1
|
Corinaldesi C, Holmes AB, Martire G, Tosato A, Rizzato D, Lovisa F, Gallingani I, Shen Q, Ferrone L, Harris M, Davies K, Molinaro L, Mortara U, Dei Tos AP, Ofori K, D'Amore ESG, Chiarle R, Ngan B, Carraro E, Pillon M, Hussein S, Bhagat G, Pizzi M, Mussolin L, Basso K. Single-cell transcriptomics of pediatric Burkitt lymphoma reveals intra-tumor heterogeneity and markers of therapy resistance. Leukemia 2025; 39:189-198. [PMID: 39424708 PMCID: PMC11717704 DOI: 10.1038/s41375-024-02431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
Burkitt lymphoma (BL) is the most frequent B-cell lymphoma in pediatric patients. While most patients are cured, a fraction of them are resistant to therapy. To investigate BL heterogeneity and the features distinguishing therapy responders (R) from non-responders (NR), we analyzed by single-cell (sc)-transcriptomics diagnostic EBV-negative BL specimens. Analysis of the non-tumor component revealed a predominance of immune cells and a small representation of fibroblasts, enriched in NR. Tumors displayed patient-specific features, as well as shared subpopulations that expressed transcripts related to cell cycle, signaling pathways and cell-of-origin signatures. Several transcripts were differentially expressed in R versus NR. The top candidate, Tropomyosin 2 (TPM2), a member of the tropomyosin actin filament binding protein family, was confirmed to be significantly higher in NR both at the transcript and protein level. Stratification of patients based on TPM2 expression at diagnosis significantly correlated with prognosis, independently of TP53 mutations. These results indicate that BL displays transcriptional heterogeneity and identify candidate biomarkers of therapy resistance.
Collapse
Affiliation(s)
| | - Antony B Holmes
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Gaia Martire
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Anna Tosato
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Domenico Rizzato
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
| | - Federica Lovisa
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Ilaria Gallingani
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Qiong Shen
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Lavinia Ferrone
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Marian Harris
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Luca Molinaro
- Department of Medical Science, University of Torino, Torino, Italy
| | - Umberto Mortara
- Department of Medical Science, University of Torino, Torino, Italy
| | - Angelo Paolo Dei Tos
- General Pathology and Cytopathology Unit, Department of Medicine-DMED, University-Hospital of Padova, Padova, Italy
| | - Kenneth Ofori
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | | | - Roberto Chiarle
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- European Institute of Oncology IRCCS, Division of Hematopathology, Milan, Italy
| | - Bo Ngan
- Hospital for Sick Children (SickKids), Toronto, ON, Canada
| | - Elisa Carraro
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
| | - Marta Pillon
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
| | - Shafinaz Hussein
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Govind Bhagat
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Marco Pizzi
- General Pathology and Cytopathology Unit, Department of Medicine-DMED, University-Hospital of Padova, Padova, Italy
| | - Lara Mussolin
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy.
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy.
| | - Katia Basso
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Solares S, León J, García-Gutiérrez L. The Functional Interaction Between Epstein-Barr Virus and MYC in the Pathogenesis of Burkitt Lymphoma. Cancers (Basel) 2024; 16:4212. [PMID: 39766110 PMCID: PMC11674381 DOI: 10.3390/cancers16244212] [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] [Received: 09/26/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The Epstein-Barr virus (EBV) is associated with a wide range of diseases, malignant and non-malignant. EBV was, in fact, the first virus described with cell transformation capacity, discovered by Epstein in 1964 in lymphoma samples from African children. Since then, EBV has been associated with several human tumors including nasopharyngeal carcinoma, gastric carcinoma, T-cell lymphoma, Hodgkin lymphoma, diffuse large B cell lymphoma, and Burkitt lymphoma among others. The molecular hallmark of Burkitt lymphoma (BL) is a chromosomal translocation that involves the MYC gene and immunoglobulin loci, resulting in the deregulated expression of MYC, an oncogenic transcription factor that appears deregulated in about half of human tumors. The role of MYC in lymphoma is well established, as MYC overexpression drives B cell proliferation through multiple mechanisms, foremost, the stimulation of the cell cycle. Indeed, MYC is found overexpressed or deregulated in several non-Hodgkin lymphomas. Most endemic and many sporadic BLs are associated with EBV infection. While some mechanisms by which EBV can contribute to BL have been reported, the mechanism that links MYC translocation and EBV infection in BL is still under debate. Here, we review the main EBV-associated diseases, with a special focus on BL, and we discuss the interaction of EBV and MYC translocation during B cell malignant transformation in BL.
Collapse
Affiliation(s)
| | | | - Lucía García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria, Departamento de Biología Molecular, Universidad de Cantabria-CSIC, Albert Einstein 22, 39011 Cantabria, Spain; (S.S.); (J.L.)
| |
Collapse
|
3
|
Fang H, Wang W, Medeiros LJ. Burkitt lymphoma. Hum Pathol 2024:105703. [PMID: 39662784 DOI: 10.1016/j.humpath.2024.105703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Burkitt lymphoma is a mature aggressive B-cell neoplasm with distinctive clinical and morphologic features, a germinal center B-cell immunophenotype, a high proliferation index and MYC rearrangement with an immunoglobulin gene partner. Initially described in equatorial Africa by a surgeon, Denis Burkitt, African (endemic) Burkitt lymphoma was the first neoplasm shown to be associated with a virus, Epstein-Barr virus (EBV), and the first neoplasm shown to be associated with a chromosomal translocation, IGH::MYC. In this article, we provide a brief historical introduction of Burkitt lymphoma, followed by a review of all aspects of this neoplasm including pathogenesis, clinical presentation, morphology, immunophenotype, cytogenetics and molecular findings. We also provide recent updates of this entity, including advances in our understanding of molecular pathogenesis of Burkitt lymphoma and the recent proposal in the current World Health Organization classification that the traditional epidemiologic variants of Burkitt lymphoma are better replaced by presence or absence of EBV infection. We also discuss the differential diagnosis of Burkitt lymphoma and how this neoplasm can be distinguished from reactive conditions and other aggressive B-cell lymphomas/leukemias. Given its very rapid growth and the unique treatment approach employed to treat these patients, it is important to recognize Burkitt lymphoma to facilitate appropriate therapy.
Collapse
Affiliation(s)
- Hong Fang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Shi A, Yun F, Shi L, Liu X, Jia Y. Research progress on the mechanism of common inflammatory pathways in the pathogenesis and development of lymphoma. Ann Med 2024; 56:2329130. [PMID: 38489405 PMCID: PMC10946270 DOI: 10.1080/07853890.2024.2329130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/25/2024] [Indexed: 03/17/2024] Open
Abstract
In recent years, the incidence and mortality rates of lymphoma have gradually increased worldwide. Tumorigenesis and drug resistance are closely related to intracellular inflammatory pathways in lymphoma. Therefore, understanding the biological role of inflammatory pathways and their abnormal activation in relation to the development of lymphoma and their selective modulation may open new avenues for targeted therapy of lymphoma. The biological functions of inflammatory pathways are extensive, and they are central hubs for regulating inflammatory responses, immune responses, and the tumour immune microenvironment. However, limited studies have investigated the role of inflammatory pathways in lymphoma development. This review summarizes the relationship between abnormal activation of common inflammatory pathways and lymphoma development to identify precise and efficient targeted therapeutic options for patients with advanced, drug-resistant lymphoma.
Collapse
Affiliation(s)
- Aorong Shi
- Department of Pathology, Basic Medical Sciences College, Inner Mongolia Medical University, Hohhot, China
| | - Fen Yun
- Department of Pathology, Basic Medical Sciences College, Inner Mongolia Medical University, Hohhot, China
- Department of Pathology, The First Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Lin Shi
- Department of Pathology, Basic Medical Sciences College, Inner Mongolia Medical University, Hohhot, China
- Department of Pathology, The First Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Xia Liu
- Department of Pathology, Basic Medical Sciences College, Inner Mongolia Medical University, Hohhot, China
- Department of Pathology, The First Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Yongfeng Jia
- Department of Pathology, Basic Medical Sciences College, Inner Mongolia Medical University, Hohhot, China
- Department of Pathology, The First Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| |
Collapse
|
5
|
Guiyedi K, Parquet M, Aoufouchi S, Chauzeix J, Rizzo D, Al Jamal I, Feuillard J, Gachard N, Peron S. Increased c-MYC Expression Associated with Active IGH Locus Rearrangement: An Emerging Role for c-MYC in Chronic Lymphocytic Leukemia. Cancers (Basel) 2024; 16:3749. [PMID: 39594704 PMCID: PMC11592262 DOI: 10.3390/cancers16223749] [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: 10/11/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
This review examines the pivotal role of c-MYC in Chronic Lymphocytic Leukemia (CLL), focusing on how its overexpression leads to increased genetic instability, thereby accelerating disease progression. MYC, a major oncogene, encodes a transcription factor that regulates essential cellular processes, including cell cycle control, proliferation, and apoptosis. In CLL cases enriched with unmutated immunoglobulin heavy chain variable (IGHV) genes, MYC is significantly overexpressed and associated with active rearrangements in the IGH immunoglobulin heavy chain locus. This overexpression results in substantial DNA damage, including double-strand breaks, chromosomal translocations, and an increase in abnormal repair events. Consequently, c-MYC plays a dual role in CLL: it promotes aggressive cell proliferation while concurrently driving genomic instability through its involvement in genetic recombination. This dynamic contributes not only to CLL progression but also to the overall aggressiveness of the disease. Additionally, the review suggests that c-MYC's influence on genetic rearrangements makes it an attractive target for therapeutic strategies aimed at mitigating CLL malignancy. These findings underscore c-MYC's critical importance in advancing CLL progression, highlighting the need for further research to explore its potential as a target in future treatment approaches.
Collapse
Affiliation(s)
- Kenza Guiyedi
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
| | - Milène Parquet
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
| | - Said Aoufouchi
- Gustave Roussy, B-Cell and Genome Plasticity Team, CNRS UMR9019, Villejuif, France and Université Paris-Saclay, 91400 Orsay, France
| | - Jasmine Chauzeix
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - David Rizzo
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - Israa Al Jamal
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Faculty of Sciences, GSBT Genomic Surveillance and Biotherapy Team, Mont Michel Campus, Lebanese University, Tripoli 1300, Lebanon
| | - Jean Feuillard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - Nathalie Gachard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - Sophie Peron
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
| |
Collapse
|
6
|
Zhou Y, Lou J, Tian Y, Ding J, Wang X, Tang B. How lactate affects immune strategies in lymphoma. Front Mol Biosci 2024; 11:1480884. [PMID: 39464313 PMCID: PMC11502318 DOI: 10.3389/fmolb.2024.1480884] [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] [Received: 08/14/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Tumor cells undergo metabolic reprogramming through shared pathways, resulting in a hypoxic, acidic, and highly permeable internal tumor microenvironment (TME). Lactate, once only regarded as a waste product of glycolysis, has an inseparable dual role with tumor immunity. It can not only provide a carbon source for immune cells to enhance immunity but also help the immune escape through a variety of ways. Lymphoma also depends on the proliferation signal of TME. This review focuses on the dynamic process of lactate metabolism and immune function changes in lymphoma and aims to comprehensively summarize and explore which genes, transcription factors, and pathways affect the biological changes and functions of immune cells. To deeply understand the complex and multifaceted role of lactate metabolism and immunity in lymphoma, the combination of lactate targeted therapy and classical immunotherapy will be a promising development direction in the future.
Collapse
Affiliation(s)
- Yuehan Zhou
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinzhan Lou
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuqin Tian
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinlei Ding
- Department of Thoracic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaobo Wang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Tang
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Kläsener K, Herrmann N, Håversen L, Sundell T, Sundqvist M, Lundqvist C, Manna PT, Jonsson CA, Visentini M, Ljung Sass D, McGrath S, Grimstad K, Aranburu A, Mellgren K, Fogelstrand L, Forsman H, Ekwall O, Borén J, Gjertsson I, Reth M, Mårtensson I, Camponeschi A. Targeting CD38 with monoclonal antibodies disrupts key survival pathways in paediatric Burkitt's lymphoma malignant B cells. Clin Transl Immunology 2024; 13:e70011. [PMID: 39364393 PMCID: PMC11447455 DOI: 10.1002/cti2.70011] [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] [Received: 02/27/2024] [Revised: 08/23/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
Objectives Paediatric Burkitt's lymphoma (pBL) is the most common childhood non-Hodgkin B-cell lymphoma. Despite the encouraging survival rates for most children, treating cases with relapse/resistance to current therapies remains challenging. CD38 is a transmembrane protein highly expressed in pBL. This study investigates the effectiveness of CD38-targeting monoclonal antibodies (mAbs), daratumumab and isatuximab, in impairing crucial cellular processes and survival pathways in pBL malignant cells. Methods In silico analyses of patient samples, combined with in vitro experiments using the Ramos cell line, were conducted to assess the impact of daratumumab and isatuximab on cellular proliferation, apoptosis and the phosphoinositide 3-kinase (PI3K) pathway. Results Isatuximab was found to be more effective than daratumumab in disrupting B-cell receptor signalling, reducing cellular proliferation and inducing apoptosis. Additionally, isatuximab caused a significant impairment of the PI3K pathway and induced metabolic reprogramming in pBL cells. The study also revealed a correlation between CD38 and MYC expression levels in pBL patient samples, suggesting CD38 involvement in key oncogenic processes. Conclusion The study emphasises the therapeutic potential of CD38-targeting mAbs, particularly isatuximab, in pBL.
