1
|
George SL, Lynn C, Stankunaite R, Hughes D, Sauer CM, Chalker J, Waqar Ahmed S, Oostveen M, Proszek PZ, Yuan L, Shaikh R, Jamal S, Brew A, Tall J, Rogers T, Clifford SC, Vormoor J, Shipley JM, Tweddle DA, Jones C, Willis C, Burke GA, Vedi A, Howell L, Johnston R, Rees H, Adams M, Jesudason A, Ronghe M, Elliott M, Ross E, Makin G, Campbell-Hewson Q, Grundy RG, Turnbull J, Wilson S, Lee V, Gray JC, Stoneham S, Gatz SA, Marshall LV, Angelini P, Anderson J, Cresswell GD, Graham TA, Al-Lazikani B, Cortés-Ciriano I, Kearns P, Hutchinson JC, Hargrave D, Jacques TS, Hubank M, Sottoriva A, Chesler L. Stratified Medicine Pediatrics: Cell-Free DNA and Serial Tumor Sequencing Identifies Subtype-Specific Cancer Evolution and Epigenetic States. Cancer Discov 2025; 15:717-732. [PMID: 39693475 PMCID: PMC11962403 DOI: 10.1158/2159-8290.cd-24-0916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/09/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024]
Abstract
SIGNIFICANCE In tumors of childhood, we identify mutations in epigenetic genes as drivers of relapse, with matched cfDNA sequencing showing significant intratumor genetic heterogeneity and cell-state specific patterns of chromatin accessibility. This highlights the power of cfDNA analysis to identify both genetic and epigenetic drivers of aggressive disease in pediatric cancers.
Collapse
Affiliation(s)
- Sally L. George
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
- Children and Young People’s Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Claire Lynn
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Reda Stankunaite
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Debbie Hughes
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Carolin M. Sauer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Jane Chalker
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Saira Waqar Ahmed
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Minou Oostveen
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Paula Z. Proszek
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Lina Yuan
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Ridwan Shaikh
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Sabri Jamal
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Ama Brew
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Jennifer Tall
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
| | - Tony Rogers
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
| | - Steven C. Clifford
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Josef Vormoor
- Princess Máxima Center and University Medical Center, Utrecht, the Netherlands
| | - Janet M. Shipley
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Deborah A. Tweddle
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chris Jones
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Courtney Willis
- Royal Aberdeen Children’s Hospital, Aberdeen, United Kingdom
| | - G.A. Amos Burke
- Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aditi Vedi
- Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Lisa Howell
- Alder-Hey Children’s Hospital, Liverpool, United Kingdom
| | - Robert Johnston
- Royal Belfast Hospital for Sick Children, Belfast, United Kingdom
| | - Helen Rees
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Madeleine Adams
- Noah’s Ark Children’s Hospital for Wales, Cardiff, United Kingdom
| | | | - Milind Ronghe
- Royal Hospital for Children, Glasgow, United Kingdom
| | | | - Emma Ross
- Leicester Royal Infirmary, Leicester, United Kingdom
| | - Guy Makin
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | | | | | | | | | - Victoria Lee
- Sheffield Children’s Hospital, Sheffield, United Kingdom
| | - Juliet C. Gray
- Southampton General Hospital, Southampton, United Kingdom
| | | | - Susanne A. Gatz
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Children’s Hospital, Birmingham, United Kingdom
| | - Lynley V. Marshall
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
- Children and Young People’s Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Paola Angelini
- Children and Young People’s Unit, The Royal Marsden Hospital, London, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - John Anderson
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, London, United Kingdom
| | - George D. Cresswell
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Trevor A. Graham
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Bissan Al-Lazikani
- Department of Genomic Medicine and the Therapeutics Discovery Division, MD Anderson Cancer Center, Houston, Texas
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Pamela Kearns
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - J. Ciaran Hutchinson
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Darren Hargrave
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, London, United Kingdom
- Haematology and Oncology Department, Great Ormond Street Hospital, London, United Kingdom
| | - Thomas S. Jacques
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, London, United Kingdom
| | - Michael Hubank
- Clinical Genomics, Centre for Molecular Pathology, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Louis Chesler
- Paediatric Oncology Experimental Medicine Centre (POEM), The Institute of Cancer Research, London, United Kingdom
- Children and Young People’s Unit, The Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
2
|
