schüler T, Escherich G, den Boer ML, Boer JM, Wessels W, Peipp M, Alten J, Antić Ž, Bergmann AK, Schrappe M, Cario G, Brüggemann M, Poirier N, Schewe DM. The IL-7R antagonist lusvertikimab reduces leukemic burden in xenograft ALL via antibody-dependent cellular phagocytosis. Blood 2024; 143:2735-2748. [PMID: 38518105 PMCID: PMC11251409 DOI: 10.1182/blood.2023021088] [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: 05/08/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024] Open
Abstract
ABSTRACT Acute lymphoblastic leukemia (ALL) arises from the uncontrolled proliferation of B-cell precursors (BCP-ALL) or T cells (T-ALL). Current treatment protocols obtain high cure rates in children but are based on toxic polychemotherapy. Novel therapies are urgently needed, especially in relapsed/refractory (R/R) disease, high-risk (HR) leukemias and T-ALL, in which immunotherapy approaches remain scarce. Although the interleukin-7 receptor (IL-7R) plays a pivotal role in ALL development, no IL-7R-targeting immunotherapy has yet reached clinical application in ALL. The IL-7Rα chain (CD127)-targeting IgG4 antibody lusvertikimab (LUSV; formerly OSE-127) is a full antagonist of the IL-7R pathway, showing a good safety profile in healthy volunteers. Here, we show that ∼85% of ALL cases express surface CD127. We demonstrate significant in vivo efficacy of LUSV immunotherapy in a heterogeneous cohort of BCP- and T-ALL patient-derived xenografts (PDX) in minimal residual disease (MRD) and overt leukemia models, including R/R and HR leukemias. Importantly, LUSV was particularly effective when combined with polychemotherapy in a phase 2-like PDX study with CD127high samples leading to MRD-negativity in >50% of mice treated with combination therapy. Mechanistically, LUSV targeted ALL cells via a dual mode of action comprising direct IL-7R antagonistic activity and induction of macrophage-mediated antibody-dependent cellular phagocytosis (ADCP). LUSV-mediated in vitro ADCP levels significantly correlated with CD127 expression levels and the reduction of leukemia burden upon treatment of PDX animals in vivo. Altogether, through its dual mode of action and good safety profile, LUSV may represent a novel immunotherapy option for any CD127+ ALL, particularly in combination with standard-of-care polychemotherapy.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Xenograft Model Antitumor Assays
- Receptors, Interleukin-7/antagonists & inhibitors
- Mice, SCID
- Phagocytosis/drug effects
- Interleukin-7 Receptor alpha Subunit
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Female
- Mice, Inbred NOD
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Cell Line, Tumor
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
Collapse
Affiliation(s)
- Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | - Anna Laqua
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Heymann
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Claas Reimer
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Anna Dietterle
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Dorothee Winterberg
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | | | | | | | | | | | | | - Fotini Vogiatzi
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Beat Bornhauser
- Department of Pediatric Hematology/Oncology, University Children's Hospital, Zurich, Switzerland
| | - Jean-Pierre Bourquin
- Department of Pediatric Hematology/Oncology, University Children's Hospital, Zurich, Switzerland
| | - Simon Raffel
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Vladyslava Dovhan
- Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Gabriele Escherich
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Wiebke Wessels
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Željko Antić
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Anke K. Bergmann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Martin Schrappe
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Gunnar Cario
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Monika Brüggemann
- Department of Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Denis M. Schewe
- Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
9
|
Qu J, Li Y, Wu B, Shen Q, Chen L, Sun W, Wang B, Ying L, Wu L, Zhou H, Zhou J, Zhou J. CD161 +CD127 +CD8 + T cell subsets can predict the efficacy of anti-PD-1 immunotherapy in non-small cell lung cancer with diabetes mellitus. Oncoimmunology 2024; 13:2371575. [PMID: 38952673 PMCID: PMC11216103 DOI: 10.1080/2162402x.2024.2371575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024] Open
Abstract
The role of CD161+CD127+CD8+ T cells in non-small cell lung cancer (NSCLC) patients with diabetes remains unexplored. This study determined the prevalence, phenotype, and function of CD8+ T cell subsets in NSCLC with diabetes. We recruited NSCLC patients (n = 436) treated with anti-PD-1 immunotherapy as first-line treatment. The progression-free survival (PFS), overall survival (OS), T cells infiltration, and peripheral blood immunological characteristics were analyzed in NSCLC patients with or without diabetes. NSCLC patients with diabetes exhibited shorter PFS and OS (p = 0.0069 and p = 0.012, respectively) and significantly lower CD8+ T cells infiltration. Mass cytometry by time-of-flight (CyTOF) showed a higher percentage of CD161+CD127+CD8+ T cells among CD8+T cells in NSCLC with diabetes before anti-PD-1 treatment (p = 0.0071) than that in NSCLC without diabetes and this trend continued after anti-PD-1 treatment (p = 0.0393). Flow cytometry and multiple-immunofluorescence confirmed that NSCLC with diabetes had significantly higher CD161+CD127+CD8+ T cells to CD8+T cells ratios than NSCLC patients without diabetes. The RNA-sequencing analysis revealed immune-cytotoxic genes were reduced in the CD161+CD127+CD8+ T cell subset compared to CD161+CD127-CD8+ T cells in NSCLC with diabetes. CD161+CD127+CD8+ T cells exhibited more T cell-exhausted phenotypes in NSCLC with diabetes. NSCLC patients with diabetes with ≥ 6.3% CD161+CD127+CD8+ T cells to CD8+T cells ratios showed worse PFS. These findings indicate that diabetes is a risk factor for NSCLC patients who undergo anti-PD-1 immunotherapy.CD161+CD127+CD8+ T cells could be a key indicator of a poor prognosis in NSCLC with diabetes. Our findings would help in advancing anti-PD-1 therapy in NSCLC patients with diabetes.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Yuekang Li
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Binggen Wu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Qian Shen
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Wenjia Sun
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Bo Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Lixiong Ying
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Li Wu
- Department of Endocrinology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Hong Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
10
|
Mandell JD, Diviti S, Xu M, Townsend JP. Rare Drivers at Low Prevalence with High Cancer Effects in T-Cell and B-Cell Pediatric Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:6589. [PMID: 38928295 PMCID: PMC11203805 DOI: 10.3390/ijms25126589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The genomic analyses of pediatric acute lymphoblastic leukemia (ALL) subtypes, particularly T-cell and B-cell lineages, have been pivotal in identifying potential therapeutic targets. Typical genomic analyses have directed attention toward the most commonly mutated genes. However, assessing the contribution of mutations to cancer phenotypes is crucial. Therefore, we estimated the cancer effects (scaled selection coefficients) for somatic substitutions in T-cell and B-cell cohorts, revealing key insights into mutation contributions. Cancer effects for well-known, frequently mutated genes like NRAS and KRAS in B-ALL were high, which underscores their importance as therapeutic targets. However, less frequently mutated genes IL7R, XBP1, and TOX also demonstrated high cancer effects, suggesting pivotal roles in the development of leukemia when present. In T-ALL, KRAS and NRAS are less frequently mutated than in B-ALL. However, their cancer effects when present are high in both subtypes. Mutations in PIK3R1 and RPL10 were not at high prevalence, yet exhibited some of the highest cancer effects in individual T-cell ALL patients. Even CDKN2A, with a low prevalence and relatively modest cancer effect, is potentially highly relevant for the epistatic effects that its mutated form exerts on other mutations. Prioritizing investigation into these moderately frequent but potentially high-impact targets not only presents novel personalized therapeutic opportunities but also enhances the understanding of disease mechanisms and advances precision therapeutics for pediatric ALL.