Collapse
Affiliation(s)
- Kathrin Kläsener
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center FreiburgFreiburgGermany
- Signaling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
| | - Nadja Herrmann
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center FreiburgFreiburgGermany
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Liliana Håversen
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Timothy Sundell
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Martina Sundqvist
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Christina Lundqvist
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Paul T Manna
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Charlotte A Jonsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Marcella Visentini
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Diana Ljung Sass
- Department of Pediatric Hematology and Oncology, The Queen Silvia's Hospital for Children and AdolescentsUniversity of GothenburgGothenburgSweden
| | - Sarah McGrath
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Kristoffer Grimstad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- School of BioscienceUniversity of SkövdeSkövdeSweden
| | - Alaitz Aranburu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Karin Mellgren
- Department of Pediatric Hematology and Oncology, The Queen Silvia's Hospital for Children and AdolescentsUniversity of GothenburgGothenburgSweden
| | - Linda Fogelstrand
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical ChemistrySahlgrenska University HospitalGothenburgSweden
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Olov Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Rheumatology, Region Västra GötalandSahlgrenska University HospitalGothenburgSweden
| | - Michael Reth
- Department of Rheumatology and Clinical ImmunologyUniversity Medical Center FreiburgFreiburgGermany
- Signaling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
| | - Inga‐Lill Mårtensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical Immunology and Transfusion Medicine, Region Västra GötalandSahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
8
|
Liu W, Li S, Yang M, Ma J, Liu L, Fei P, Xiang Q, Huang L, Zhao P, Yang Z, Zhu X. Dysfunction of Calcyphosine-Like gene impairs retinal angiogenesis through the MYC axis and is associated with familial exudative vitreoretinopathy. eLife 2024; 13:RP96907. [PMID: 39264149 PMCID: PMC11392532 DOI: 10.7554/elife.96907] [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] [Indexed: 09/13/2024] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a severe genetic disorder characterized by incomplete vascularization of the peripheral retina and associated symptoms that can lead to vision loss. However, the underlying genetic causes of approximately 50% of FEVR cases remain unknown. Here, we report two heterozygous variants in calcyphosine-like gene (CAPSL) that is associated with FEVR. Both variants exhibited compromised CAPSL protein expression. Vascular endothelial cell (EC)-specific inactivation of Capsl resulted in delayed radial/vertical vascular progression, compromised endothelial proliferation/migration, recapitulating the human FEVR phenotypes. CAPSL-depleted human retinal microvascular endothelial cells (HRECs) exhibited impaired tube formation, decreased cell proliferation, disrupted cell polarity establishment, and filopodia/lamellipodia formation, as well as disrupted collective cell migration. Transcriptomic and proteomic profiling revealed that CAPSL abolition inhibited the MYC signaling axis, in which the expression of core MYC targeted genes were profoundly decreased. Furthermore, a combined analysis of CAPSL-depleted HRECs and c-MYC-depleted human umbilical vein endothelial cells uncovered similar transcription patterns. Collectively, this study reports a novel FEVR-associated candidate gene, CAPSL, which provides valuable information for genetic counseling of FEVR. This study also reveals that compromised CAPSL function may cause FEVR through MYC axis, shedding light on the potential involvement of MYC signaling in the pathogenesis of FEVR.
Collapse
Affiliation(s)
- Wenjing Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shujin Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Mu Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Jie Ma
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianchun Xiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lulin Huang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenglin Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- Jinfeng Laboratory, Chongqing, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
- Jinfeng Laboratory, Chongqing, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
9
|
Wu S, Zhang J, Chen S, Zhou X, Liu Y, Hua H, Qi X, Mao Y, Young KH, Lu T. Low NDRG2, regulated by the MYC/MIZ-1 complex and methylation, predicts poor outcomes in DLBCL patients. Ann Hematol 2024; 103:2877-2892. [PMID: 38842567 DOI: 10.1007/s00277-024-05829-2] [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: 10/12/2023] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) represents the most common tumor in non-Hodgkin's lymphoma. N-Myc downstream-regulated gene 2 (NDRG2) is a tumor suppressor highly expressed in healthy tissues but downregulated in many cancers. Although cell proliferation-related metabolism rewiring has been well characterized, less is known about the mechanism of metabolic changes with DLBCL. Herein, we investigated the expressions of NDRG2, MYC and Myc-interacting zinc finger protein 1 (MIZ-1) in seven human lymphoma (mostly DLBCLs) cell lines. NDRG2 expression was inversely correlated with the expressions of MYC and MIZ-1. Further, we explored the regulatory mechanism and biological functions underlying the lymphomagenesis involving NDRG2, MYC and MIZ-1. MYC and MIZ-1 promoted DLBCL cell proliferation, while NDRG2 induced apoptosis in LY8 cells. Moreover, NDRG2 methylation was reversed by the 5-Aza-2'-deoxycytidine (5-Aza-CDR) treatment, triggering the downregulation of MYC and inhibiting DLBCL cell survival. MYC interacts with NDRG2 to regulate energy metabolism associated with mTOR. Remarkably, supporting the biological significance, the converse correlation between NDRG2 and MYC was observed in human DLBCL tumor tissues (R = -0.557). Bioinformatics analysis further validated the association among NDRG2, MYC, MIZ-1, mTOR, and related metabolism genes. Additionally, NDRG2 (P = 0.001) and MYC (P < 0.001) were identified as promising prognostic biomarkers in DLBCL patients through survival analysis. Together, our data demonstrate that the MYC/MIZ-1 complex interplays with NDRG2 to influence the proliferation and apoptosis of DLBCL cells and show the characterizations of NDRG2, MYC and MIZ-1 for metabolism features and prediction prognosis in DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Gene Expression Regulation, Neoplastic
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Male
- Prognosis
- Cell Line, Tumor
- Female
- Middle Aged
- DNA Methylation
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Aged
- Cell Proliferation
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Shuang Wu
- Department of Hematology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Jie Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- Department of Oncology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Shan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- Department of Oncology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Xinyi Zhou
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Yankui Liu
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Haiying Hua
- Department of Hematology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Xiaowei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Yong Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- Department of Oncology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu Province, China
| | - Ken H Young
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, 27710, USA
| | - Tingxun Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
- Department of Oncology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu Province, China.
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
10
|
Sureda-Gómez M, Iaccarino I, De Bolòs A, Meyer M, Balsas P, Richter J, Rodríguez ML, López C, Carreras-Caballé M, Glaser S, Nadeu F, Jares P, Clot G, Siciliano MC, Bellan C, Tornambè S, Boccacci R, Leoncini L, Campo E, Siebert R, Amador V, Klapper W. SOX11 expression is restricted to EBV-negative Burkitt lymphoma and is associated with molecular genetic features. Blood 2024; 144:187-200. [PMID: 38620074 DOI: 10.1182/blood.2023023242] [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: 11/16/2023] [Revised: 02/27/2024] [Accepted: 03/21/2024] [Indexed: 04/17/2024] Open
Abstract
ABSTRACT SRY-related HMG-box gene 11 (SOX11) is a transcription factor overexpressed in mantle cell lymphoma (MCL), a subset of Burkitt lymphomas (BL) and precursor lymphoid cell neoplasms but is absent in normal B cells and other B-cell lymphomas. SOX11 has an oncogenic role in MCL but its contribution to BL pathogenesis remains uncertain. Here, we observed that the presence of Epstein-Barr virus (EBV) and SOX11 expression were mutually exclusive in BL. SOX11 expression in EBV-negative (EVB-) BL was associated with an IG∷MYC translocation generated by aberrant class switch recombination, whereas in EBV-negative (EBV-)/SOX11-negative (SOX11-) tumors the IG∷MYC translocation was mediated by mistaken somatic hypermutations. Interestingly, EBV- SOX11-expressing BL showed higher frequency of SMARCA4 and ID3 mutations than EBV-/SOX11- cases. By RNA sequencing, we identified a SOX11-associated gene expression profile, with functional annotations showing partial overlap with the SOX11 transcriptional program of MCL. Contrary to MCL, no differences on cell migration or B-cell receptor signaling were found between SOX11- and SOX11-positive (SOX11+) BL cells. However, SOX11+ BL showed higher adhesion to vascular cell adhesion molecule 1 (VCAM-1) than SOX11- BL cell lines. Here, we demonstrate that EBV- BL comprises 2 subsets of cases based on SOX11 expression. The mutual exclusion of SOX11 and EBV, and the association of SOX11 with a specific genetic landscape suggest a role of SOX11 in the early pathogenesis of BL.
Collapse
Affiliation(s)
- Marta Sureda-Gómez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Ingram Iaccarino
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Kiel, Kiel, Germany
| | - Anna De Bolòs
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Mieke Meyer
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Kiel, Kiel, Germany
| | - Patricia Balsas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Julia Richter
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Kiel, Kiel, Germany
| | | | - Cristina López
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | | | - Selina Glaser
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Ferran Nadeu
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Pedro Jares
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Department of Hematology Hospital Clinic of Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Guillem Clot
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- University of Barcelona, Barcelona, Spain
| | - Maria Chiara Siciliano
- Department of Medical Biotechnologies, Section of Pathological Anatomy, University of Siena, Siena, Italy
| | - Cristiana Bellan
- Department of Medical Biotechnologies, Section of Pathological Anatomy, University of Siena, Siena, Italy
| | - Salvatore Tornambè
- Department of Medical Biotechnologies, Section of Pathological Anatomy, University of Siena, Siena, Italy
| | - Roberto Boccacci
- Department of Medical Biotechnologies, Section of Pathological Anatomy, University of Siena, Siena, Italy
| | - Lorenzo Leoncini
- Department of Medical Biotechnologies, Section of Pathological Anatomy, University of Siena, Siena, Italy
| | - Elias Campo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Department of Hematology Hospital Clinic of Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Virginia Amador
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Kiel, Kiel, Germany
| |
Collapse
|
11
|
Yurtdas ZY, Kilic E, Boor P, Wyler E, Landthaler M, Jung K, Schmidt-Ott KM. Grainyhead-like 2 Deficiency and Kidney Cyst Growth in a Mouse Model. J Am Soc Nephrol 2024; 35:838-853. [PMID: 38656794 PMCID: PMC11230724 DOI: 10.1681/asn.0000000000000353] [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: 04/01/2023] [Accepted: 04/08/2024] [Indexed: 04/26/2024] Open
Abstract
Key Points Our study reveals segment-specific mechanisms in cystic kidney disease and suggests Grhl2 as a modifier of collecting duct–derived cyst progression. Our data demonstrate that genetic deletion of Grhl2 accelerates disease progression in a cystic mouse model. Background The transcription factor grainyhead-like 2 (GRHL2) plays a crucial role in maintaining the epithelial barrier properties of the kidney collecting duct and is important to osmoregulation. We noticed a reduction in GRHL2 expression in cysts derived from the collecting ducts in kidneys affected by autosomal dominant polycystic kidney disease (ADPKD). However, the specific role of GRHL2 in cystic kidney disease remains unknown. Methods The functional role of the transcription factor Grhl2 in the context of cystic kidney disease was examined through analysis of its expression pattern in patient samples with ADPKD and generating a transgenic cystic kidney disease (TCKD) mouse model by overexpressing the human proto-oncogene c-MYC in kidney collecting ducts. Next, TCKD mice bred with collecting duct–specific Grhl2 knockout mice (Grhl2KO). The resulting TCKD-Grhl2 KO mice and their littermates were examined by various types of histological and biochemical assays and gene profiling analysis through RNA sequencing. Results A comprehensive examination of kidney samples from patients with ADPKD revealed GRHL2 downregulation in collecting duct–derived cyst epithelia. Comparative analysis of TCKD and TCKD-Grhl2 KO mice exhibited that the collecting duct–specific deletion of Grhl2 resulted in markedly aggravated cyst growth, worsened kidney dysfunction, and shortened life span. Furthermore, transcriptomic analyses indicated sequential downregulation of kidney epithelial cyst development regulators (Frem2 , Muc1 , Cdkn2c , Pkd2 , and Tsc1 ) during cyst progression in kidneys of TCKD-Grhl2 KO mice, which included presumed direct Grhl2 target genes. Conclusions These results suggest Grhl2 as a potential progression modifier, especially for cysts originating from collecting ducts.
Collapse
Affiliation(s)
- Zeliha Yesim Yurtdas
- Molecular and Translational Kidney Research, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute for Urologic Research, Berlin, Germany
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ergin Kilic
- Medical School Hamburg, Department of Pathology, Hamburg, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute für Biologie, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klaus Jung
- Berlin Institute for Urologic Research, Berlin, Germany
- Department of Urology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kai M. Schmidt-Ott
- Molecular and Translational Kidney Research, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Wang JN, Zheng G, Wu W, Huang H. Follicular helper T cells: emerging roles in lymphomagenesis. J Leukoc Biol 2024; 116:54-63. [PMID: 37939814 DOI: 10.1093/jleuko/qiad140] [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: 04/29/2023] [Revised: 08/11/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
Follicular helper T cells are a subset of CD4+ T cells that are fundamental to forming germinal centers, which are the primary sites of antibody affinity maturation and the proliferation of activated B cells. Follicular helper T cells have been extensively studied over the past 10 years, especially regarding their roles in cancer genesis. This review describes the characteristics of normal follicular helper T cells and focuses on the emerging link between follicular helper T cells and lymphomagenesis. Advances in lymphoma genetics have substantially expanded our understanding of the role of follicular helper T cells in lymphomagenesis. Moreover, we detail a range of agents and new therapies, with a major focus on chimeric antigen receptor T-cell therapy; these novel approaches may offer new treatment opportunities for patients with lymphomas.