Zhao Z, Wang D, Sheng X, Li S, Liu T, Chang M, Feng L, Zhang D, Ji C, Lu F, Ye J. Identification and validation of BATF as a prognostic biomarker and regulator of immune cell infiltration in acute myeloid leukemia. Front Immunol 2025; 15:1429855. [PMID: 39872530 PMCID: PMC11769954 DOI: 10.3389/fimmu.2024.1429855] [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: 05/08/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Background Basic leucine zipper ATF-like transcription factor (BATF) is a nuclear basic leucine zipper protein affiliated with the AP-1/ATF superfamily. Previous research has confirmed that BATF expression plays a significant role in the tumour microenvironment. However, the associations between BATF expression and prognoses in acute myeloid leukaemia (AML) patients and their immunological effects remain unclear. Methods Genomic and clinical AML data were got from the TCGA (TCGA-LAML) and GEO (GSE37642) databases for the subsequent analysis. The expression levels of BATF in AML patients were assessed using GEPIA, and the results were verified by qRT-PCR and Western blotting. In the meantime, the prognostic value of BATF was evaluated using univariate and multivariate analyses, receiver operating characteristic (ROC) curve (AUC) analysis, and Kaplan-Meier (KM) survival analysis. EdU, colony formation, and CCK-8 assays were employed to evaluate the proliferation of cells. Moreover, we detected the association of BATF expression with drug sensitivity through database analysis and in vivo experiments. To further investigate the mechanism of action of BATF in AML, RNA sequencing (RNA-seq) analysis was performed, followed by pathway enrichment analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Meanwhile, we detected the connection between BATF expression and the proportions of immune cells via flow cytometry in C1498 mouse model of AML. Finally, we investigated the association between BATF expression and cell-cell communication within the AML cell population using single-cell sequencing. Results In this study, we thoroughly investigated the role of BATF in AML. First, we observed a significant elevation in the expression of BATF in patients and cells in AML. Further analysis revealed an association between high BATF expression and poor prognosis in AML. Additionally, BATF expression was found to promote the proliferative capacity of AML cells. Moreover, the results showed that the expression level of BATF dramatically affected the effect of chemotherapy in AML patients. We also discovered that BATF expression could activate multiple immune-related pathways, altering the proportions of CD8+T cells and NK cells, suggesting that BATF may be a regulator of immune cell infiltration. Finally, there were differences in receptor ligand pairs between AML cells with high and low expression of BATF and immune cells. Conclusion Bioinformatics analysis and experimental verification revealed that BATF expression could alter the proportions of CD8+T cells and NK cells in AML and affect drug sensitivity, making it a potential treatment target for AML.
Collapse
MESH Headings
- Humans
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Animals
- Prognosis
- Mice
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Female
- Tumor Microenvironment/immunology
- Male
- Cell Line, Tumor
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Cell Proliferation
- Gene Expression Regulation, Leukemic
- Middle Aged
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Dongmei Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xue Sheng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Shuying Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Mengyuan Chang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Feng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Di Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fei Lu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Jingjing Ye
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
3
|
Yin K, Guo Y, Wang J, Guo S, Zhang C, Dai Y, Guo Y, Dai C. Identification of a novel immune checkpoint-related gene signature predicts prognosis and immunotherapy in breast cancer and experiment verification. Sci Rep 2024; 14:31065. [PMID: 39730892 DOI: 10.1038/s41598-024-82266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/04/2024] [Indexed: 12/29/2024] Open
Abstract
Breast cancer (BRCA) is one of the pivotal causes of female death worldwide. And the morbidity and mortality of breast cancer have increased rapidly. Immune checkpoints are important to maintain immune tolerance and are regarded as important therapeutic targets. However, research for BRCA were limited to single immune checkpoint-related gene (ICG) and few studies have systematically explored expression profile of Immune checkpoint-related genes or attempted to construct a prognostic gene risk model based on immune checkpoint-related genes. We identified immune checkpoint-related differentially expressed genes (DEGs) in BRCA and normal breast tissues from TCGA database. A 7-gene signature was created by utilizing the univariate Cox regression model with least absolute shrinkage and selection operator (LASSO) Cox regression method. In addition, we conducted a nomogram to predict the prognostic significance. This tool enables quantitative prediction of patient prognosis, serving as a valuable reference for clinical decision-making, thereby improving patient outcomes. Relationships between our risk model and clinical indicators, TME (Tumor Microenvironment), immune cell infiltration, immune response and drug susceptibility were investigated. A set of in vitro cell assays was conducted to decipher the relationship between MAP2K6 and proliferation, invasion, migration, colony formation and apoptosis rate of breast cancer cells. As a result, we established a prognostic model composed of seven ICGs in BRCA. Based on the median risk score, BRCA patients were equally assigned into two groups of high- and low-risk. High-risk BRCA patients have poorer OS (overall survival) than low-risk patients. In addition, there were remarkable differences between these two groups in clinicopathological features, TME, immune cell infiltration, immune response and drug susceptibility. The results of GO and KEGG analyses indicated that DEGs between the high- and low-risk groups were involved in immune-related biological processes and pathways. GSEA analysis also showed that a number of immune-related pathways were notably enriched in the low-risk group. Finally, results of cell-based assays indicated that MAP2K6 may play a pivotal role in the initiation and progression of breast cancer as a tumor suppressor gene. In conclusion, we created a novel ICG signature that has the potential to predict the survival and drug sensitivity of BRCA patients. Furthermore, this study indicated that MAP2K6 may serve as a novel target for BRCA therapy.