Collapse
Affiliation(s)
- Jeffrey D. Mandell
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA;
| | | | - Mina Xu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA;
- Program in Genetics, Genomics, and Epigenetics, Yale Cancer Center, New Haven, CT 06520, USA
| | - Jeffrey P. Townsend
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA;
- Program in Genetics, Genomics, and Epigenetics, Yale Cancer Center, New Haven, CT 06520, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
11
|
Xu Y, Zhou Q, Wang X, Zhang A, Qi W, Li Y, Zheng C, Guan J, Sun T, Li J, Lu C, Shen Y, Zhao B. PELI2 regulates early B-cell progenitor differentiation and related leukemia via the IL-7R expression. Haematologica 2024; 109:1800-1814. [PMID: 38058209 PMCID: PMC11141684 DOI: 10.3324/haematol.2023.284041] [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: 08/06/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023] Open
Abstract
Little is known about the transition mechanisms that govern early lymphoid lineage progenitors from common lymphoid progenitors (CLP). Pellino2 (PELI2) is a newly discovered E3 ubiquitin ligase, which plays important roles in inflammation and the immune system. However, the physiological and molecular roles of PELI2 in the differentiation of immune cells are largely unknown. Here, by using a conditional knockout mouse model, we demonstrated that PELI2 is required for early B-cell development and stressed hematopoiesis. PELI2 interacted with and stabilized PU.1 via K63-polyubiquitination to regulate IL-7R expression. The defects of B-cell development induced by PELI2 deletion were restored by overexpression of PU.1. Similarly, PELI2 promoted TCF3 protein stability via K63-polyubiquitination to regulate IL-7R expression, which is required for the proliferation of B-cell precursor acute lymphoblastic leukemia (BCP-ALL) cells. These results underscore the significance of PELI2 in both normal B lymphopoiesis and malignant B-cell acute lymphoblastic leukemia via the regulation of IL-7R expression, providing a potential therapeutic approach for BCP-ALL.
Collapse
Affiliation(s)
- Yan Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012
| | - Qian Zhou
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012
| | - Xiaoming Wang
- Department of Pediatrics, Qilu hospital of Shandong University, Jinan, Shandong, 250012
| | - Aijun Zhang
- Department of Pediatrics, Qilu hospital of Shandong University, Jinan, Shandong, 250012
| | - Wentao Qi
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012
| | - Yuan Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012
| | - Chengzu Zheng
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012
| | - Jianmin Guan
- Department of Hematology, Heze Municipal Hospital, Heze, Shandong
| | - Tao Sun
- Department of Hematology, Qilu hospital of Shandong University, Jinan, Shandong, 250012
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012
| | - Chunhua Lu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012
| | - Yuemao Shen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012
| | - Baobing Zhao
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012.
| |
Collapse
|
12
|
Zhang Y, Morris R, Brown GJ, Lorenzo AMD, Meng X, Kershaw NJ, Kiridena P, Burgio G, Gross S, Cappello JY, Shen Q, Wang H, Turnbull C, Lea-Henry T, Stanley M, Yu Z, Ballard FD, Chuah A, Lee JC, Hatch AM, Enders A, Masters SL, Headley AP, Trnka P, Mallon D, Fletcher JT, Walters GD, Šestan M, Jelušić M, Cook MC, Athanasopoulos V, Fulcher DA, Babon JJ, Vinuesa CG, Ellyard JI. Rare SH2B3 coding variants in lupus patients impair B cell tolerance and predispose to autoimmunity. J Exp Med 2024; 221:e20221080. [PMID: 38417019 PMCID: PMC10901239 DOI: 10.1084/jem.20221080] [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: 06/23/2022] [Revised: 03/14/2023] [Accepted: 01/17/2024] [Indexed: 03/01/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease with a clear genetic component. While most SLE patients carry rare gene variants in lupus risk genes, little is known about their contribution to disease pathogenesis. Amongst them, SH2B3-a negative regulator of cytokine and growth factor receptor signaling-harbors rare coding variants in over 5% of SLE patients. Here, we show that unlike the variant found exclusively in healthy controls, SH2B3 rare variants found in lupus patients are predominantly hypomorphic alleles, failing to suppress IFNGR signaling via JAK2-STAT1. The generation of two mouse lines carrying patients' variants revealed that SH2B3 is important in limiting the number of immature and transitional B cells. Furthermore, hypomorphic SH2B3 was shown to impair the negative selection of immature/transitional self-reactive B cells and accelerate autoimmunity in sensitized mice, at least in part due to increased IL-4R signaling and BAFF-R expression. This work identifies a previously unappreciated role for SH2B3 in human B cell tolerance and lupus risk.