Collapse
Affiliation(s)
- Ji-Nuo Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| | - Gaofeng Zheng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| | - Wenjun Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| |
Collapse
|
13
|
Magazzino F, Aristei C, Passarelli A, Pierini A, De Giorgi U, Martinello R, Domenici L, Pignata S, Mangili G, Cormio G. Lymphomas of the Vulva: A Review of the MITO Rare Cancer Group. Cancers (Basel) 2024; 16:2102. [PMID: 38893221 PMCID: PMC11171216 DOI: 10.3390/cancers16112102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Since they are very rare tumors, lymphomas of the vulva are often not properly recognized. Patients with vulvar lymphoma are generally elderly and the classical manifestation of the disease is a vulvar mass. No significant age differences have been found between primary and secondary lymphoma. To make a correct diagnosis, it is therefore necessary to use not only histological examination but also the genetic and molecular profile in order to establish optimal therapeutic management. Literature analysis confirm the good prognosis of this disease.
Collapse
Affiliation(s)
- Francescapaola Magazzino
- Complex Operating Unit Obstetrics and Gynaecology, Ospedale Civile di San Donà di Piave-Venezia, AULSS4 Veneto Orientale, 30027 San Donà di Piave, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Medicine and Surgery, Perugia General Hospital Sant’Andrea delle Fratte, University of Perugia, 06156 Perugia, Italy;
| | - Anna Passarelli
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80144 Napoli, Italy; (A.P.); (S.P.)
| | - Antonio Pierini
- Division of Hematolgy and Clinical Immunolgy, Department of Medicine and Surgery, University of Perugia, 06156 Perugia, Italy;
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Ruby Martinello
- Department of Medical Sciences, Institute of Obstetrics and Gynecology, University of Ferrara, 44121 Ferrara, Italy;
| | - Lavinia Domenici
- 2nd Division of Obstetrics and Gynaecology, Azienda Ospedaliera Universitaria Pisana, University of Pisa, 56126 Pisa, Italy;
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80144 Napoli, Italy; (A.P.); (S.P.)
| | - Giorgia Mangili
- Department of Obstetrics and Gynaecology, San Raffaele Scientific Institute, 20132 Milano, Italy;
| | - Gennaro Cormio
- Gynecologic Oncoly Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
- Department of Interdisciplinary Medicine, University of Bari, 70124 Bari, Italy
| |
Collapse
|
14
|
Witte H, Künstner A, Gebauer N. Update: The molecular spectrum of virus-associated high-grade B-cell non-Hodgkin lymphomas. Blood Rev 2024; 65:101172. [PMID: 38267313 DOI: 10.1016/j.blre.2024.101172] [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: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
The vast spectrum of aggressive B-cell non-Hodgkin neoplasms (B-NHL) encompasses several infrequent entities occurring in association with viral infections, posing diagnostic challenges for practitioners. In the emerging era of precision oncology, the molecular characterization of malignancies has acquired paramount significance. The pathophysiological comprehension of specific entities and the identification of targeted therapeutic options have seen rapid development. However, owing to their rarity, not all entities have undergone exhaustive molecular characterization. Considerable heterogeneity exists in the extant body of work, both in terms of employed methodologies and the scale of cases studied. Presently, therapeutic strategies are predominantly derived from observations in diffuse large B-cell lymphoma (DLBCL), the most prevalent subset of aggressive B-NHL. Ongoing investigations into the molecular profiles of these uncommon virus-associated entities are progressively facilitating a clearer distinction from DLBCL, ultimately paving the way towards individualized therapeutic approaches. This review consolidates the current molecular insights into aggressive and virus-associated B-NHL, taking into consideration the recently updated 5th edition of the WHO classification of hematolymphoid tumors (WHO-5HAEM) and the International Consensus Classification (ICC). Additionally, potential therapeutically targetable susceptibilities are highlighted, offering a comprehensive overview of the present scientific landscape in the field.
Collapse
Affiliation(s)
- H Witte
- Department of Hematology and Oncology, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081 Ulm, Germany; Department of Hematology and Oncology, University Hospital Schleswig-Holstein (UKSH) Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany.
| | - A Künstner
- University Cancer Center Schleswig-Holstein (UCCSH), Ratzeburger Allee 160, 23538 Lübeck, Germany; Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - N Gebauer
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein (UKSH) Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; University Cancer Center Schleswig-Holstein (UCCSH), Ratzeburger Allee 160, 23538 Lübeck, Germany
| |
Collapse
|
15
|
Mayr F, Kruse V, Fuhrmann DC, Wolf S, Löber J, Alsouri S, Paglilla N, Lee K, Chapuy B, Brüne B, Zenz T, Häupl B, Oellerich T, Engelke M. SH2 domain-containing inositol 5-phosphatases support the survival of Burkitt lymphoma cells by promoting energy metabolism. Haematologica 2024; 109:1445-1459. [PMID: 37916396 PMCID: PMC11063853 DOI: 10.3324/haematol.2023.283663] [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: 06/02/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Burkitt lymphoma cells (BL) exploit antigen-independent tonic signals transduced by the B-cell antigen receptor (BCR) for their survival, but the molecular details of the rewired BL-specific BCR signal network remain unclear. A loss of function screen revealed the SH2 domain-containing 5`-inositol phosphatase 2 (SHIP2) as a potential modulator of BL fitness. We characterized the role of SHIP2 in BL survival in several BL cell models and show that perturbing SHIP2 function renders cells more susceptible to apoptosis, while attenuating proliferation in a BCR-dependent manner. Unexpectedly, SHIP2 deficiency did neither affect PI3K survival signals nor MAPK activity, but attenuated ATP production. We found that an efficient energy metabolism in BL cells requires phosphatidylinositol-3,4-bisphosphate (PI(3,4)P2), which is the enzymatic product of SHIP proteins. Consistently, interference with the function of SHIP1 and SHIP2 augments BL cell susceptibility to PI3K inhibition. Notably, we provide here a molecular basis of how tonic BCR signals are connected to energy supply, which is particularly important for such an aggressively growing neoplasia. These findings may help to improve therapies for the treatment of BL by limiting energy metabolism through the inhibition of SHIP proteins, which renders BL cells more susceptible to the targeting of survival signals.
Collapse
Affiliation(s)
- Florian Mayr
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen
| | - Vanessa Kruse
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen
| | - Dominik C Fuhrmann
- Institute for Biochemistry I, Faculty of Medicine, Johann Wolfgang Goethe-University Frankfurt
| | - Sebastian Wolf
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt
| | - Jens Löber
- Department of Hematology, Oncology and Cancer Immunology, Charité, Campus Benjamin Franklin
| | - Saed Alsouri
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen
| | - Nadia Paglilla
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen
| | - Kwang Lee
- Translational Medical Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg
| | - Björn Chapuy
- Department of Hematology, Oncology and Cancer Immunology, Charité, Campus Benjamin Franklin
| | - Bernhard Brüne
- Institute for Biochemistry I, Faculty of Medicine, Johann Wolfgang Goethe-University Frankfurt
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital Zurich
| | - Björn Häupl
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Frankfurt Cancer Institute, Johann Wolfgang Goethe University Frankfurt, Frankfurt
| | - Thomas Oellerich
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Frankfurt Cancer Institute, Johann Wolfgang Goethe University Frankfurt, Frankfurt
| | - Michael Engelke
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen.
| |
Collapse
|
16
|
Alsouri S, Ambrose A, Mougios N, Paglilla N, Mayr F, Choi K, Loeber J, Chapuy B, Haeupl B, Opazo F, Oellerich T, Gold M, Engelke M. Actinin-4 controls survival signaling in B cells by limiting the lateral mobility of B-cell antigen receptors. Eur J Immunol 2024; 54:e2350774. [PMID: 38299456 DOI: 10.1002/eji.202350774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024]
Abstract
The structure and dynamics of F-actin networks in the cortical area of B cells control the signal efficiency of B-cell antigen receptors (BCRs). Although antigen-induced signaling has been studied extensively, the role of cortical F-actin in antigen-independent tonic BCR signaling is less well understood. Because these signals are essential for the survival of B cells and are consequently exploited by several B-cell lymphomas, we assessed how the cortical F-actin structure influences tonic BCR signal transduction. We employed genetic variants of a primary cell-like B-cell line that can be rendered quiescent to show that cross-linking of actin filaments by α-actinin-4 (ACTN4), but not ACTN1, is required to preserve the dense architecture of F-actin in the cortical area of B cells. The reduced cortical F-actin density in the absence of ACTN4 resulted in increased lateral BCR diffusion. Surprisingly, this was associated with reduced tonic activation of BCR-proximal effector proteins, extracellular signal-regulated kinase, and pro-survival pathways. Accordingly, ACTN4-deficient B-cell lines and primary human B cells exhibit augmented apoptosis. Hence, our findings reveal that cortical F-actin architecture regulates antigen-independent tonic BCR survival signals in human B cells.
Collapse
Affiliation(s)
- Saed Alsouri
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Ashley Ambrose
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Department of Mathematics, University of British Columbia, Vancouver, Canada
| | - Nikolaos Mougios
- Center for Biostructural Imaging of Neurodegeneration (BIN), Goettingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Goettingen, Goettingen, Germany
| | - Nadia Paglilla
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Florian Mayr
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Kate Choi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Jens Loeber
- Department of Hematology, Oncology and Cancer Immunology, Charité - University Medical Center Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Björn Chapuy
- Department of Hematology, Oncology and Cancer Immunology, Charité - University Medical Center Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Björn Haeupl
- Frankfurt Cancer Institute, Johann Wolfgang Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration (BIN), Goettingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas Oellerich
- Frankfurt Cancer Institute, Johann Wolfgang Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Gold
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Michael Engelke
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
17
|
Katsuragawa‐Taminishi Y, Mizutani S, Kawaji‐Kanayama Y, Onishi A, Okamoto H, Isa R, Mizuhara K, Muramatsu A, Fujino T, Tsukamoto T, Shimura Y, Taniwaki M, Miyagawa‐Hayashino A, Konishi E, Kuroda J. Triple targeting of RSK, AKT, and S6K as pivotal downstream effectors of PDPK1 by TAS0612 in B-cell lymphomas. Cancer Sci 2023; 114:4691-4705. [PMID: 37840379 PMCID: PMC10728023 DOI: 10.1111/cas.15995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/09/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
B-cell lymphomas (BCLs) are the most common disease entity among hematological malignancies and have various genetically and molecularly distinct subtypes. In this study, we revealed that the blockade of phosphoinositide-dependent kinase-1 (PDPK1), the master kinase of AGC kinases, induces a growth inhibition via cell cycle arrest and the induction of apoptosis in all eight BCL-derived cell lines examined, including those from activated B-cell-like diffuse large B-cell lymphoma (DLBCL), double expressor DLBCL, Burkitt lymphoma, and follicular lymphoma. We also demonstrated that, in these cell lines, RSK2, AKT, and S6K, but not PLK1, SGK, or PKC, are the major downstream therapeutic target molecules of PDPK1 and that RSK2 plays a central role and AKT and S6K play subsidiary functional roles as the downstream effectors of PDPK1 in cell survival and proliferation. Following these results, we confirmed the antilymphoma efficacy of TAS0612, a triple inhibitor for total RSK, including RSK2, AKT, and S6K, not only in these cell lines, regardless of disease subtypes, but also in all 25 patient-derived B lymphoma cells of various disease subtypes. At the molecular level, TAS0612 caused significant downregulation of MYC and mTOR target genes while inducing the tumor suppressor TP53INP1 protein in these cell lines. These results prove that the simultaneous blockade of RSK2, AKT, and S6K, which are the pivotal downstream substrates of PDPK1, is a novel therapeutic target for the various disease subtypes of BCLs and line up TAS0612 as an attractive candidate agent for BCLs for future clinical development.
Collapse
Affiliation(s)
- Yoko Katsuragawa‐Taminishi
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Shinsuke Mizutani
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Yuka Kawaji‐Kanayama
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Akio Onishi
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Haruya Okamoto
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Reiko Isa
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Kentaro Mizuhara
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Ayako Muramatsu
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Takahiro Fujino
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Taku Tsukamoto
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
- Department of Blood TransfusionKyoto Prefectural University of MedicineKyotoJapan
| | - Masafumi Taniwaki
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | | | - Eiichi Konishi
- Department of Surgical PathologyKyoto Prefectural University of MedicineKyotoJapan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
18
|
Wang ZQ, Zhang ZC, Wu YY, Pi YN, Lou SH, Liu TB, Lou G, Yang C. Bromodomain and extraterminal (BET) proteins: biological functions, diseases, and targeted therapy. Signal Transduct Target Ther 2023; 8:420. [PMID: 37926722 PMCID: PMC10625992 DOI: 10.1038/s41392-023-01647-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 11/07/2023] Open
Abstract
BET proteins, which influence gene expression and contribute to the development of cancer, are epigenetic interpreters. Thus, BET inhibitors represent a novel form of epigenetic anticancer treatment. Although preliminary clinical trials have shown the anticancer potential of BET inhibitors, it appears that these drugs have limited effectiveness when used alone. Therefore, given the limited monotherapeutic activity of BET inhibitors, their use in combination with other drugs warrants attention, including the meaningful variations in pharmacodynamic activity among chosen drug combinations. In this paper, we review the function of BET proteins, the preclinical justification for BET protein targeting in cancer, recent advances in small-molecule BET inhibitors, and preliminary clinical trial findings. We elucidate BET inhibitor resistance mechanisms, shed light on the associated adverse events, investigate the potential of combining these inhibitors with diverse therapeutic agents, present a comprehensive compilation of synergistic treatments involving BET inhibitors, and provide an outlook on their future prospects as potent antitumor agents. We conclude by suggesting that combining BET inhibitors with other anticancer drugs and innovative next-generation agents holds great potential for advancing the effective targeting of BET proteins as a promising anticancer strategy.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Zhao-Cong Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Yu-Yang Wu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ya-Nan Pi
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Sheng-Han Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tian-Bo Liu
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Ge Lou
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China.
| | - Chang Yang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China.
| |
Collapse
|
19
|
Gómez-Escolar C, Marina-Zárate E, Ramiro AR. Activation-induced deaminase expression defines mature B cell lymphoma in the mouse. Front Immunol 2023; 14:1268930. [PMID: 37809061 PMCID: PMC10558245 DOI: 10.3389/fimmu.2023.1268930] [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: 07/28/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Germinal centers (GCs) are the sites of secondary antibody diversification and underlie the mechanism of action of many vaccination strategies. Activation-induced deaminase (AID) triggers secondary antibody diversification through the introduction of somatic changes in immunoglobulin genes which lead to the generation of antibodies of higher affinity and more specialized effector functions. However, AID can also target other genomic regions, giving rise to mutations and chromosome translocations with oncogenic potential. Many human lymphomas originate from mature B cells that have undergone the GC reaction, such as the diffuse large B cell lymphoma, the follicular lymphoma and Burkitt lymphoma, and carry chromosome translocations. Mature B cell lymphomagenesis has been modeled in the mouse by the genetic introduction of chromosome translocations. Here, we present an in-depth characterization of one such model, λ-MYC mice. We found that young pre-tumor stage mice had a prominent block in early B cell differentiation that resulted in the generation of very aggressive tumors lacking surface B cell receptor (BCR) expression, indicating that a large fraction of tumors in λ-MYC mice arise from B cell precursors rather than from mature B cells. Further, we assessed the contribution of AID to B cell lymphomagenesis in λ-MYC mice by using a genetic tracer of historical AID expression. Only a fraction of tumors contained cells of GC origin as defined by AID expression. AID-experienced tumors associated with longer survival and resembled mature B cell lymphomas. Thus, AID expression defines Burkitt lymphomagenesis in λ-MYC mice.