Collapse
Affiliation(s)
- Ke Yin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yangyang Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jinqiu Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shenchao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chunxu Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yongping Dai
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Yu Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Chen Dai
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
4
|
Iqbal J, Inghirami G, Chan WC. New insights into the biology of T-cell lymphomas. Blood 2024; 144:1873-1886. [PMID: 39213420 PMCID: PMC11551850 DOI: 10.1182/blood.2023021787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
ABSTRACT Peripheral T-cell lymphomas (PTCLs) encompass a heterogeneous group of postthymic T-cell lymphomas with >30 distinct subtypes associated with varied clinicopathological features. Unfortunately, the overall survival of the major PTCL subtypes is dismal and has not improved for decades; thus, there is an urgent unmet clinical need to improve diagnosis, therapies, and clinical outcomes. The diagnosis is often challenging, requiring a combinatorial evaluation of clinical, morphologic, and immunophenotypic features. PTCL pathobiology is difficult to investigate due to enormous intertumor and intratumor heterogeneity, limited tissue availability, and the paucity of authentic T-cell lymphoma cell lines or genetically faithful animal models. The application of transcriptomic profiling and genomic sequencing has markedly accelerated the discovery of new biomarkers, molecular signatures, and genetic lesions, and some of the discoveries have been included in the revised World Health Organization or International Consensus Classification. Genome-wide investigations have revealed the mutational landscape and transcriptomic profiles of PTCL entities, defined the cell of origin as a major determinant of T-cell lymphoma biology, and allowed for the refinement of biologically and clinically meaningful entities for precision therapy. In this review, we prioritize the discussion on common nodal PTCL subtypes together with 2 virus-associated T-cell and natural killer cell lymphomas. We succinctly review normal T-cell development, differentiation, and T-cell receptor signaling as they relate to PTCL pathogenesis and biology. This review will facilitate a better biological understanding of the different PTCL entities and their stratification for additional studies and target-directed clinical trials.
Collapse
Affiliation(s)
- Javeed Iqbal
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
5
|
Liu X, Zhou W, Zheng D, Yang X, Qing Y, Liao W, Zeng W. BATF-Activated AIM2 Mediates Immune Escape in Lung Adenocarcinoma by Regulating PD-L1. Int Arch Allergy Immunol 2024; 186:345-357. [PMID: 39471785 DOI: 10.1159/000540875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 07/26/2024] [Indexed: 11/01/2024] Open
Abstract
INTRODUCTION Immunotherapy has demonstrated encouraging outcomes in tackling lung adenocarcinoma (LUAD), but immune escape may bring negative impacts. Only a single study has demonstrated the function of AIM2 in LUAD and reported that NF-κB and STAT1 are the chief transcription factors, this study is designed to analyze the role of AIM2 and examine the transcription factor, BATF in LUAD immunotherapy. METHODS Bioinformatics methods to analyze the expression and binding sites of AIM2 and BATF in LUAD, as well as the correlation between AIM2 and PD-L1. Dual-luciferase and chromatin immunoprecipitation assays were used to verify the binding of AIM2 and BATF. qRT-PCR and Western blot assayed expression of AIM2, BATF, and PD-L1 in LUAD. MTT measured cell viability, flow cytometry detected cell apoptosis, cytotoxicity assays measured the toxicity of CD8+ T cells to cancer cells, and enzyme-linked immunosorbent assay measured the expression of related cytokines. Immunohistochemistry detected the protein expression levels of AIM2, BATF, PD-L1, and CD8 in tumor tissue. RESULTS AIM2 and BATF were both highly expressed in LUAD, and there was a targeted binding relationship. BATF promoted LUAD cell proliferation and inhibited apoptosis by affecting AIM2 expression. The downregulation of AIM2 and PD-L1 expression inhibited PD-L1 and activated CD8+ T cells. The rescue experiment manifested that increased BATF weakened repression of AIM2 silencing on LUAD tumor immune escape in vitro and in vivo. CONCLUSION BATF promoted AIM2 expression, upregulated PD-L1, inhibited CD8+ T cell activity, and ultimately led to immune escape in LUAD. Our research uncovered an innovative outlook on the intricate regulation of immune checkpoint molecules and proposed a new approach to target the BATF/AIM2 axis in tumor immunotherapy.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wangyan Zhou