Collapse
Affiliation(s)
- Yaoyuan Zhang
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Rhiannon Morris
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Grant J. Brown
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Ayla May D. Lorenzo
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Xiangpeng Meng
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Nadia J. Kershaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Pamudika Kiridena
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Gaétan Burgio
- Division of Genome Sciences and Cancer, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Simon Gross
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Jean Y. Cappello
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Qian Shen
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Francis Crick Institute, London, UK
| | - Hao Wang
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Francis Crick Institute, London, UK
| | - Cynthia Turnbull
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Tom Lea-Henry
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Canberra Hospital, Garran, Australia
| | - Maurice Stanley
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Zhijia Yu
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Fiona D. Ballard
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Aaron Chuah
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - James C. Lee
- Francis Crick Institute, London, UK
- Department of Gastroenterology, Division of Medicine, Institute for Liver and Digestive Health, University College London, London, UK
| | - Ann-Maree Hatch
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Canberra Hospital, Garran, Australia
| | - Anselm Enders
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Seth L. Masters
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | | | - Peter Trnka
- Queensland Children’s Hospital, South Brisbane, Australia
| | | | | | | | - Mario Šestan
- Department of Pediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marija Jelušić
- Department of Pediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Matthew C. Cook
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Canberra Hospital, Garran, Australia
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | - Vicki Athanasopoulos
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - David A. Fulcher
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Jeffrey J. Babon
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Carola G. Vinuesa
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Francis Crick Institute, London, UK
| | - Julia I. Ellyard
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| |
Collapse
|
13
|
Rodrigues de Oliveira B, Iansavitchous J, Rysan H, Wang WC, Sams MP, Knight D, Xu LS, Jeong J, Qu TP, Zorzi AP, DeKoter RP. IKZF3/Aiolos H195Y mutation identified in a mouse model of B cell leukemia results in altered DNA binding and altered STAT5-dependent gene expression. Gene 2024; 900:148131. [PMID: 38216003 DOI: 10.1016/j.gene.2024.148131] [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: 11/21/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Precursor B cell acute lymphoblastic leukemia (Pre-B-ALL) arises from developing B cells and frequently involves mutations in genes encoding transcription factors. In this study, we investigated the function of mutations in the transcription factor IKZF3 (Aiolos), R137* and H195Y, discovered in a mouse model of pre-B-ALL. R137* IKZF3 mutation resulted in a truncated protein, while electrophoretic mobility shift assay showed that H195Y IKZF3 mutation resulted in a protein with altered DNA binding. 38B9 pre-B cell lines were generated expressing WT and H195Y IKZF3 proteins. Anti-IKZF3 ChIP-seq showed that H195Y IKZF3 interacted with a larger number of sites that were different than WT IKZF3. Treatment with interleukin-7 induced changes in gene expression in 38B9 cells expressing WT IKZF3, but did not induce any changes in gene expression in cells expressing H195Y IKZF3. Anti-STAT5 ChIP-seq showed that expression of H195Y IKZF3 resulted in redistribution of STAT5 binding sites in the genome. H195Y IKZF3 binding sites overlapped with a subset of STAT5 binding sites, including in the promoter of the Cish gene. These findings suggest that H195Y mutation of IKZF3 results in altered DNA binding specificity and altered binding of STAT5 to target genes.
Collapse
Affiliation(s)
- Bruno Rodrigues de Oliveira
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - James Iansavitchous
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Heidi Rysan
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Wei Cen Wang
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mia P Sams
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Devon Knight
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Li S Xu
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Jeewoo Jeong
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Thomas P Qu
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Alexandra P Zorzi
- Department of Paediatrics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Rodney P DeKoter
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, Ontario, Canada.
| |
Collapse
|
14
|
Salimi A, Schemionek‐Reinders M, Huber M, Vieri M, Patterson JB, Alten J, Brümmendorf TH, Kharabi Masouleh B, Appelmann I. XBP1 promotes NRAS G12D pre-B acute lymphoblastic leukaemia through IL-7 receptor signalling and provides a therapeutic vulnerability for oncogenic RAS. J Cell Mol Med 2023; 27:3363-3377. [PMID: 37753803 PMCID: PMC10623536 DOI: 10.1111/jcmm.17904] [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: 04/13/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/28/2023] Open
Abstract
Activating point mutations of the RAS gene act as driver mutations for a subset of precursor-B cell acute lymphoblastic leukaemias (pre-B ALL) and represent an ambitious target for therapeutic approaches. The X box-binding protein 1 (XBP1), a key regulator of the unfolded protein response (UPR), is critical for pre-B ALL cell survival, and high expression of XBP1 confers poor prognosis in ALL patients. However, the mechanism of XBP1 activation has not yet been elucidated in RAS mutated pre-B ALL. Here, we demonstrate that XBP1 acts as a downstream linchpin of the IL-7 receptor signalling pathway and that pharmacological inhibition or genetic ablation of XBP1 selectively abrogates IL-7 receptor signalling via inhibition of its downstream effectors, JAK1 and STAT5. We show that XBP1 supports malignant cell growth of pre-B NRASG12D ALL cells and that genetic loss of XBP1 consequently leads to cell cycle arrest and apoptosis. Our findings reveal that active XBP1 prevents the cytotoxic effects of a dual PI3K/mTOR pathway inhibitor (BEZ235) in pre-B NRASG12D ALL cells. This implies targeting XBP1 in combination with BEZ235 as a promising new targeted strategy against the oncogenic RAS in NRASG12D -mutated pre-B ALL.
Collapse
Affiliation(s)
- Azam Salimi
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging CenterEberhard Karls University TübingenTübingenGermany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies"TübingenGermany
| | - Mirle Schemionek‐Reinders
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Michael Huber
- Medical Faculty, Institute of Biochemistry and Molecular ImmunologyRWTH Aachen UniversityAachenGermany
| | - Margherita Vieri
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | | | - Julia Alten
- Department of PediatricsUniversity Medical Centre Schleswig‐HolsteinKielGermany
| | - Tim H. Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Behzad Kharabi Masouleh
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Iris Appelmann
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| |
Collapse
|
15
|
Casado-García A, Isidro-Hernández M, Alemán-Arteaga S, Ruiz-Corzo B, Riesco S, Prieto-Matos P, Sánchez L, Sánchez-García I, Vicente-Dueñas C. Lessons from mouse models in the impact of risk factors on the genesis of childhood B-cell leukemia. Front Immunol 2023; 14:1285743. [PMID: 37901253 PMCID: PMC10602728 DOI: 10.3389/fimmu.2023.1285743] [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: 08/30/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) stands as the primary contributor to childhood cancer-related mortality on a global scale. The development of the most conventional forms of this disease has been proposed to be conducted by two different steps influenced by different types of risk factors. The first step is led by a genetic insult that is presumably acquired before birth that transforms a healthy cell into a preleukemic one, which is maintained untransformed until the second step takes place. This necessary next step to leukemia development will be triggered by different risk factors to which children are exposed after birth. Murine models that recap the stepwise progression of B-ALL have been instrumental in identifying environmental and genetic factors that contribute to disease risk. Recent evidence from these models has demonstrated that specific environmental risk factors, such as common infections or gut microbiome dysbiosis, induce immune stress, driving the transformation of preleukemic cells, and harboring genetic alterations, into fully transformed leukemic cells. Such models serve as valuable tools for investigating the mechanisms underlying preleukemic events and can aid in the development of preventive approaches for leukemia in child. Here, we discuss the existing knowledge, learned from mouse models, of the impact of genetic and environmental risk factors on childhood B-ALL evolution and how B-ALL prevention could be reached by interfering with preleukemic cells.