Collapse
Affiliation(s)
| | | | - Almudena R. Ramiro
- B Lymphocyte Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
20
|
Pang Y, Lu T, Xu-Monette ZY, Young KH. Metabolic Reprogramming and Potential Therapeutic Targets in Lymphoma. Int J Mol Sci 2023; 24:5493. [PMID: 36982568 PMCID: PMC10052731 DOI: 10.3390/ijms24065493] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Lymphoma is a heterogeneous group of diseases that often require their metabolism program to fulfill the demand of cell proliferation. Features of metabolism in lymphoma cells include high glucose uptake, deregulated expression of enzymes related to glycolysis, dual capacity for glycolytic and oxidative metabolism, elevated glutamine metabolism, and fatty acid synthesis. These aberrant metabolic changes lead to tumorigenesis, disease progression, and resistance to lymphoma chemotherapy. This metabolic reprogramming, including glucose, nucleic acid, fatty acid, and amino acid metabolism, is a dynamic process caused not only by genetic and epigenetic changes, but also by changes in the microenvironment affected by viral infections. Notably, some critical metabolic enzymes and metabolites may play vital roles in lymphomagenesis and progression. Recent studies have uncovered that metabolic pathways might have clinical impacts on the diagnosis, characterization, and treatment of lymphoma subtypes. However, determining the clinical relevance of biomarkers and therapeutic targets related to lymphoma metabolism is still challenging. In this review, we systematically summarize current studies on metabolism reprogramming in lymphoma, and we mainly focus on disorders of glucose, amino acids, and lipid metabolisms, as well as dysregulation of molecules in metabolic pathways, oncometabolites, and potential metabolic biomarkers. We then discuss strategies directly or indirectly for those potential therapeutic targets. Finally, we prospect the future directions of lymphoma treatment on metabolic reprogramming.
Collapse
Affiliation(s)
- Yuyang Pang
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Hematology, Ninth People’s Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Tingxun Lu
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Durham, NC 27710, USA
| | - Zijun Y. Xu-Monette
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Durham, NC 27710, USA
| | - Ken H. Young
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Durham, NC 27710, USA
| |
Collapse
|
21
|
Najmi A, Thangavel N, Mohanan AT, Qadri M, Albratty M, Ashraf SE, Saleh SF, Nayeem M, Mohan S. Structural Complementarity of Bruton’s Tyrosine Kinase and Its Inhibitors for Implication in B-Cell Malignancies and Autoimmune Diseases. Pharmaceuticals (Basel) 2023; 16:ph16030400. [PMID: 36986499 PMCID: PMC10051736 DOI: 10.3390/ph16030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Bruton’s tyrosine kinase (BTK) is a critical component in B-cell receptor (BCR) signaling and is also expressed in haematogenic and innate immune cells. Inhibition of BTK hyperactivity is implicated in B-cell malignancies and autoimmune diseases. This review derives the structural complementarity of the BTK-kinase domain and its inhibitors from recent three-dimensional structures of inhibitor-bound BTK in the protein data bank (PDB). Additionally, this review analyzes BTK-mediated effector responses of B-cell development and antibody production. Covalent inhibitors contain an α, β-unsaturated carbonyl moiety that forms a covalent bond with Cys481, stabilizing αC-helix in inactive-out conformation which inhibits Tyr551 autophosphorylation. Asn484, located two carbons far from Cys481, influences the stability of the BTK-transition complex. Non-covalent inhibitors engage the BTK-kinase domain through an induced-fit mechanism independent of Cys481 interaction and bind to Tyr551 in the activation kink resulting in H3 cleft, determining BTK selectivity. Covalent and non-covalent binding to the kinase domain of BTK shall induce conformational changes in other domains; therefore, investigating the whole-length BTK conformation is necessary to comprehend BTK’s autophosphorylation inhibition. Knowledge about the structural complementarity of BTK and its inhibitors supports the optimization of existing drugs and the discovery of drugs for implication in B-cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Neelaveni Thangavel
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
- Correspondence: (N.T.); (S.M.)
| | | | - Marwa Qadri
- Department of Pharmacology, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
- Medical Research Center, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Safeena Eranhiyil Ashraf
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Safaa Fathy Saleh
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Maryam Nayeem
- Department of Pharmacology, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Syam Mohan
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt
- Substance Abuse and Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun 248007, India
- Correspondence: (N.T.); (S.M.)
| |
Collapse
|
22
|
ten Hacken E, Sewastianik T, Yin S, Hoffmann GB, Gruber M, Clement K, Penter L, Redd RA, Ruthen N, Hergalant S, Sholokhova A, Fell G, Parry EM, Broséus J, Guieze R, Lucas F, Hernández-Sánchez M, Baranowski K, Southard J, Joyal H, Billington L, Regis FFD, Witten E, Uduman M, Knisbacher BA, Li S, Lyu H, Vaisitti T, Deaglio S, Inghirami G, Feugier P, Stilgenbauer S, Tausch E, Davids MS, Getz G, Livak KJ, Bozic I, Neuberg DS, Carrasco RD, Wu CJ. In Vivo Modeling of CLL Transformation to Richter Syndrome Reveals Convergent Evolutionary Paths and Therapeutic Vulnerabilities. Blood Cancer Discov 2023; 4:150-169. [PMID: 36468984 PMCID: PMC9975769 DOI: 10.1158/2643-3230.bcd-22-0082] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/16/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Transformation to aggressive disease histologies generates formidable clinical challenges across cancers, but biological insights remain few. We modeled the genetic heterogeneity of chronic lymphocytic leukemia (CLL) through multiplexed in vivo CRISPR-Cas9 B-cell editing of recurrent CLL loss-of-function drivers in mice and recapitulated the process of transformation from indolent CLL into large cell lymphoma [i.e., Richter syndrome (RS)]. Evolutionary trajectories of 64 mice carrying diverse combinatorial gene assortments revealed coselection of mutations in Trp53, Mga, and Chd2 and the dual impact of clonal Mga/Chd2 mutations on E2F/MYC and interferon signaling dysregulation. Comparative human and murine RS analyses demonstrated tonic PI3K signaling as a key feature of transformed disease, with constitutive activation of the AKT and S6 kinases, downmodulation of the PTEN phosphatase, and convergent activation of MYC/PI3K transcriptional programs underlying enhanced sensitivity to MYC/mTOR/PI3K inhibition. This robust experimental system presents a unique framework to study lymphoid biology and therapy. SIGNIFICANCE Mouse models reflective of the genetic complexity and heterogeneity of human tumors remain few, including those able to recapitulate transformation to aggressive disease histologies. Herein, we model CLL transformation into RS through multiplexed in vivo gene editing, providing key insight into the pathophysiology and therapeutic vulnerabilities of transformed disease. This article is highlighted in the In This Issue feature, p. 101.
Collapse
Affiliation(s)
- Elisa ten Hacken
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Tomasz Sewastianik
- Harvard Medical School, Boston, Massachusetts
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Shanye Yin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Michaela Gruber
- CEMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kendell Clement
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Molecular Pathology Unit, Center for Cancer Research and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité – Universitätsmedizin Berlin (corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin), Berlin, Germany
| | - Robert A. Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Neil Ruthen
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sébastien Hergalant
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risques Environnementaux (N-GERE), Université de Lorraine, Nancy, France
| | - Alanna Sholokhova
- Department of Applied Mathematics, University of Washington, Seattle, Washington
| | - Geoffrey Fell
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Erin M. Parry
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Julien Broséus
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risques Environnementaux (N-GERE), Université de Lorraine, Nancy, France
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, Nancy, France
| | | | - Fabienne Lucas
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - María Hernández-Sánchez
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, Madrid, Spain
| | - Kaitlyn Baranowski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jackson Southard
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Heather Joyal
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Leah Billington
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Fara Faye D. Regis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elizabeth Witten
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mohamed Uduman
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Binyamin A. Knisbacher
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Shuqiang Li
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Haoxiang Lyu
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Pierre Feugier
- Inserm UMRS1256 Nutrition-Génétique et Exposition aux Risques Environnementaux (N-GERE), Université de Lorraine, Nancy, France
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, Nancy, France
| | - Stephan Stilgenbauer
- Department III of Internal Medicine III, Division of CLL, Ulm University, Ulm, Germany
| | - Eugen Tausch
- Department III of Internal Medicine III, Division of CLL, Ulm University, Ulm, Germany
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Kenneth J. Livak
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ivana Bozic
- Department of Applied Mathematics, University of Washington, Seattle, Washington
| | - Donna S. Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ruben D. Carrasco
- Harvard Medical School, Boston, Massachusetts
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
23
|
Larrayoz M, Garcia-Barchino MJ, Celay J, Etxebeste A, Jimenez M, Perez C, Ordoñez R, Cobaleda C, Botta C, Fresquet V, Roa S, Goicoechea I, Maia C, Lasaga M, Chesi M, Bergsagel PL, Larrayoz MJ, Calasanz MJ, Campos-Sanchez E, Martinez-Cano J, Panizo C, Rodriguez-Otero P, Vicent S, Roncador G, Gonzalez P, Takahashi S, Katz SG, Walensky LD, Ruppert SM, Lasater EA, Amann M, Lozano T, Llopiz D, Sarobe P, Lasarte JJ, Planell N, Gomez-Cabrero D, Kudryashova O, Kurilovich A, Revuelta MV, Cerchietti L, Agirre X, San Miguel J, Paiva B, Prosper F, Martinez-Climent JA. Preclinical models for prediction of immunotherapy outcomes and immune evasion mechanisms in genetically heterogeneous multiple myeloma. Nat Med 2023; 29:632-645. [PMID: 36928817 PMCID: PMC10033443 DOI: 10.1038/s41591-022-02178-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 12/09/2022] [Indexed: 03/17/2023]
Abstract
The historical lack of preclinical models reflecting the genetic heterogeneity of multiple myeloma (MM) hampers the advance of therapeutic discoveries. To circumvent this limitation, we screened mice engineered to carry eight MM lesions (NF-κB, KRAS, MYC, TP53, BCL2, cyclin D1, MMSET/NSD2 and c-MAF) combinatorially activated in B lymphocytes following T cell-driven immunization. Fifteen genetically diverse models developed bone marrow (BM) tumors fulfilling MM pathogenesis. Integrative analyses of ∼500 mice and ∼1,000 patients revealed a common MAPK-MYC genetic pathway that accelerated time to progression from precursor states across genetically heterogeneous MM. MYC-dependent time to progression conditioned immune evasion mechanisms that remodeled the BM microenvironment differently. Rapid MYC-driven progressors exhibited a high number of activated/exhausted CD8+ T cells with reduced immunosuppressive regulatory T (Treg) cells, while late MYC acquisition in slow progressors was associated with lower CD8+ T cell infiltration and more abundant Treg cells. Single-cell transcriptomics and functional assays defined a high ratio of CD8+ T cells versus Treg cells as a predictor of response to immune checkpoint blockade (ICB). In clinical series, high CD8+ T/Treg cell ratios underlie early progression in untreated smoldering MM, and correlated with early relapse in newly diagnosed patients with MM under Len/Dex therapy. In ICB-refractory MM models, increasing CD8+ T cell cytotoxicity or depleting Treg cells reversed immunotherapy resistance and yielded prolonged MM control. Our experimental models enable the correlation of MM genetic and immunological traits with preclinical therapy responses, which may inform the next-generation immunotherapy trials.
Collapse
Affiliation(s)
- Marta Larrayoz
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Maria J Garcia-Barchino
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Jon Celay
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Amaia Etxebeste
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Maddalen Jimenez
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Cristina Perez
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Raquel Ordoñez
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Cesar Cobaleda
- Immune System Development and Function Unit, Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas/Universidad Autonoma, Madrid, Spain
| | - Cirino Botta
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Vicente Fresquet
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Sergio Roa
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Ibai Goicoechea
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Catarina Maia
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Miren Lasaga
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Marta Chesi
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - P Leif Bergsagel
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Maria J Larrayoz
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Maria J Calasanz
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Elena Campos-Sanchez
- Immune System Development and Function Unit, Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas/Universidad Autonoma, Madrid, Spain
| | - Jorge Martinez-Cano
- Immune System Development and Function Unit, Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas/Universidad Autonoma, Madrid, Spain
| | - Carlos Panizo
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Silvestre Vicent
- Program in Solid Tumors, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBERONC, Pamplona, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Research Centre CNIO, Madrid, Spain
| | - Patricia Gonzalez
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Research Centre CNIO, Madrid, Spain
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Loren D Walensky
- Department of Pediatric Oncology and Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shannon M Ruppert
- Oncology Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Elisabeth A Lasater
- Department of Translational Oncology, Genentech, South San Francisco, CA, USA
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Schlieren, Switzerland
| | - Teresa Lozano
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Diana Llopiz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Pablo Sarobe
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Juan J Lasarte
- Program of Immunology and Immunotherapy, Center for Applied Medical Research CIMA, University of Navarra, IDISNA, CIBEREHD, Pamplona, Spain
| | - Nuria Planell
- Translational Bioinformatics Unit, Navarra-Biomed, Public University of Navarra, IDISNA, Pamplona, Spain
| | - David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarra-Biomed, Public University of Navarra, IDISNA, Pamplona, Spain
- Biological and Environmental Sciences & Engineering Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | | | | | - Maria V Revuelta
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Leandro Cerchietti
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Xabier Agirre
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
| | - Jesus San Miguel
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Bruno Paiva
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Felipe Prosper
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain
- Department of Hematology, Clinica Universidad de Navarra, CCUN, IDISNA, CIBERONC, Pamplona, Spain
| | - Jose A Martinez-Climent
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, Cancer Center University of Navarra (CCUN), Navarra Institute for Health Research (IDISNA), CIBERONC, Pamplona, Spain.
| |
Collapse
|
24
|
Chen Z, Han F, Du Y, Shi H, Zhou W. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:70. [PMID: 36797231 PMCID: PMC9935926 DOI: 10.1038/s41392-023-01332-8] [Citation(s) in RCA: 308] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/20/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Having a hypoxic microenvironment is a common and salient feature of most solid tumors. Hypoxia has a profound effect on the biological behavior and malignant phenotype of cancer cells, mediates the effects of cancer chemotherapy, radiotherapy, and immunotherapy through complex mechanisms, and is closely associated with poor prognosis in various cancer patients. Accumulating studies have demonstrated that through normalization of the tumor vasculature, nanoparticle carriers and biocarriers can effectively increase the oxygen concentration in the tumor microenvironment, improve drug delivery and the efficacy of radiotherapy. They also increase infiltration of innate and adaptive anti-tumor immune cells to enhance the efficacy of immunotherapy. Furthermore, drugs targeting key genes associated with hypoxia, including hypoxia tracers, hypoxia-activated prodrugs, and drugs targeting hypoxia-inducible factors and downstream targets, can be used for visualization and quantitative analysis of tumor hypoxia and antitumor activity. However, the relationship between hypoxia and cancer is an area of research that requires further exploration. Here, we investigated the potential factors in the development of hypoxia in cancer, changes in signaling pathways that occur in cancer cells to adapt to hypoxic environments, the mechanisms of hypoxia-induced cancer immune tolerance, chemotherapeutic tolerance, and enhanced radiation tolerance, as well as the insights and applications of hypoxia in cancer therapy.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Fangfang Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yan Du
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huaqing Shi
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China. .,Lanzhou University Sencond Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
25
|
Lacroix M, Beauchemin H, Khandanpour C, Möröy T. The RNA helicase DDX3 and its role in c-MYC driven germinal center-derived B-cell lymphoma. Front Oncol 2023; 13:1148936. [PMID: 37035206 PMCID: PMC10081492 DOI: 10.3389/fonc.2023.1148936] [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] [Received: 01/20/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
DDX3X is an RNA helicase with many functions in RNA metabolism such as mRNA translation, alternative pre-mRNA splicing and mRNA stability, but also plays a role as a regulator of transcription as well as in the Wnt/beta-catenin- and Nf-κB signaling pathways. The gene encoding DDX3X is located on the X-chromosome, but escapes X-inactivation. Hence females have two active copies and males only one. However, the Y chromosome contains the gene for the male DDX3 homologue, called DDX3Y, which has a very high sequence similarity and functional redundancy with DDX3X, but shows a more restricted protein expression pattern than DDX3X. High throughput sequencing of germinal center (GC)-derived B-cell malignancies such as Burkitt Lymphoma (BL) and Diffuse large B-cell lymphoma (DLBCL) samples showed a high frequency of loss-of-function (LOF) mutations in the DDX3X gene revealing several features that distinguish this gene from others. First, DDX3X mutations occur with high frequency particularly in those GC-derived B-cell lymphomas that also show translocations of the c-MYC proto-oncogene, which occurs in almost all BL and a subset of DLBCL. Second, DDX3X LOF mutations occur almost exclusively in males and is very rarely found in females. Third, mutations in the male homologue DDX3Y have never been found in any type of malignancy. Studies with human primary GC B cells from male donors showed that a loss of DDX3X function helps the initial process of B-cell lymphomagenesis by buffering the proteotoxic stress induced by c-MYC activation. However, full lymphomagenesis requires DDX3 activity since an upregulation of DDX3Y expression is invariably found in GC derived B-cell lymphoma with DDX3X LOF mutation. Other studies with male transgenic mice that lack Ddx3x, but constitutively express activated c-Myc transgenes in B cells and are therefore prone to develop B-cell malignancies, also showed upregulation of the DDX3Y protein expression during the process of lymphomagenesis. Since DDX3Y is not expressed in normal human cells, these data suggest that DDX3Y may represent a new cancer cell specific target to develop adjuvant therapies for male patients with BL and DLBCL and LOF mutations in the DDX3X gene.
Collapse
Affiliation(s)
- Marion Lacroix
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Hugues Beauchemin
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, Canada
| | - Cyrus Khandanpour
- Klinik für Hämatologie und Onkologie, University Hospital Schleswig Holstein, University Lübeck, Lübeck, Germany
- *Correspondence: Tarik Möröy, ; Cyrus Khandanpour,
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, IRCM, Montréal, QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Tarik Möröy, ; Cyrus Khandanpour,
| |
Collapse
|
26
|
Wen KW, Wang L, Menke JR, Damania B. Cancers associated with human gammaherpesviruses. FEBS J 2022; 289:7631-7669. [PMID: 34536980 PMCID: PMC9019786 DOI: 10.1111/febs.16206] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/10/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023]
Abstract
Epstein-Barr virus (EBV; human herpesvirus 4; HHV-4) and Kaposi sarcoma-associated herpesvirus (KSHV; human herpesvirus 8; HHV-8) are human gammaherpesviruses that have oncogenic properties. EBV is a lymphocryptovirus, whereas HHV-8/KSHV is a rhadinovirus. As lymphotropic viruses, EBV and KSHV are associated with several lymphoproliferative diseases or plasmacytic/plasmablastic neoplasms. Interestingly, these viruses can also infect epithelial cells causing carcinomas and, in the case of KSHV, endothelial cells, causing sarcoma. EBV is associated with Burkitt lymphoma, classic Hodgkin lymphoma, nasopharyngeal carcinoma, plasmablastic lymphoma, lymphomatoid granulomatosis, leiomyosarcoma, and subsets of diffuse large B-cell lymphoma, post-transplant lymphoproliferative disorder, and gastric carcinoma. KSHV is implicated in Kaposi sarcoma, primary effusion lymphoma, multicentric Castleman disease, and KSHV-positive diffuse large B-cell lymphoma. Pathogenesis by these two herpesviruses is intrinsically linked to viral proteins expressed during the lytic and latent lifecycles. This comprehensive review intends to provide an overview of the EBV and KSHV viral cycles, viral proteins that contribute to oncogenesis, and the current understanding of the pathogenesis and clinicopathology of their related neoplastic entities.
Collapse
Affiliation(s)
- Kwun Wah Wen
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158
| | - Linlin Wang
- Department of Laboratory Medicine, University of California, San Francisco, CA 94158
| | - Joshua R. Menke
- Department of Pathology, Stanford University, Palo Alto, CA 94304
| | - Blossom Damania
- Department of Microbiology & Immunology & Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
27
|
Advances in Understanding of Metabolism of B-Cell Lymphoma: Implications for Therapy. Cancers (Basel) 2022; 14:cancers14225552. [PMID: 36428647 PMCID: PMC9688663 DOI: 10.3390/cancers14225552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
There have been significant recent advances in the understanding of the role of metabolism in normal and malignant B-cell biology. Previous research has focused on the role of MYC and mammalian target of rapamycin (mTOR) and how these interact with B-cell receptor signaling and hypoxia to regulate glycolysis, glutaminolysis, oxidative phosphorylation (OXPHOS) and related metabolic pathways in germinal centers. Many of the commonest forms of lymphoma arise from germinal center B-cells, reflecting the physiological attenuation of normal DNA damage checkpoints to facilitate somatic hypermutation of the immunoglobulin genes. As a result, these lymphomas can inherit the metabolic state of their cell-of-origin. There is increasing interest in the potential of targeting metabolic pathways for anti-cancer therapy. Some metabolic inhibitors such as methotrexate have been used to treat lymphoma for decades, with several new agents being recently licensed such as inhibitors of phosphoinositide-3-kinase. Several other inhibitors are in development including those blocking mTOR, glutaminase, OXPHOS and monocarboxylate transporters. In addition, recent work has highlighted the importance of the interaction between diet and cancer, with particular focus on dietary modifications that restrict carbohydrates and specific amino acids. This article will review the current state of this field and discuss future developments.
Collapse
|
28
|
Chen G, Wu K, Li H, Xia D, He T. Role of hypoxia in the tumor microenvironment and targeted therapy. Front Oncol 2022; 12:961637. [PMID: 36212414 PMCID: PMC9545774 DOI: 10.3389/fonc.2022.961637] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
Tumor microenvironment (TME), which is characterized by hypoxia, widely exists in solid tumors. As a current research hotspot in the TME, hypoxia is expected to become a key element to break through the bottleneck of tumor treatment. More and more research results show that a variety of biological behaviors of tumor cells are affected by many factors in TME which are closely related to hypoxia. In order to inhibiting the immune response in TME, hypoxia plays an important role in tumor cell metabolism and anti-apoptosis. Therefore, exploring the molecular mechanism of hypoxia mediated malignant tumor behavior and therapeutic targets is expected to provide new ideas for anti-tumor therapy. In this review, we discussed the effects of hypoxia on tumor behavior and its interaction with TME from the perspectives of immune cells, cell metabolism, oxidative stress and hypoxia inducible factor (HIF), and listed the therapeutic targets or signal pathways found so far. Finally, we summarize the current therapies targeting hypoxia, such as glycolysis inhibitors, anti-angiogenesis drugs, HIF inhibitors, hypoxia-activated prodrugs, and hyperbaric medicine.
Collapse
Affiliation(s)
- Gaoqi Chen
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Kaiwen Wu
- Department of Gastroenterology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hao Li
- Deparment of Neurology, Affiliated Hospital of Jiangsu University, Jiang Su University, Zhenjiang, China
| | - Demeng Xia
- Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
- *Correspondence: Demeng Xia, ; Tianlin He,
| | - Tianlin He
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Demeng Xia, ; Tianlin He,
| |
Collapse
|
29
|
Affiliation(s)
- Mark Roschewski
- From the Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Louis M Staudt
- From the Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Wyndham H Wilson
- From the Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
30
|
Tamura Y, Yamane K, Kawano Y, Bullinger L, Wirtz T, Weber T, Sander S, Ohki S, Kitajima Y, Okada S, Rajewsky K, Yasuda T. Concomitant Cytotoxic Effector Differentiation of CD4+ and CD8+ T Cells in Response to EBV-Infected B Cells. Cancers (Basel) 2022; 14:cancers14174118. [PMID: 36077655 PMCID: PMC9454722 DOI: 10.3390/cancers14174118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The Epstein–Barr virus (EBV) is a γ-herpes virus that primarily infects human B cells, and more than 90% of adults have experienced infection. EBV+ B cells express several viral proteins, transmitting signals important for the transformation and tumorigenesis of the infected B cells. Immune surveillance by the host immune system is important to suppress such abnormal expansion of EBV-infected B cells. Here we found that both CD4+ T cells and CD8+ T cells show similar gene expression patterns relating to cytotoxicity towards EBV-infected B cells. EBV-specific cytotoxic CD4+ T cells markedly expressed T-bet, Granzyme B, and Perforin alongside killing activity, which could reflect mechanisms shared with cytotoxic CD8+ T cells. Our findings support the concept that, upon EBV and perhaps other viral infections, T cells of different subsets can be drawn into common pathways mediating immune surveillance through cytotoxicity. Abstract Most people infected by EBV acquire specific immunity, which then controls latent infection throughout their life. Immune surveillance of EBV-infected cells by cytotoxic CD4+ T cells has been recognized; however, the molecular mechanism of generating cytotoxic effector T cells of the CD4+ subset remains poorly understood. Here we compared phenotypic features and the transcriptome of EBV-specific effector-memory CD4+ T cells and CD8+ T cells in mice and found that both T cell types show cytotoxicity and, to our surprise, widely similar gene expression patterns relating to cytotoxicity. Similar to cytotoxic CD8+ T cells, EBV-specific cytotoxic CD4+ T cells from human peripheral blood expressed T-bet, Granzyme B, and Perforin and upregulated the degranulation marker, CD107a, immediately after restimulation. Furthermore, T-bet expression in cytotoxic CD4+ T cells was highly correlated with Granzyme B and Perforin expression at the protein level. Thus, differentiation of EBV-specific cytotoxic CD4+ T cells is possibly controlled by mechanisms shared by cytotoxic CD8+ T cells. T-bet-mediated transcriptional regulation may explain the similarity of cytotoxic effector differentiation between CD4+ T cells and CD8+ T cells, implicating that this differentiation pathway may be directed by environmental input rather than T cell subset.