Collapse
Affiliation(s)
- Ana Casado-García
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Marta Isidro-Hernández
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Silvia Alemán-Arteaga
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Belén Ruiz-Corzo
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Susana Riesco
- Department of Pediatrics, Hospital Universitario de Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Pablo Prieto-Matos
- Department of Pediatrics, Hospital Universitario de Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Lucía Sánchez
- School of Law, University of Salamanca, Salamanca, Spain
| | - Isidro Sánchez-García
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Carolina Vicente-Dueñas
- Department of Pediatrics, Hospital Universitario de Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
16
|
Kaiser FMP, Janowska I, Menafra R, de Gier M, Korzhenevich J, Pico-Knijnenburg I, Khatri I, Schulz A, Kuijpers TW, Lankester AC, Konstantinidis L, Erlacher M, Kloet S, van Schouwenburg PA, Rizzi M, van der Burg M. IL-7 receptor signaling drives human B-cell progenitor differentiation and expansion. Blood 2023; 142:1113-1130. [PMID: 37369082 PMCID: PMC10644098 DOI: 10.1182/blood.2023019721] [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: 01/26/2023] [Revised: 05/18/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Although absence of interleukin-7 (IL-7) signaling completely abrogates T and B lymphopoiesis in mice, patients with severe combined immunodeficiency caused by mutations in the IL-7 receptor α chain (IL-7Rα) still generate peripheral blood B cells. Consequently, human B lymphopoiesis has been thought to be independent of IL-7 signaling. Using flow cytometric analysis and single-cell RNA sequencing of bone marrow samples from healthy controls and patients who are IL-7Rα deficient, in combination with in vitro modeling of human B-cell differentiation, we demonstrate that IL-7R signaling plays a crucial role in human B lymphopoiesis. IL-7 drives proliferation and expansion of early B-cell progenitors but not of pre-BII large cells and has a limited role in the prevention of cell death. Furthermore, IL-7 guides cell fate decisions by enhancing the expression of BACH2, EBF1, and PAX5, which jointly orchestrate the specification and commitment of early B-cell progenitors. In line with this observation, early B-cell progenitors of patients with IL-7Rα deficiency still expressed myeloid-specific genes. Collectively, our results unveil a previously unknown role for IL-7 signaling in promoting the B-lymphoid fate and expanding early human B-cell progenitors while defining important differences between mice and humans. Our results have implications for hematopoietic stem cell transplantation strategies in patients with T- B+ severe combined immunodeficiency and provide insights into the role of IL-7R signaling in leukemogenesis.
Collapse
Affiliation(s)
- Fabian M. P. Kaiser
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Iga Janowska
- Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Melanie de Gier
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Jakov Korzhenevich
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pico-Knijnenburg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Indu Khatri
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ansgar Schulz
- Department of Pediatrics and Adolescent Medicine, University Medical Center, University Ulm, Ulm, Germany
| | - Taco W. Kuijpers
- Department of Pediatrics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Arjan C. Lankester
- Department of Pediatrics, Hematology and Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Lukas Konstantinidis
- Department of Orthopedics and Trauma Surgery, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
| | - Susan Kloet
- Leiden Genome Technology Center, Leiden, The Netherlands
| | - Pauline A. van Schouwenburg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Marta Rizzi
- Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
17
|
Khanolkar A, Liu G, Simpson Schneider BM. Defining the Basal and Immunomodulatory Mediator-Induced Phosphoprotein Signature in Pediatric B Cell Acute Lymphoblastic Leukemia (B-ALL) Diagnostic Samples. Int J Mol Sci 2023; 24:13937. [PMID: 37762241 PMCID: PMC10531382 DOI: 10.3390/ijms241813937] [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: 06/29/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
It is theorized that dysregulated immune responses to infectious insults contribute to the development of pediatric B-ALL. In this context, our understanding of the immunomodulatory-mediator-induced signaling responses of leukemic blasts in pediatric B-ALL diagnostic samples is rather limited. Hence, in this study, we defined the signaling landscape of leukemic blasts, as well as normal mature B cells and T cells residing in diagnostic samples from 63 pediatric B-ALL patients. These samples were interrogated with a range of immunomodulatory-mediators within 24 h of collection, and phosflow analyses of downstream proximal signaling nodes were performed. Our data reveal evidence of basal hyperphosphorylation across a broad swath of these signaling nodes in leukemic blasts in contrast to normal mature B cells and T cells in the same sample. We also detected similarities in the phosphoprotein signature between blasts and mature B cells in response to IFNγ and IL-2 treatment, but significant divergence in the phosphoprotein signature was observed between blasts and mature B cells in response to IL-4, IL-7, IL-10, IL-21 and CD40 ligand treatment. Our results demonstrate the existence of both symmetry and asymmetry in the phosphoprotein signature between leukemic and non-leukemic cells in pediatric B-ALL diagnostic samples.
Collapse
Affiliation(s)
- Aaruni Khanolkar
- Department of Pathology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA
| | - Guorong Liu
- Department of Pathology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | | |
Collapse
|
18
|
Korzhenevich J, Janowska I, van der Burg M, Rizzi M. Human and mouse early B cell development: So similar but so different. Immunol Lett 2023; 261:1-12. [PMID: 37442242 DOI: 10.1016/j.imlet.2023.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/09/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Early B cell development in the bone marrow ensures the replenishment of the peripheral B cell pool. Immature B cells continuously develop from hematopoietic stem cells, in a process guided by an intricate network of transcription factors as well as chemokine and cytokine signals. Humans and mice possess somewhat similar regulatory mechanisms of B lymphopoiesis. The continuous discovery of monogenetic defects that impact early B cell development in humans substantiates the similarities and differences with B cell development in mice. These differences become relevant when targeted therapeutic approaches are used in patients; therefore, predicting potential immunological adverse events is crucial. In this review, we have provided a phenotypical classification of human and murine early progenitors and B cell stages, based on surface and intracellular protein expression. Further, we have critically compared the role of key transcription factors (Ikaros, E2A, EBF1, PAX5, and Aiolos) and chemo- or cytokine signals (FLT3, c-kit, IL-7R, and CXCR4) during homeostatic and aberrant B lymphopoiesis in both humans and mice.