Collapse
Affiliation(s)
- Yumi Tamura
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Keita Yamane
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yohei Kawano
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Chariteé-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Tristan Wirtz
- Immune Regulation and Cancer, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Timm Weber
- Immune Regulation and Cancer, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Sandrine Sander
- Immune Regulation and Cancer, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Shun Ohki
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yasuo Kitajima
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Klaus Rajewsky
- Immune Regulation and Cancer, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Tomoharu Yasuda
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
- Immune Regulation and Cancer, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Correspondence: ; Tel.: +81-82-257-5175
| |
Collapse
|
31
|
Sang W, Tu D, Zhang M, Qin Y, Yin W, Song X, Sun C, Yan D, Wang X, Zeng L, Li Z, Xu K, Xu L. l-Asparaginase synergizes with etoposide via the PI3K/Akt/mTOR pathway in Epstein-Barr virus-positive Burkitt lymphoma. J Biochem Mol Toxicol 2022; 36:e23117. [PMID: 35757978 DOI: 10.1002/jbt.23117] [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: 09/24/2021] [Revised: 04/24/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022]
Abstract
Burkitt lymphoma (BL) is an aggressive Epstein-Barr virus (EBV)-driven B-cell lymphoma characterized by the translocation and rearrangement of the c-Myc proto-oncogene. High-intensity multidrug chemotherapy regimens have a limited effect on the survival of refractory or relapsed BL patients, mainly owing to the high EBV load and drug resistance. l-asparaginase ( l-Asp) and etoposide (VP-16) play a beneficial role in EBV-related lymphoproliferative diseases; however, their roles and mechanisms in BL remain unclear. In this study, we found that VP-16 inhibited BL cell proliferation and arrested the cell cycle at the G2 /M phase. It also induced autophagy and activated the extrinsic and intrinsic apoptotic signaling pathways in BL cells. Mechanistically, VP-16 inhibited c-Myc expression and regulated the PI3K/Akt/mTOR signaling pathway. Notably, VP-16 also showed a specific synergistic effect with l-Asp to induce apoptosis in EBV-positive BL cells but not in EBV-negative BL cells. VP-16 combined with l-Asp further inhibited c-Myc expression and downregulated the PI3K/Akt/mTOR signaling pathway. Additionally, we found that VP-16 inhibited the expression of latent membrane protein 1 (LMP1), and in combination with l-Asp further decreased LMP1 expression in Raji cells. Our in vivo data also showed that the dual-drug combination significantly inhibited the growth of BL tumors and prolonged the survival of mice compared to VP-16 alone. In conclusion, this study provides new evidence that l-Asp may enhance the antitumor effect of VP-16 by inhibiting the PI3K/Akt/mTOR signaling pathway in EBV-positive BL cells.
Collapse
Affiliation(s)
- Wei Sang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Dongyun Tu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China.,Department of Cardiology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu, China
| | - Meng Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Yuanyuan Qin
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Wenjing Yin
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Xuguang Song
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cai Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongmei Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangmin Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Linyan Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| |
Collapse
|
32
|
Man J, Wang H, Qian X, Chen L, Ma Y, Qian M, Zhai X. TCF3 protein was highly expressed in pediatric Burkitt lymphoma and predicts poor prognosis: a single-center study. Leuk Lymphoma 2022; 63:2453-2460. [PMID: 35617527 DOI: 10.1080/10428194.2022.2076852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This retrospective single-center study was to evaluate the expression of TCF3 protein in pediatric Burkitt lymphomas (pBLs) and analyze its relations with clinical characteristics and prognosis. A total of 58 pBLs and 30 reactive hyperplastic lymphadenites (RH) were recruited. The high expression rate of TCF3 was 67.24% in pBLs, significantly higher than that in the RHs (36.67%, p = .01), which was consistent with the findings in biopsy specimens from mRNA and protein level, respectively. The expression of TCF3 was significantly associated with tumor localization and size. A total of 54 patients having received short-intensive chemotherapy had a median follow-up of 54.15 months (range: 1-111 months). Log-rank test of Kaplan-Meier survival curves indicated an inverse correlation of TCF3 expression with the overall survival (OS) and event-free survival (EFS). Univariate analysis showed that high TCF3 expression was significantly associated with poor EFS. The result of multivariate COX regression analysis indicated that the TCF3 expression was an independent prognostic factor for EFS.
Collapse
Affiliation(s)
- Jie Man
- Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, PR China
| | - Hongsheng Wang
- Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, PR China
| | - Xiaowen Qian
- Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, PR China
| | - Lian Chen
- Pathology, Children's Hospital of Fudan University, Shanghai, PR China
| | - Yangyang Ma
- Pathology, Children's Hospital of Fudan University, Shanghai, PR China
| | - Maoxiang Qian
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, PR China.,The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Xiaowen Zhai
- Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, PR China
| |
Collapse
|
33
|
Latent Membrane Proteins from EBV Differentially Target Cellular Pathways to Accelerate MYC-induced Lymphomagenesis. Blood Adv 2022; 6:4283-4296. [PMID: 35605249 PMCID: PMC9327557 DOI: 10.1182/bloodadvances.2022007695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/12/2022] [Indexed: 11/20/2022] Open
Abstract
EBV LMP1 enhances MYC-mediated degradation of the p27kip1 tumor suppressor and accelerates MYC-induced lymphomagenesis. EBV LMP1 and LMP2A differentially use G1-specific cell cycle and BCR-mediated signaling to accelerate MYC-induced lymphomagenesis.
MYC translocations in association with Epstein-Barr virus (EBV) infection are often observed in B-cell lymphomas. A subset of Burkitt lymphoma (BL) expresses EBV latent membrane proteins 1 and 2A (LMP1 and LMP2A) in addition to the typical restricted EBV latent gene expression. EBV-associated diffuse large B-cell lymphoma (DLBCL) typically exhibits latency type II or III and expresses LMP1. Here, we investigate the role of LMP1 in MYC-driven lymphomagenesis in our murine model. λ-MYC mice develop tumors having a “starry sky” appearance and have abnormal p53 expression that is also observed in human BL. LMP2A/λ-MYC double-transgenic mice develop tumors significantly faster than mice only expressing MYC. Similar to LMP2A/λ-MYC mice, LMP1/λ-MYC mice also have accelerated MYC-driven lymphomagenesis. As observed in LMP2A/λ-MYC mice, p27kip1 was degraded in LMP1/λ-MYC pretumor and tumor B cells. Coexpression of LMP1 and LMP2A resulted in the enhancement of B cell proliferation. In contrast to LMP2A, the inhibition of Syk or cyclin-dependant kinase (CDK)4/6 activity did not effectively inhibit LMP1-mediated MYC lymphomagenesis. Also, in contrast to LMP2A, LMP1 did not lessen abnormal p53 expression in λ-MYC tumors. To investigate the significance of LMP1 expression in human BL development, we reanalyzed RNA sequencing (RNA-Seq) data of primary human BL from previous studies. Interestingly, p53 mutations were less observed in LMP1-expressing BL, although they were not significantly changed by EBV infection, indicating LMP1 may lessen p53 mutations in human primary BL. This suggests that LMP1 effects in EBV-associated human BL vary from what we observe in our murine model. Finally, our studies suggest a novel pathogenic role of LMP1 in lymphomagenesis.
Collapse
|
34
|
Summerauer AM, Jäggi V, Ogwang R, Traxel S, Colombo L, Amundsen E, Eyer T, Subramanian B, Fehr J, Mantel P, Idro R, Bürgler S. Epstein-Barr virus and malaria upregulate AID and APOBEC3 enzymes, but only AID seems to play a major mutagenic role in Burkitt lymphoma. Eur J Immunol 2022; 52:1273-1284. [PMID: 35503749 PMCID: PMC7613445 DOI: 10.1002/eji.202249820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/01/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022]
Abstract
Endemic Burkitt lymphoma (eBL) is characterized by an oncogenic IGH/c‐MYC translocation and Epstein–Barr virus (EBV) positivity, and is epidemiologically linked to Plasmodium falciparum malaria. Both EBV and malaria are thought to contribute to eBL by inducing the expression of activation‐induced cytidine deaminase (AID), an enzyme involved in the IGH/c‐MYC translocation. AID/apolipoprotein B mRNA editing catalytic polypeptide‐like (AID/APOBEC) family enzymes have recently emerged as potent mutagenic sources in a variety of cancers, but apart from AID, their involvement in eBL and their regulation by EBV and P. falciparum is unknown. Here, we show that upon inoculation with EBV, human B cells strongly upregulate the expression of enzymatically active APOBEC3B and APOBEC3G. In addition, we found significantly increased levels of APOBEC3A in B cells of malaria patients, which correlated with parasite load. Interestingly, despite the fact that APOBEC3A, APOBEC3B, and APOBEC3G caused c‐MYC mutations when overexpressed in HEK293T cells, a mutational enrichment in eBL tumors was only detected in AID motifs. This suggests that even though the EBV‐ and P. falciparum‐directed immune response triggers the expression and activity of several AID/APOBEC members, only the upregulation of AID has oncogenic consequences, while the induction of the APOBEC3 subfamily may primarily have immunoprotective functions.
Collapse
Affiliation(s)
- Andrea M. Summerauer
- Experimental Infectious Diseases and Cancer Research, Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichZurichSwitzerland
| | - Vera Jäggi
- Experimental Infectious Diseases and Cancer Research, Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Rodney Ogwang
- College of Health SciencesMakerere UniversityKampalaUganda
- Centre of Tropical NeuroscienceKitgum SiteKampalaUganda
- KEMRI‐Wellcome Trust Research ProgrammeCentre for Geographic Medicine CoastKilifiKenya
| | - Sabrina Traxel
- Experimental Infectious Diseases and Cancer Research, Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Lorenzo Colombo
- Experimental Infectious Diseases and Cancer Research, Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Eivind Amundsen
- KG Jebsen Centre for B Cell Malignancies, Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Tatjana Eyer
- Experimental Infectious Diseases and Cancer Research, Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Bibin Subramanian
- Department of Oncology, Microbiology, and Immunology, Faculty of Science and MedicineUniversity of FribourgFribourgSwitzerland
| | - Jan Fehr
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichZurichSwitzerland
| | - Pierre‐Yves Mantel
- Department of Oncology, Microbiology, and Immunology, Faculty of Science and MedicineUniversity of FribourgFribourgSwitzerland
| | - Richard Idro
- College of Health SciencesMakerere UniversityKampalaUganda
- Centre of Tropical NeuroscienceKitgum SiteKampalaUganda
| | - Simone Bürgler
- Experimental Infectious Diseases and Cancer Research, Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| |
Collapse
|
35
|
Xu H, Zhao C, Wang Q, Chen Y, Zhang W, Zhuang Y, Chen R, Zhang H. Primary ovarian Burkitt lymphoma: report of a case and review of literature. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2022; 15:110-119. [PMID: 35414839 PMCID: PMC8986468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
We report a case of primary ovarian Burkitt lymphoma that occurred in a 25-year-old woman. The patient complained of a mass in the right ovary discerned by physical examination 2 months prior. Ultrasound examination indicated that the right ovary was enlarged and abundant blood flow signals were observed. Right salpingo-oophorectomy was subsequently performed. Histology was characterized by diffuse sheets of monotonous medium-sized lymphoid cells with plentiful mitotic figures and apoptosis. Numerous tingible-body macrophages were found in the ovarian tissue, presenting a starry sky pattern. The tumor cells expressed CD20, CD10, BCL6 and MYC in the absence of BCL2. Ki-67 proliferative index was very high with a proliferation rate of near 100%. MYC (8q24) rearrangement was detected by fluorescence in situ hybridization (FISH) with no BCL2 (18q21) and BCL6 (3q37) gene rearrangements. Cumulative evidence established primary ovarian Burkitt lymphoma as the final histopathologic diagnosis with clinical stage I (FIGO). The patient received HyperCVAD chemotherapy after surgery and remained complete response (CR) for 18 months. We aim to provide insight into the future treatment of this rare but lethal disease.
Collapse
Affiliation(s)
- Hongliang Xu
- Department of Pathology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| | - Caixia Zhao
- Department of Pathology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| | - Qing Wang
- Department of Pathology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| | - Yong Chen
- Department of Pathology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| | - Weiqin Zhang
- Department of Pathology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| | - Yali Zhuang
- Department of Gynecology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| | - Rongming Chen
- Department of Pathology, The People’s Hospital of Changfeng CountyHefei 230000, People’s Republic of China
| | - Heping Zhang
- Department of Pathology, Anhui Province Maternity and Child Health Hospital, Anhui Medical UniversityHefei 230000, People’s Republic of China
| |
Collapse
|
36
|
Metabolic path toward TCF3 inactivation in Burkitt lymphoma. Blood 2022; 139:475-476. [PMID: 35084477 DOI: 10.1182/blood.2021014133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/05/2021] [Indexed: 11/20/2022] Open
|
37
|
SHMT2 inhibition disrupts the TCF3 transcriptional survival program in Burkitt lymphoma. Blood 2022; 139:538-553. [PMID: 34624079 PMCID: PMC8938936 DOI: 10.1182/blood.2021012081] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/11/2021] [Indexed: 01/29/2023] Open
Abstract
Burkitt lymphoma (BL) is an aggressive lymphoma type that is currently treated by intensive chemoimmunotherapy. Despite the favorable clinical outcome for most patients with BL, chemotherapy-related toxicity and disease relapse remain major clinical challenges, emphasizing the need for innovative therapies. Using genome-scale CRISPR-Cas9 screens, we identified B-cell receptor (BCR) signaling, specific transcriptional regulators, and one-carbon metabolism as vulnerabilities in BL. We focused on serine hydroxymethyltransferase 2 (SHMT2), a key enzyme in one-carbon metabolism. Inhibition of SHMT2 by either knockdown or pharmacological compounds induced anti-BL effects in vitro and in vivo. Mechanistically, SHMT2 inhibition led to a significant reduction of intracellular glycine and formate levels, which inhibited the mTOR pathway and thereby triggered autophagic degradation of the oncogenic transcription factor TCF3. Consequently, this led to a collapse of tonic BCR signaling, which is controlled by TCF3 and is essential for BL cell survival. In terms of clinical translation, we also identified drugs such as methotrexate that synergized with SHMT inhibitors. Overall, our study has uncovered the dependency landscape in BL, identified and validated SHMT2 as a drug target, and revealed a mechanistic link between SHMT2 and the transcriptional master regulator TCF3, opening up new perspectives for innovative therapies.
Collapse
|
38
|
Witte HM, Künstner A, Hertel N, Bernd HW, Bernard V, Stölting S, Merz H, von Bubnoff N, Busch H, Feller AC, Gebauer N. Integrative genomic and transcriptomic analysis in plasmablastic lymphoma identifies disruption of key regulatory pathways. Blood Adv 2022; 6:637-651. [PMID: 34714908 PMCID: PMC8791589 DOI: 10.1182/bloodadvances.2021005486] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/02/2021] [Indexed: 11/20/2022] Open
Abstract
Plasmablastic lymphoma (PBL) represents a clinically heterogeneous subtype of aggressive B-cell non-Hodgkin lymphoma. Targeted-sequencing studies and a single-center whole-exome sequencing (WES) study in HIV-positive patients recently revealed several genes associated with PBL pathogenesis; however, the global mutational landscape and transcriptional profile of PBL remain elusive. To inform on disease-associated mutational drivers, mutational patterns, and perturbed pathways in HIV-positive and HIV-negative PBL, we performed WES and transcriptome sequencing (RNA-sequencing) of 33 PBL tumors. Integrative analysis of somatic mutations and gene expression profiles was performed to acquire insights into the divergent genotype-phenotype correlation in Epstein-Barr virus-positive (EBV+) and EBV- PBL. We describe a significant accumulation of mutations in the JAK signal transducer and transcription activator (OSMR, STAT3, PIM1, and SOCS1), as well as receptor tyrosine-kinase RAS (ERBB3, NRAS, PDGFRB, and NTRK) pathways. We provide further evidence of frequent perturbances of NF-κB signaling (NFKB2 and BTK). Induced pathways, identified by RNA-sequencing, closely resemble the mutational profile regarding alterations accentuated in interleukin-6/JAK/STAT signaling, NF-κB activity, and MYC signaling. Moreover, class I major histocompatibility complex-mediated antigen processing and cell cycle regulation were significantly affected by EBV status. An almost exclusive upregulation of phosphatidylinositol 3-kinase/AKT/mTOR signaling in EBV+ PBL and a significantly induced expression of NTRK3 in concert with recurrent oncogenic mutations in EBV- PBL hint at a specific therapeutically targetable mechanism in PBL subgroups. Our characterization of a mutational and transcriptomic landscape in PBL, distinct from that of diffuse large B-cell lymphoma and multiple myeloma, substantiates the pathobiological independence of PBL in the spectrum of B-cell malignancies and thereby refines the taxonomy for aggressive lymphomas.
Collapse
Affiliation(s)
- Hanno M. Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Ulm, Germany
| | - Axel Künstner
- Medical Systems Biology Group, and
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany; and
| | - Nadine Hertel
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Heinz-Wolfram Bernd
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Veronica Bernard
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Stephanie Stölting
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Hartmut Merz
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany; and
| | - Hauke Busch
- Medical Systems Biology Group, and
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany; and
| | - Alfred C. Feller
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany; and
| |
Collapse
|
39
|
Chakravorty S, Afzali B, Kazemian M. EBV-associated diseases: Current therapeutics and emerging technologies. Front Immunol 2022; 13:1059133. [PMID: 36389670 PMCID: PMC9647127 DOI: 10.3389/fimmu.2022.1059133] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/14/2022] [Indexed: 11/13/2022] Open
Abstract
EBV is a prevalent virus, infecting >90% of the world's population. This is an oncogenic virus that causes ~200,000 cancer-related deaths annually. It is, in addition, a significant contributor to the burden of autoimmune diseases. Thus, EBV represents a significant public health burden. Upon infection, EBV remains dormant in host cells for long periods of time. However, the presence or episodic reactivation of the virus increases the risk of transforming healthy cells to malignant cells that routinely escape host immune surveillance or of producing pathogenic autoantibodies. Cancers caused by EBV display distinct molecular behaviors compared to those of the same tissue type that are not caused by EBV, presenting opportunities for targeted treatments. Despite some encouraging results from exploration of vaccines, antiviral agents and immune- and cell-based treatments, the efficacy and safety of most therapeutics remain unclear. Here, we provide an up-to-date review focusing on underlying immune and environmental mechanisms, current therapeutics and vaccines, animal models and emerging technologies to study EBV-associated diseases that may help provide insights for the development of novel effective treatments.
Collapse
Affiliation(s)
- Srishti Chakravorty
- Department of Biochemistry, Purdue University, West Lafayette, IN, United States
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Majid Kazemian
- Department of Biochemistry, Purdue University, West Lafayette, IN, United States.,Department of Computer Science, Purdue University, West Lafayette IN, United States
| |
Collapse
|
40
|
Deenick EK, Bier J, Lau A. PI3K Isoforms in B Cells. Curr Top Microbiol Immunol 2022; 436:235-254. [PMID: 36243847 DOI: 10.1007/978-3-031-06566-8_10] [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: 06/16/2023]
Abstract
Phosphatidylinositol-3-kinases (PI3K) control many aspects of cellular activation and differentiation and play an important role in B cells biology. Three different classes of PI3K have been described, all of which are expressed in B cells. However, it is the class IA PI3Ks, and the p110δ catalytic subunit in particular, which seem to play the most critical role in B cells. Here we discuss the important role that class IA PI3K plays in B cell development, activation and differentiation, as well as examine what is known about the other classes of PI3Ks in B cells.
Collapse
Affiliation(s)
- Elissa K Deenick
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.
- Faculty of Medicine and Health, UNSW, Sydney, Australia.
| | - Julia Bier
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW, Sydney, Australia
| | - Anthony Lau
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW, Sydney, Australia
| |
Collapse
|
41
|
Tonc E, Takeuchi Y, Chou C, Xia Y, Holmgren M, Fujii C, Raju S, Chang GS, Iwamoto M, Egawa T. Unexpected suppression of tumorigenesis by c-MYC via TFAP4-dependent restriction of stemness in B lymphocytes. Blood 2021; 138:2526-2538. [PMID: 34283887 PMCID: PMC8678995 DOI: 10.1182/blood.2021011711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022] Open
Abstract
The proliferative burst of B lymphocytes is essential for antigen receptor repertoire diversification during the development and selective expansion of antigen-specific clones during immune responses. High proliferative activity inevitably promotes oncogenesis, the risk of which is further elevated in B lymphocytes by endogenous gene rearrangement and somatic mutations. However, B-cell-derived cancers are rare, perhaps owing to putative intrinsic tumor-suppressive mechanisms. We show that c-MYC facilitates B-cell proliferation as a protumorigenic driver and unexpectedly coengages counteracting tumor suppression through its downstream factor TFAP4. TFAP4 is mutated in human lymphoid malignancies, particularly in >10% of Burkitt lymphomas, and reduced TFAP4 expression was associated with poor survival of patients with MYC-high B-cell acute lymphoblastic leukemia. In mice, insufficient TFAP4 expression accelerated c-MYC-driven transformation of B cells. Mechanistically, c-MYC suppresses the stemness of developing B cells by inducing TFAP4 and restricting self-renewal of proliferating B cells. Thus, the pursuant transcription factor cascade functions as a tumor suppressor module that safeguards against the transformation of developing B cells.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- DNA-Binding Proteins/genetics
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Humans
- Leukemia, Lymphoid/genetics
- Leukemia, Lymphoid/pathology
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/pathology
- Mice, Inbred C57BL
- Mutation
- Proto-Oncogene Proteins c-myc/genetics
- Transcription Factors/genetics
- Tumor Cells, Cultured
- Mice
Collapse
Affiliation(s)
- Elena Tonc
- Department of Pathology and Immunology and
| | | | - Chun Chou
- Department of Pathology and Immunology and
| | - Yu Xia
- Department of Pathology and Immunology and
| | | | | | | | - Gue Su Chang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO; and
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland, Baltimore, MD
| | | |
Collapse
|
42
|
Stergiou IE, Chatzis L, Papanikolaou A, Giannouli S, Tzioufas AG, Voulgarelis M, Kapsogeorgou EK. Akt Signaling Pathway Is Activated in the Minor Salivary Glands of Patients with Primary Sjögren's Syndrome. Int J Mol Sci 2021; 22:ijms222413441. [PMID: 34948236 PMCID: PMC8709495 DOI: 10.3390/ijms222413441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune exocrinopathy of mainly the salivary and lacrimal glands associated with high prevalence of lymphoma. Akt is a phosphoinositide-dependent serine/threonine kinase, controlling numerous pathological processes, including oncogenesis and autoimmunity. Herein, we sought to examine its implication in pSS pathogenesis and related lymphomagenesis. The expression of the entire and activated forms of Akt (partially and fully activated: phosphorylated at threonine-308 (T308) and serine-473 (S473), respectively), and two of its substrates, the proline-rich Akt-substrate of 40 kDa (PRAS40) and FoxO1 transcription factor has been immunohistochemically examined in minor salivary glands (MSG) of pSS patients (n = 29; including 9 with pSS-associated lymphoma) and sicca-complaining controls (sicca-controls; n = 10). The entire and phosphorylated Akt, PRAS40, and FoxO1 molecules were strongly, uniformly expressed in the MSG epithelia and infiltrating mononuclear cells of pSS patients, but not sicca-controls. Morphometric analysis revealed that the staining intensity of the fully activated phospho-Akt-S473 in pSS patients (with or without lymphoma) was significantly higher than sicca-controls. Akt pathway activation was independent from the extent or proximity of infiltrates, as well as other disease features, including lymphoma. Our findings support that the Akt pathway is specifically activated in MSGs of pSS patients, revealing novel therapeutic targets.
Collapse
Affiliation(s)
- Ioanna E. Stergiou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (L.C.); (A.G.T.); (M.V.)
| | - Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (L.C.); (A.G.T.); (M.V.)
| | | | - Stavroula Giannouli
- Hematology Unit, Second Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (L.C.); (A.G.T.); (M.V.)
| | - Michael Voulgarelis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (L.C.); (A.G.T.); (M.V.)
| | - Efstathia K. Kapsogeorgou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (L.C.); (A.G.T.); (M.V.)
- Correspondence: ; Tel.: +30-210-746-2670
| |
Collapse
|
43
|
Harrington CT, Sotillo E, Dang CV, Thomas-Tikhonenko A. Tilting MYC toward cancer cell death. Trends Cancer 2021; 7:982-994. [PMID: 34481764 PMCID: PMC8541926 DOI: 10.1016/j.trecan.2021.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022]
Abstract
MYC oncoprotein promotes cell proliferation and serves as the key driver in many human cancers; therefore, considerable effort has been expended to develop reliable pharmacological methods to suppress its expression or function. Despite impressive progress, MYC-targeting drugs have not reached the clinic. Recent advances suggest that within a limited expression range unique to each tumor, MYC oncoprotein can have a paradoxical, proapoptotic function. Here we introduce a counterintuitive idea that modestly and transiently elevating MYC levels could aid chemotherapy-induced apoptosis and thus benefit the patients as much, if not more than MYC inhibition.
Collapse
Affiliation(s)
- Colleen T Harrington
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elena Sotillo
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chi V Dang
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA; Ludwig Institute for Cancer Research, New York, NY 10017, USA
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
44
|
Calvo-Vidal MN, Zamponi N, Krumsiek J, Stockslager MA, Revuelta MV, Phillip JM, Marullo R, Tikhonova E, Kotlov N, Patel J, Yang SN, Yang L, Taldone T, Thieblemont C, Leonard JP, Martin P, Inghirami G, Chiosis G, Manalis SR, Cerchietti L. Oncogenic HSP90 Facilitates Metabolic Alterations in Aggressive B-cell Lymphomas. Cancer Res 2021; 81:5202-5216. [PMID: 34479963 PMCID: PMC8530929 DOI: 10.1158/0008-5472.can-21-2734] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022]
Abstract
HSP90 is critical for maintenance of the cellular proteostasis. In cancer cells, HSP90 also becomes a nucleating site for the stabilization of multiprotein complexes including signaling pathways and transcription complexes. Here we described the role of this HSP90 form, referred to as oncogenic HSP90, in the regulation of cytosolic metabolic pathways in proliferating B-cell lymphoma cells. Oncogenic HSP90 assisted in the organization of metabolic enzymes into non-membrane-bound functional compartments. Under experimental conditions that conserved cellular proteostasis, oncogenic HSP90 coordinated and sustained multiple metabolic pathways required for energy production and maintenance of cellular biomass as well as for secretion of extracellular metabolites. Conversely, inhibition of oncogenic HSP90, in absence of apparent client protein degradation, decreased the efficiency of MYC-driven metabolic reprogramming. This study reveals that oncogenic HSP90 supports metabolism in B-cell lymphoma cells and patients with diffuse large B-cell lymphoma, providing a novel mechanism of activity for HSP90 inhibitors. SIGNIFICANCE: The oncogenic form of HSP90 organizes and maintains functional multienzymatic metabolic hubs in cancer cells, suggesting the potential of repurposing oncogenic HSP90 selective inhibitors to disrupt metabolism in lymphoma cells.