Collapse
Affiliation(s)
- Jakov Korzhenevich
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Iga Janowska
- Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, University of Freiburg, 79106, Freiburg, Germany
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, 2333, ZA Leiden, The Netherlands
| | - Marta Rizzi
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria; Department of Rheumatology and Clinical Immunology, Freiburg University Medical Center, University of Freiburg, 79106, Freiburg, Germany; Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
19
|
Wang H, Qiu Y, Zhang H, Chang N, Hu Y, Chen J, Hu R, Liao P, Li Z, Yang Y, Cen Q, Ding X, Li M, Xie X, Li Y. Histone acetylation by HBO1 (KAT7) activates Wnt/β-catenin signaling to promote leukemogenesis in B-cell acute lymphoblastic leukemia. Cell Death Dis 2023; 14:498. [PMID: 37542030 PMCID: PMC10403501 DOI: 10.1038/s41419-023-06019-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive hematological disorder with a dismal prognosis. The dysregulation of histone acetylation is of great significance in the pathogenesis and progression of B-ALL. Regarded as a fundamental acetyltransferase gene, the role of HBO1 (lysine acetyltransferase 7/KAT7) in B-ALL has not been investigated. Herein, we found that HBO1 expression was elevated in human B-ALL cells and associated with poor disease-free survival. Strikingly, HBO1 knockdown inhibited viability, proliferation, and G1-S cycle progression in B-ALL cells, while provoking apoptosis. In contrast, ectopic overexpression of HBO1 enhanced cell viability and proliferation but inhibited apoptotic activation. The results of in vivo experiments also certificated the inhibitory effect of HBO1 knockdown on tumor growth. Mechanistically, HBO1 acetylated histone H3K14, H4K8, and H4K12, followed by upregulating CTNNB1 expression, resulting in activation of the Wnt/β-catenin signaling pathway. Moreover, a novel small molecule inhibitor of HBO1, WM-3835, potently inhibited the progression of B-ALL. Our data identified HBO1 as an efficacious regulator of CTNNB1 with therapeutic potential in B-ALL.
Collapse
Affiliation(s)
- Hao Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Honghao Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China.
| | - Ning Chang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Jianyu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Rong Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Peiyun Liao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Zhongwei Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Yulu Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Qingyan Cen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Xiangyang Ding
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Meifang Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China
| | - Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, P. R. China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, 510005, P. R. China.
| |
Collapse
|
20
|
Zhu S, Zhao Y, Xing C, Guo W, Huang Z, Zhang H, Yin L, Ruan X, Li H, Cheng Z, Wang Z, Peng H. Immune infiltration and drug specificity analysis of different subtypes based on functional status in angioimmunoblastic T-cell lymphoma. Heliyon 2023; 9:e18836. [PMID: 37576233 PMCID: PMC10412840 DOI: 10.1016/j.heliyon.2023.e18836] [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: 02/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a subtype of peripheral T-cell lymphoma (PTCL) strongly correlated with worse clinical outcomes. However, the role of characteristic pathway-related genes in patients with AITL (e.g., subtype typing and pathogenesis) remains unknown. In this study, we intended to understand the potential role and prognostic value of characteristic pathways in AITL and identified a model for subtype identification based on pathway-related functional status. Transcriptomic (RNA-seq) data were obtained from the Gene Expression Omnibus database for three sets of tumor tissues from AITL patients. AITL was divided into three clusters based on the pathway profile of patients and the best clustering k = 3, and differentially expressed genes (DEGs) in the three clusters were analyzed. The top 45 important variables associated with characteristic pathways, such as Huntington's disease, VEGF signaling pathway, nucleotide excision repair, ubiquitin-mediated proteolysis, purine metabolism, olfactory transduction, etc., were used to construct a subtype identification model. The model was experimentally validated and proved to possess good predictive efficacy. In addition, pathway-related subtype typing was significantly associated with different immune cell infiltration in AITL. Further analysis revealed that the drug IC50 values predicted also differed markedly among the different subtypes, thus further identifying some subtype-specific drugs. Our study indicates a potential role of characteristic pathways in AITL staging for the first time, provides novel insights for future research targeting AITL, and points to potential therapeutic options for patients with different subtypes of AITL.
Collapse
Affiliation(s)
- Shicong Zhu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Cheng Xing
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Wancheng Guo
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Huifang Zhang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Xueqin Ruan
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zhihua Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan 410011, China
| |
Collapse
|
21
|
Tang J, Yang L, Guan F, Miller H, Camara NOS, James LK, Benlagha K, Kubo M, Heegaard S, Lee P, Lei J, Zeng H, He C, Zhai Z, Liu C. The role of Raptor in lymphocytes differentiation and function. Front Immunol 2023; 14:1146628. [PMID: 37283744 PMCID: PMC10239924 DOI: 10.3389/fimmu.2023.1146628] [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: 01/17/2023] [Accepted: 04/28/2023] [Indexed: 06/08/2023] Open
Abstract
Raptor, a key component of mTORC1, is required for recruiting substrates to mTORC1 and contributing to its subcellular localization. Raptor has a highly conserved N-terminus domain and seven WD40 repeats, which interact with mTOR and other mTORC1-related proteins. mTORC1 participates in various cellular events and mediates differentiation and metabolism. Directly or indirectly, many factors mediate the differentiation and function of lymphocytes that is essential for immunity. In this review, we summarize the role of Raptor in lymphocytes differentiation and function, whereby Raptor mediates the secretion of cytokines to induce early lymphocyte metabolism, development, proliferation and migration. Additionally, Raptor regulates the function of lymphocytes by regulating their steady-state maintenance and activation.
Collapse
Affiliation(s)
- Jianing Tang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Louisa K. James
- Centre for Immunobiology, Bizard Institute, Queen Mary University of London, London, United Kingdom
| | - Kamel Benlagha
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, Paris, France
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), Rikagaku Kenkyusho, Institute of Physical and Chemical Research (RIKEN) Yokohama Institute, Yokohama, Japan
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet Glostrup, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hu Zeng
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
22
|
Das Gupta D, Lohoff M. Puppet masters of B-cell progenitor acute lymphoblastic leukemia: The preB cell receptor and the interleukin 7 receptor α. Eur J Immunol 2023; 53:e2250093. [PMID: 36805963 DOI: 10.1002/eji.202250093] [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: 10/05/2022] [Revised: 12/02/2022] [Accepted: 01/13/2023] [Indexed: 02/23/2023]
Abstract
B-cell progenitor acute lymphoblastic leukemia (BCP-ALL) is enriched for a preB cell phenotype, hinting at a specific vulnerability of this cell stage. Two signaling pathways via the preB cell receptor (preBCR) and the interleukin 7 receptor α (IL-7Rα) chain govern the balance between differentiation and proliferation at this stage and both receptor pathways are routinely altered in human BCP-ALL. Here, we review the immunobiology of both the preBCR as well as the IL-7Rα and analyze the human BCP-ALL spectrum in the light of these signaling complexes. Finally, we present a terminology for preBCR signaling modules that distinguishes a pro-proliferative "phase-I" module from a pro-differentiative "phase-II" module. This terminology might serve as a framework to better address shared oncogenic mechanics of preB cell stage BCP-ALL.