Collapse
Affiliation(s)
- M Nieves Calvo-Vidal
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Nahuel Zamponi
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York
| | - Max A Stockslager
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Maria V Revuelta
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Jude M Phillip
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Rossella Marullo
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | | | | | - Jayeshkumar Patel
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Shao Ning Yang
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Lucy Yang
- Koch Institute for Integrative Cancer Research and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Tony Taldone
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Institute, New York, New York
| | - Catherine Thieblemont
- APHP, Saint-Louis Hospital, Hemato-Oncology, Paris - Paris Diderot University, Paris, France.,EA3788, Paris Descartes University, Paris, France
| | - John P Leonard
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Peter Martin
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Giorgio Inghirami
- Deparment of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Gabriela Chiosis
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Institute, New York, New York
| | - Scott R Manalis
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Koch Institute for Integrative Cancer Research and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Leandro Cerchietti
- Hematology and Oncology Division, Department of Medicine, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
45
|
Gong C, Krupka JA, Gao J, Grigoropoulos NF, Giotopoulos G, Asby R, Screen M, Usheva Z, Cucco F, Barrans S, Painter D, Zaini NBM, Haupl B, Bornelöv S, Ruiz De Los Mozos I, Meng W, Zhou P, Blain AE, Forde S, Matthews J, Khim Tan MG, Burke GAA, Sze SK, Beer P, Burton C, Campbell P, Rand V, Turner SD, Ule J, Roman E, Tooze R, Oellerich T, Huntly BJ, Turner M, Du MQ, Samarajiwa SA, Hodson DJ. Sequential inverse dysregulation of the RNA helicases DDX3X and DDX3Y facilitates MYC-driven lymphomagenesis. Mol Cell 2021; 81:4059-4075.e11. [PMID: 34437837 DOI: 10.1016/j.molcel.2021.07.041] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/17/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022]
Abstract
DDX3X is a ubiquitously expressed RNA helicase involved in multiple stages of RNA biogenesis. DDX3X is frequently mutated in Burkitt lymphoma, but the functional basis for this is unknown. Here, we show that loss-of-function DDX3X mutations are also enriched in MYC-translocated diffuse large B cell lymphoma and reveal functional cooperation between mutant DDX3X and MYC. DDX3X promotes the translation of mRNA encoding components of the core translational machinery, thereby driving global protein synthesis. Loss-of-function DDX3X mutations moderate MYC-driven global protein synthesis, thereby buffering MYC-induced proteotoxic stress during early lymphomagenesis. Established lymphoma cells restore full protein synthetic capacity by aberrant expression of DDX3Y, a Y chromosome homolog, the expression of which is normally restricted to the testis. These findings show that DDX3X loss of function can buffer MYC-driven proteotoxic stress and highlight the capacity of male B cell lymphomas to then compensate for this loss by ectopic DDX3Y expression.
Collapse
Affiliation(s)
- Chun Gong
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
| | - Joanna A Krupka
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK; MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge CB2 0XZ, UK
| | - Jie Gao
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
| | | | - George Giotopoulos
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
| | - Ryan Asby
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
| | - Michael Screen
- Immunology Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Zelvera Usheva
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
| | - Francesco Cucco
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK
| | - Sharon Barrans
- Haematological Malignancy Diagnostic Service, St. James's Institute of Oncology, Leeds LS9 7TF, UK
| | - Daniel Painter
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York YO10 5DD, UK
| | | | - Björn Haupl
- Department of Medicine II, Hematology/Oncology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt, Germany
| | - Susanne Bornelöv
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Igor Ruiz De Los Mozos
- The Francis Crick Institute, London NW1 1AT, UK; Department for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Wei Meng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, Singapore
| | - Peixun Zhou
- National Horizons Centre, Teesside University, 38 John Dixon Lane, Darlington DL1 1HG, UK; School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BA, UK
| | - Alex E Blain
- National Horizons Centre, Teesside University, 38 John Dixon Lane, Darlington DL1 1HG, UK; Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK; School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BA, UK
| | - Sorcha Forde
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK
| | - Jamie Matthews
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK
| | - Michelle Guet Khim Tan
- Department of Clinical Translational Research, Singapore General Hospital, Outram Road, Singapore 169856, Singapore
| | - G A Amos Burke
- Department of Paediatric Oncology, Addenbrooke's Hospital, Cambridge, UK
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, Singapore
| | - Philip Beer
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Cathy Burton
- Haematological Malignancy Diagnostic Service, St. James's Institute of Oncology, Leeds LS9 7TF, UK
| | - Peter Campbell
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Vikki Rand
- National Horizons Centre, Teesside University, 38 John Dixon Lane, Darlington DL1 1HG, UK; School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BA, UK
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK; CEITEC, Masaryk University, Brno, Czech Republic
| | - Jernej Ule
- The Francis Crick Institute, London NW1 1AT, UK; Department for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Eve Roman
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York YO10 5DD, UK
| | - Reuben Tooze
- Haematological Malignancy Diagnostic Service, St. James's Institute of Oncology, Leeds LS9 7TF, UK; Section of Experimental Haematology, Leeds Institute of Molecular Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Thomas Oellerich
- Department of Medicine II, Hematology/Oncology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt, Germany
| | - Brian J Huntly
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
| | - Martin Turner
- Immunology Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Ming-Qing Du
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK
| | - Shamith A Samarajiwa
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge CB2 0XZ, UK
| | - Daniel J Hodson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK.
| |
Collapse
|
46
|
Grenov AC, Moss L, Edelheit S, Cordiner R, Schmiedel D, Biram A, Hanna JH, Jensen TH, Schwartz S, Shulman Z. The germinal center reaction depends on RNA methylation and divergent functions of specific methyl readers. J Exp Med 2021; 218:e20210360. [PMID: 34402854 PMCID: PMC8374864 DOI: 10.1084/jem.20210360] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/02/2021] [Accepted: 07/22/2021] [Indexed: 12/19/2022] Open
Abstract
Long-lasting immunity depends on the generation of protective antibodies through the germinal center (GC) reaction. N6-methyladenosine (m6A) modification of mRNAs by METTL3 activity modulates transcript lifetime primarily through the function of m6A readers; however, the physiological role of this molecular machinery in the GC remains unknown. Here, we show that m6A modifications by METTL3 are required for GC maintenance through the differential functions of m6A readers. Mettl3-deficient GC B cells exhibited reduced cell-cycle progression and decreased expression of proliferation- and oxidative phosphorylation-related genes. The m6A binder, IGF2BP3, was required for stabilization of Myc mRNA and expression of its target genes, whereas the m6A reader, YTHDF2, indirectly regulated the expression of the oxidative phosphorylation gene program. Our findings demonstrate how two independent gene networks that support critical GC functions are modulated by m6A through distinct mRNA binders.
Collapse
Affiliation(s)
- Amalie C. Grenov
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lihee Moss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sarit Edelheit
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ross Cordiner
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dominik Schmiedel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob H. Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Torben Heick Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Schraga Schwartz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
47
|
Human CD22-Transgenic, Primary Murine Lymphoma Challenges Immunotherapies in Organ-Specific Tumor Microenvironments. Int J Mol Sci 2021; 22:ijms221910433. [PMID: 34638774 PMCID: PMC8508822 DOI: 10.3390/ijms221910433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022] Open
Abstract
Targeted immunotherapies have greatly changed treatment of patients with B cell malignancies. To further enhance immunotherapies, research increasingly focuses on the tumor microenvironment (TME), which differs considerably by organ site. However, immunocompetent mouse models of disease to study immunotherapies targeting human molecules within organ-specific TME are surprisingly rare. We developed a myc-driven, primary murine lymphoma model expressing a human-mouse chimeric CD22 (h/mCD22). Stable engraftment of three distinct h/mCD22+ lymphoma was established after subcutaneous and systemic injection. However, only systemic lymphoma showed immune infiltration that reflected human disease. In this model, myeloid cells supported lymphoma growth and showed a phenotype of myeloid-derived suppressor cells. The human CD22-targeted immunotoxin Moxetumomab was highly active against h/mCD22+ lymphoma and similarly reduced infiltration of bone marrow and spleen of all three models up to 90-fold while efficacy against lymphoma in lymph nodes varied substantially, highlighting relevance of organ-specific TME. As in human aggressive lymphoma, anti-PD-L1 as monotherapy was not efficient. However, anti-PD-L1 enhanced efficacy of Moxetumomab suggesting potential for future clinical application. The novel model system of h/mCD22+ lymphoma provides a unique platform to test targeted immunotherapies and may be amenable for other human B cell targets such as CD19 and CD20.
Collapse
|
48
|
Dawes JC, Uren AG. Forward and Reverse Genetics of B Cell Malignancies: From Insertional Mutagenesis to CRISPR-Cas. Front Immunol 2021; 12:670280. [PMID: 34484175 PMCID: PMC8414522 DOI: 10.3389/fimmu.2021.670280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer genome sequencing has identified dozens of mutations with a putative role in lymphomagenesis and leukemogenesis. Validation of driver mutations responsible for B cell neoplasms is complicated by the volume of mutations worthy of investigation and by the complex ways that multiple mutations arising from different stages of B cell development can cooperate. Forward and reverse genetic strategies in mice can provide complementary validation of human driver genes and in some cases comparative genomics of these models with human tumors has directed the identification of new drivers in human malignancies. We review a collection of forward genetic screens performed using insertional mutagenesis, chemical mutagenesis and exome sequencing and discuss how the high coverage of subclonal mutations in insertional mutagenesis screens can identify cooperating mutations at rates not possible using human tumor genomes. We also compare a set of independently conducted screens from Pax5 mutant mice that converge upon a common set of mutations observed in human acute lymphoblastic leukemia (ALL). We also discuss reverse genetic models and screens that use CRISPR-Cas, ORFs and shRNAs to provide high throughput in vivo proof of oncogenic function, with an emphasis on models using adoptive transfer of ex vivo cultured cells. Finally, we summarize mouse models that offer temporal regulation of candidate genes in an in vivo setting to demonstrate the potential of their encoded proteins as therapeutic targets.
Collapse
Affiliation(s)
- Joanna C Dawes
- Medical Research Council, London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Anthony G Uren
- Medical Research Council, London Institute of Medical Sciences, London, United Kingdom.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
49
|
Meyer SN, Koul S, Pasqualucci L. Mouse Models of Germinal Center Derived B-Cell Lymphomas. Front Immunol 2021; 12:710711. [PMID: 34456919 PMCID: PMC8387591 DOI: 10.3389/fimmu.2021.710711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/28/2021] [Indexed: 12/19/2022] Open
Abstract
Over the last decades, the revolution in DNA sequencing has changed the way we understand the genetics and biology of B-cell lymphomas by uncovering a large number of recurrently mutated genes, whose aberrant function is likely to play an important role in the initiation and/or maintenance of these cancers. Dissecting how the involved genes contribute to the physiology and pathology of germinal center (GC) B cells -the origin of most B-cell lymphomas- will be key to advance our ability to diagnose and treat these patients. Genetically engineered mouse models (GEMM) that faithfully recapitulate lymphoma-associated genetic alterations offer a valuable platform to investigate the pathogenic roles of candidate oncogenes and tumor suppressors in vivo, and to pre-clinically develop new therapeutic principles in the context of an intact tumor immune microenvironment. In this review, we provide a summary of state-of-the art GEMMs obtained by accurately modelling the most common genetic alterations found in human GC B cell malignancies, with a focus on Burkitt lymphoma, follicular lymphoma, and diffuse large B-cell lymphoma, and we discuss how lessons learned from these models can help guide the design of novel therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Stefanie N. Meyer
- Institute for Cancer Genetics, Columbia University, New York, NY, United States
| | - Sanjay Koul
- Department of Biological Sciences & Geology, Queensborough Community College (City University of New York), Bayside, NY, United States
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, NY, United States
- Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
- The Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, United States
| |
Collapse
|
50
|
Mossadegh-Keller N, Brisou G, Beyou A, Nadel B, Roulland S. Human B Lymphomas Reveal Their Secrets Through Genetic Mouse Models. Front Immunol 2021; 12:683597. [PMID: 34335584 PMCID: PMC8323519 DOI: 10.3389/fimmu.2021.683597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/12/2021] [Indexed: 12/18/2022] Open
Abstract
Lymphomas are cancers deriving from lymphocytes, arising preferentially in secondary lymphoid organs, and represent the 6th cancer worldwide and the most frequent blood cancer. The majority of B cell Non-Hodgkin lymphomas (B-NHL) develop from germinal center (GC) experienced mature B cells. GCs are transient structures that form in lymphoid organs in response to antigen exposure of naive B cells, and where B cell receptor (BCR) affinity maturation occurs to promote B cell differentiation into memory B and plasma cells producing high-affinity antibodies. Genomic instability associated with the somatic hypermutation (SHM) and class-switch recombination (CSR) processes during GC transit enhance susceptibility to malignant transformation. Most B cell differentiation steps in the GC are at the origin of frequent B cell malignant entities, namely Follicular Lymphoma (FL) and GCB diffuse large B cell lymphomas (GCB-DLBCL). Over the past decade, large sequencing efforts have provided a great boost in the identification of candidate oncogenes and tumor suppressors involved in FL and DLBCL oncogenesis. Mouse models have been instrumental to accurately mimic in vivo lymphoma-specific mutations and interrogate their normal function in the GC context and their oncogenic function leading to lymphoma onset. The limited access of biopsies during the initiating steps of the disease, the cellular and (epi)genetic heterogeneity of individual tumors across and within patients linked to perturbed dynamics of GC ecosystems make the development of genetically engineered mouse models crucial to decipher lymphomagenesis and disease progression and eventually to test the effects of novel targeted therapies. In this review, we provide an overview of some of the important genetically engineered mouse models that have been developed to recapitulate lymphoma-associated (epi)genetic alterations of two frequent GC-derived lymphoma entities: FL and GCB-DLCBL and describe how those mouse models have improved our knowledge of the molecular processes supporting GC B cell transformation.
Collapse
Affiliation(s)
| | - Gabriel Brisou
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Alicia Beyou
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Bertrand Nadel
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|