Collapse
Affiliation(s)
- Dennis Das Gupta
- Institute for Medical Microbiology & Hospital Hygiene, Philipps University Marburg, Marburg, Germany.,Medical Department II, Hematology and Oncology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Michael Lohoff
- Institute for Medical Microbiology & Hospital Hygiene, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
23
|
Paisana E, Cascão R, Custódia C, Qin N, Picard D, Pauck D, Carvalho T, Ruivo P, Barreto C, Doutel D, Cabeçadas J, Roque R, Pimentel J, Miguéns J, Remke M, Barata JT, Faria CC. UBE2C promotes leptomeningeal dissemination and is a therapeutic target in brain metastatic disease. Neurooncol Adv 2023; 5:vdad048. [PMID: 37215954 PMCID: PMC10195208 DOI: 10.1093/noajnl/vdad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Background Despite current improvements in systemic cancer treatment, brain metastases (BM) remain incurable, and there is an unmet clinical need for effective targeted therapies. Methods Here, we sought common molecular events in brain metastatic disease. RNA sequencing of thirty human BM identified the upregulation of UBE2C, a gene that ensures the correct transition from metaphase to anaphase, across different primary tumor origins. Results Tissue microarray analysis of an independent BM patient cohort revealed that high expression of UBE2C was associated with decreased survival. UBE2C-driven orthotopic mouse models developed extensive leptomeningeal dissemination, likely due to increased migration and invasion. Early cancer treatment with dactolisib (dual PI3K/mTOR inhibitor) prevented the development of UBE2C-induced leptomeningeal metastases. Conclusions Our findings reveal UBE2C as a key player in the development of metastatic brain disease and highlight PI3K/mTOR inhibition as a promising anticancer therapy to prevent late-stage metastatic brain cancer.
Collapse
Affiliation(s)
- Eunice Paisana
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa; Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Rita Cascão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa; Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Carlos Custódia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa; Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Nan Qin
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University Düsseldorf, Medical Faculty, and University Hospital Düsseldorf; Moorenstraße 5, 40225 Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University Düsseldorf, Medical Faculty, and University Hospital Düsseldorf; Moorenstraße 5, 40225 Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Moorenstraße 5, 40225 Düsseldorf, Germany
| | - David Pauck
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University Düsseldorf, Medical Faculty, and University Hospital Düsseldorf; Moorenstraße 5, 40225 Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa; Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Pedro Ruivo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa; Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Clara Barreto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa; Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Delfim Doutel
- Anatomic Pathology Department, Instituto Português de Oncologia Francisco Gentil, R. Prof. Lima Basto, 1099-023, Lisboa, Portugal
| | - José Cabeçadas
- Anatomic Pathology Department, Instituto Português de Oncologia Francisco Gentil, R. Prof. Lima Basto, 1099-023, Lisboa, Portugal
| | - Rafael Roque
- Neurology Department, Laboratory of Neuropathology, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - José Pimentel
- Neurology Department, Laboratory of Neuropathology, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - José Miguéns
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Marc Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University Düsseldorf, Medical Faculty, and University Hospital Düsseldorf; Moorenstraße 5, 40225 Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Moorenstraße 5, 40225 Düsseldorf, Germany
| | | | - Claudia C Faria
- Corresponding Author: Claudia C. Faria, Instituto de Medicina Molecular João Lobo Antunes, Edifício Egas Moniz, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, Lisboa, 1649-028, Portugal ()
| |
Collapse
|
24
|
Fernandes MB, Barata JT. IL-7 and IL-7R in health and disease: An update through COVID times. Adv Biol Regul 2023; 87:100940. [PMID: 36503870 DOI: 10.1016/j.jbior.2022.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The role of IL-7 and IL-7R for normal lymphoid development and an adequately functioning immune system has been recognized for long, with severe immune deficiency and lymphoid leukemia as extreme examples of the consequences of deregulation of the IL-7-IL-7R axis. In this review, we provide an update (focusing on the past couple of years) on IL-7 and IL-7R in health and disease. We highlight the findings on IL-7/IL-7R signaling mechanisms and the, sometimes controversial, impact of IL-7 and its receptor on leukocyte biology, COVID-19, acute lymphoblastic leukemia, and different solid tumors, as well as their relevance as therapeutic tools or targets.
Collapse
Affiliation(s)
- Marta B Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
25
|
Renatino Canevarolo R, Pereira de Souza Melo C, Moreno Cury N, Luiz Artico L, Ronchi Corrêa J, Tonhasca Lau Y, Sousa Mariano S, Reddy Sudalagunta P, Regina Brandalise S, Carolina de Mattos Zeri A, Andrés Yunes J. Glutathione levels are associated with methotrexate resistance in acute lymphoblastic leukemia cell lines. Front Oncol 2022; 12:1032336. [PMID: 36531023 PMCID: PMC9751399 DOI: 10.3389/fonc.2022.1032336] [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: 08/30/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Methotrexate (MTX), a folic acid antagonist and nucleotide synthesis inhibitor, is a cornerstone drug used against acute lymphoblastic leukemia (ALL), but its mechanism of action and resistance continues to be unraveled even after decades of clinical use. Methods To better understand the mechanisms of this drug, we accessed the intracellular metabolic content of 13 ALL cell lines treated with MTX by 1H-NMR, and correlated metabolome data with cell proliferation and gene expression. Further, we validated these findings by inhibiting the cellular antioxidant system of the cells in vitro and in vivo in the presence of MTX. Results MTX altered the concentration of 31 out of 70 metabolites analyzed, suggesting inhibition of the glycine cleavage system, the pentose phosphate pathway, purine and pyrimidine synthesis, phospholipid metabolism, and bile acid uptake. We found that glutathione (GSH) levels were associated with MTX resistance in both treated and untreated cells, suggesting a new constitutive metabolic-based mechanism of resistance to the drug. Gene expression analyses showed that eight genes involved in GSH metabolism were correlated to GSH concentrations, 2 of which (gamma-glutamyltransferase 1 [GGT1] and thioredoxin reductase 3 [TXNRD3]) were also correlated to MTX resistance. Gene set enrichment analysis (GSEA) confirmed the association between GSH metabolism and MTX resistance. Pharmacological inhibition or stimulation of the main antioxidant systems of the cell, GSH and thioredoxin, confirmed their importance in MTX resistance. Arsenic trioxide (ATO), a thioredoxin inhibitor used against acute promyelocytic leukemia, potentiated MTX cytotoxicity in vitro in some of the ALL cell lines tested. Likewise, the ATO+MTX combination decreased tumor burden and extended the survival of NOD scid gamma (NSG) mice transplanted with patient-derived ALL xenograft, but only in one of four ALLs tested. Conclusion Altogether, our results show that the cellular antioxidant defense systems contribute to leukemia resistance to MTX, and targeting these pathways, especially the thioredoxin antioxidant system, may be a promising strategy for resensitizing ALL to MTX.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanca Tonhasca Lau
- Centro de Pesquisa Boldrini, Centro Infantil Boldrini, Campinas, SP, Brazil
| | | | - Praneeth Reddy Sudalagunta
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | | | - Ana Carolina de Mattos Zeri
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - José Andrés Yunes
- Centro de Pesquisa Boldrini, Centro Infantil Boldrini, Campinas, SP, Brazil,Medical Genetics Department, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil,*Correspondence: José Andrés Yunes,
| |
Collapse
|
26
|
Childhood B-Cell Preleukemia Mouse Modeling. Int J Mol Sci 2022; 23:ijms23147562. [PMID: 35886910 PMCID: PMC9317949 DOI: 10.3390/ijms23147562] [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: 06/15/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 02/04/2023] Open
Abstract
Leukemia is the most usual childhood cancer, and B-cell acute lymphoblastic leukemia (B-ALL) is its most common presentation. It has been proposed that pediatric leukemogenesis occurs through a “multi-step” or “multi-hit” mechanism that includes both in utero and postnatal steps. Many childhood leukemia-initiating events, such as chromosomal translocations, originate in utero, and studies so far suggest that these “first-hits” occur at a far higher frequency than the incidence of childhood leukemia itself. The reason why only a small percentage of the children born with such preleukemic “hits” will develop full-blown leukemia is still a mystery. In order to better understand childhood leukemia, mouse modeling is essential, but only if the multistage process of leukemia can be recapitulated in the model. Therefore, mouse models naturally reproducing the “multi-step” process of childhood B-ALL will be essential to identify environmental or other factors that are directly linked to increased risk of disease.
Collapse
|
27
|
Oliveira ML, Veloso A, Garcia EG, Iyer S, Pereira C, Barreto VM, Langenau DM, Barata JT. Mutant IL7R collaborates with MYC to induce T-cell acute lymphoblastic leukemia. Leukemia 2022; 36:1533-1540. [PMID: 35581375 PMCID: PMC9162918 DOI: 10.1038/s41375-022-01590-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/09/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive pediatric cancer. Amongst the wide array of driver mutations, 10% of T-ALL patients display gain-of-function mutations in the IL-7 receptor α chain (IL-7Rα, encoded by IL7R), which occur in different molecular subtypes of this disease. However, it is still unclear whether IL-7R mutational activation is sufficient to transform T-cell precursors. Also, which genes cooperate with IL7R to drive leukemogenesis remain poorly defined. Here, we demonstrate that mutant IL7R alone is capable of inducing T-ALL with long-latency in stable transgenic zebrafish and transformation is associated with MYC transcriptional activation. Additionally, we find that mutant IL7R collaborates with Myc to induce early onset T-ALL in transgenic zebrafish, supporting a model where these pathways collaborate to drive leukemogenesis. T-ALLs co-expressing mutant IL7R and Myc activate STAT5 and AKT pathways, harbor reduced numbers of apoptotic cells and remake tumors in transplanted zebrafish faster than T-ALLs expressing Myc alone. Moreover, limiting-dilution cell transplantation experiments reveal that activated IL-7R signaling increases the overall frequency of leukemia propagating cells. Our work highlights a synergy between mutant IL7R and Myc in inducing T-ALL and demonstrates that mutant IL7R enriches for leukemia propagating potential.
Collapse
Affiliation(s)
- Mariana L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Alexandra Veloso
- Molecular Pathology Unit, MGH Research Institute, Charlestown, MA, 02129, USA
- MGH Cancer Center, Harvard Medical School, Charlestown, MA, 02129, USA
- Center for Regenerative Medicine, MGH, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02139, USA
| | - Elaine G Garcia
- Molecular Pathology Unit, MGH Research Institute, Charlestown, MA, 02129, USA
- MGH Cancer Center, Harvard Medical School, Charlestown, MA, 02129, USA
- Center for Regenerative Medicine, MGH, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02139, USA
| | - Sowmya Iyer
- Molecular Pathology Unit, MGH Research Institute, Charlestown, MA, 02129, USA
- MGH Cancer Center, Harvard Medical School, Charlestown, MA, 02129, USA
- Center for Regenerative Medicine, MGH, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02139, USA
| | - Clara Pereira
- Smurfit Institute of Genetics, Trinity College Dublin, University of Dublin, Dublin 2, Ireland
| | - Vasco M Barreto
- DNA Breaks Laboratory, CEDOC - Chronic Diseases Research Center, NOVA Medical School - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - David M Langenau
- Molecular Pathology Unit, MGH Research Institute, Charlestown, MA, 02129, USA.
- MGH Cancer Center, Harvard Medical School, Charlestown, MA, 02129, USA.
- Center for Regenerative Medicine, MGH, Boston, MA, 02114, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02139, USA.
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
28
|
In Utero Development and Immunosurveillance of B Cell Acute Lymphoblastic Leukemia. Curr Treat Options Oncol 2022; 23:543-561. [PMID: 35294722 PMCID: PMC8924576 DOI: 10.1007/s11864-022-00963-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2022] [Indexed: 11/06/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most frequent type of pediatric cancer with a peak incidence at 2–5 years of age. ALL frequently begins in utero with the emergence of clinically silent, preleukemic cells. Underlying leukemia-predisposing germline and acquired somatic mutations define distinct ALL subtypes that vary dramatically in treatment outcomes. In addition to genetic predisposition, a second hit, which usually occurs postnatally, is required for development of overt leukemia in most ALL subtypes. An untrained, dysregulated immune response, possibly due to an abnormal response to infection, may be an important co-factor triggering the onset of leukemia. Furthermore, the involvement of natural killer (NK) cells and T helper (Th) cells in controlling the preleukemic cells has been discussed. Identifying the cell of origin of the preleukemia-initiating event might give additional insights into potential options for prevention. Modulation of the immune system to achieve prolonged immunosurveillance of the preleukemic clone that eventually dies out in later years might present a future directive. Herein, we review the concepts of prenatal origin as well as potential preventive approaches to pediatric B cell precursor (BCP) ALL.
Collapse
|
29
|
Liu L, Cheng X, Yang H, Lian S, Jiang Y, Liang J, Chen X, Mo S, Shi Y, Zhao S, Li J, Jiang R, Yang DH, Wu Y. BCL-2 expression promotes immunosuppression in chronic lymphocytic leukemia by enhancing regulatory T cell differentiation and cytotoxic T cell exhaustion. Mol Cancer 2022; 21:59. [PMID: 35193595 PMCID: PMC8862474 DOI: 10.1186/s12943-022-01516-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) results in increased susceptibility to infections. T cell dysfunction is not associated with CLL in all patients; therefore, it is important to identify CLL patients with T cell defects. The role of B-cell lymphoma-2 (BCL-2) in CLL has been explored; however, few studies have examined its role in T cells in CLL patients. Herein, we have investigated the regulatory role of BCL-2 in T cells in the CLL tumor microenvironment. METHODS The expression of BCL-2 in T cells was evaluated using flow cytometry. The regulatory roles of BCL-2 were investigated using single-cell RNA sequencing (scRNA-seq) and verified using multi-parameter flow cytometry on CD4 and CD8 T cells. The clinical features of BCL-2 expression in T cells in CLL were also explored. RESULTS We found a significant increase in BCL-2 expression in the T cells of CLL patients (n = 266). Single cell RNA sequencing (scRNA-seq) indicated that BCL-2+CD4+ T cells had the gene signature of increased regulatory T cells (Treg); BCL-2+CD8+ T cells showed the gene signature of exhausted cytotoxic T lymphocytes (CTL); and increased expression of BCL-2 was associated with T cell activation and cellular adhesion. The results from scRNA-seq were verified in peripheral T cells from 70 patients with CLL, wherein BCL-2+CD4+ T cells were enriched with Tregs and had higher expression of interleukin-10 and transforming growth factor-β than BCL-2-CD4+ T cells. BCL-2 expression in CD8+T cells was associated with exhausted cells (PD-1+Tim-3+) and weak expression of granzyme B and perforin. T cell-associated cytokine profiling revealed a negative association between BCL-2+ T cells and T cell activation. Decreased frequencies and recovery functions of BCL-2+T cells were observed in CLL patients in complete remission after treatment with venetoclax. CONCLUSION BCL-2 expression in the T cells of CLL patients is associated with immunosuppression via promotion of Treg abundance and CTL exhaustion.
Collapse
Affiliation(s)
- Lu Liu
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Xianfeng Cheng
- Department of Clinical laboratory, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Hui Yang
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Senlin Lian
- Jiangsu Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China.,State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuegen Jiang
- Department of Clinical laboratory, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Jinhua Liang
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Xiao Chen
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Suo Mo
- Department of Clinical laboratory, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yu Shi
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Sishu Zhao
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Jianyong Li
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Runqiu Jiang
- Jiangsu Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China. .,State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China. .,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Dong-Hua Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Yujie Wu
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China. .,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
30
|
Geron I, Savino AM, Fishman H, Tal N, Brown J, Turati VA, James C, Sarno J, Hameiri-Grossman M, Lee YN, Rein A, Maniriho H, Birger Y, Zemlyansky A, Muler I, Davis KL, Marcu-Malina V, Mattson N, Parnas O, Wagener R, Fischer U, Barata JT, Jamieson CHM, Müschen M, Chen CW, Borkhardt A, Kirsch IR, Nagler A, Enver T, Izraeli S. An instructive role for Interleukin-7 receptor α in the development of human B-cell precursor leukemia. Nat Commun 2022; 13:659. [PMID: 35115489 PMCID: PMC8814001 DOI: 10.1038/s41467-022-28218-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Kinase signaling fuels growth of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Yet its role in leukemia initiation is unclear and has not been shown in primary human hematopoietic cells. We previously described activating mutations in interleukin-7 receptor alpha (IL7RA) in poor-prognosis "ph-like" BCP-ALL. Here we show that expression of activated mutant IL7RA in human CD34+ hematopoietic stem and progenitor cells induces a preleukemic state in transplanted immunodeficient NOD/LtSz-scid IL2Rγnull mice, characterized by persistence of self-renewing Pro-B cells with non-productive V(D)J gene rearrangements. Preleukemic CD34+CD10highCD19+ cells evolve into BCP-ALL with spontaneously acquired Cyclin Dependent Kinase Inhibitor 2 A (CDKN2A) deletions, as commonly observed in primary human BCP-ALL. CRISPR mediated gene silencing of CDKN2A in primary human CD34+ cells transduced with activated IL7RA results in robust development of BCP-ALLs in-vivo. Thus, we demonstrate that constitutive activation of IL7RA can initiate preleukemia in primary human hematopoietic progenitors and cooperates with CDKN2A silencing in progression into BCP-ALL.
Collapse
MESH Headings
- Animals
- Antigens, CD34/genetics
- Antigens, CD34/immunology
- Antigens, CD34/metabolism
- Base Sequence
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/immunology
- Cyclin-Dependent Kinase Inhibitor p16/metabolism
- Gene Expression/immunology
- Humans
- Interleukin-7 Receptor alpha Subunit/genetics
- Interleukin-7 Receptor alpha Subunit/immunology
- Interleukin-7 Receptor alpha Subunit/metabolism
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cells, B-Lymphoid/immunology
- Precursor Cells, B-Lymphoid/metabolism
- RNA-Seq/methods
- Receptors, Cytokine/genetics
- Receptors, Cytokine/immunology
- Receptors, Cytokine/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- Single-Cell Analysis/methods
- Transplantation, Heterologous
- Mice
Collapse
Affiliation(s)
- Ifat Geron
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Angela Maria Savino
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Hila Fishman
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Noa Tal
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - John Brown
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | | | - Chela James
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Jolanda Sarno
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Stanford University, Stanford, CA, USA
| | - Michal Hameiri-Grossman
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Yu Nee Lee
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Pediatric Department and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital Sheba Medical Center, Tel-Hashomer, Israel
| | - Avigail Rein
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Hillary Maniriho
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Yehudit Birger
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Anna Zemlyansky
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Inna Muler
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Kara L Davis
- Department of Pediatrics, Bass Center for Childhood Cancer and Blood Disorders, Stanford University, Stanford, CA, USA
| | - Victoria Marcu-Malina
- Cytogenetic Unit laboratory of Hematology, Chaim Sheba Medical Center Tel Hashomer, Tel Hashomer, Israel
| | - Nicole Mattson
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Oren Parnas
- The Concern Foundation Laboratories at the Lautenberg Center for immunology and Cancer Research, IMRIC, Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Rabea Wagener
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Catriona H M Jamieson
- UC San Diego, Moores Cancer Center, Division of Regenerative Medicine, Department of Medicine and Sanford Stem Cell Clinical Center, Ja Jolla, CA, USA
| | - Markus Müschen
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Chun-Wei Chen
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Arnon Nagler
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel
- Hematology Division BMT and Cord Blood Bank Chaim Sheba Medical Center Tel-Hashomer, Tel-Hashomer, Israel
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, UCL, London, UK
| | - Shai Izraeli
- Felsenstein Medical Research Center and The Molecular Genetics and Biochemistry Department, Sackler Faculty of Medicine, Tel Aviv University, Petach Tikva, Israel.
- Institute of Pediatric Research, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel.
- The Rina Zaizov Pediatric Hematology and Oncology Division Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA.
| |
Collapse
|