- Department of Medical Record, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Dayang Zheng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xu Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yongcheng Qing
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Weijun Liao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wei Zeng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
6
|
Dai W, Yin S, Wang F, Kuang T, Xiao H, Kang W, Yun C, Wang F, Luo L, Ao S, Zhou J, Yang X, Fan C, Li W, He D, Jin H, Tang W, Liu L, Wang R, Liang H, Zhu J. Punicalagin as a novel selective aryl hydrocarbon receptor (AhR) modulator upregulates AhR expression through the PDK1/p90RSK/AP-1 pathway to promote the anti-inflammatory response and bactericidal activity of macrophages. Cell Commun Signal 2024; 22:473. [PMID: 39363344 PMCID: PMC11448010 DOI: 10.1186/s12964-024-01847-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024] Open
Abstract
Aryl hydrocarbon receptor (AhR) plays an important role in inflammation and immunity as a new therapeutic target for infectious disease and sepsis. Punicalagin (PUN) is a Chinese herbal monomer extract of pomegranate peel that has beneficial anti-inflammatory, antioxidant and anti-infective effects. However, whether PUN is a ligand of AhR, its effect on AhR expression, and its signaling pathway remain poorly understood. In this study, we found that PUN was a unique polyphenolic compound that upregulated AhR expression at the transcriptional level, and regulated the AhR nongenomic pathway. AhR expression in lipopolysaccharide-induced macrophages was upregulated by PUN in vitro and in vivo in a time- and dose-dependent manner. Using specific inhibitors and siRNA, induction of AhR by PUN depended on sequential phosphorylation of 90-kDa ribosomal S6 kinase (p90RSK), which was activated by the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and phosphoinositide-dependent protein kinase (PDK)1 pathways. PUN promoted p90RSK-mediated activator protein-1 (AP-1) activation. AhR knockout or inhibitors reversed suppression of interleukin (IL)-6 and IL-1β expression by PUN. PUN decreased Listeria load and increased macrophage survival via AhR upregulation. In conclusion, we identified PUN as a novel selective AhR modulator involved in AhR expression via the MEK/ERK and PDK1 pathways targeting p90RSK/AP-1 in inflammatory macrophages, which inhibited macrophage inflammation and promoted bactericidal activity.
Collapse
Affiliation(s)
- Weihong Dai
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Shuangqin Yin
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fangjie Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tianyin Kuang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hongyan Xiao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wenyuan Kang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education & Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Caihong Yun
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Fei Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Li Luo
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shengxiang Ao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jing Zhou
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xue Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Chao Fan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wei Li
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dongmei He
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - He Jin
- Department of Cardiothoracic Surgery, 926th Hospital of Joint Logistics Support Force of PLA, Kaiyuan, 661600, China
| | - Wanqi Tang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lizhu Liu
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Rixing Wang
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China.
| | - Huaping Liang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Junyu Zhu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
7
|
Jin X, Zhou K, Zhang R, Li J, Guo M, Qiao H, Wu M, Cao X, Dong G, Zhang S. Construction and validation of prognostic signature for transcription factors regulating T cell exhaustion in hepatocellular carcinoma. Medicine (Baltimore) 2024; 103:e38713. [PMID: 38968464 PMCID: PMC11224837 DOI: 10.1097/md.0000000000038713] [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: 12/17/2023] [Accepted: 06/06/2024] [Indexed: 07/07/2024] Open
Abstract
In the tumor microenvironment (TME), CD8+ T cells showed stage exhaustion due to the continuous stimulation of tumor antigens. To evaluate the status of CD8+ T cells and reverse the exhaustion is the key to evaluate the prognosis and therapeutic effect of tumor patients. The aim of this study was to establish a prognostic signature that could effectively predict prognosis and response to immunotherapy in patients with hepatocellular carcinoma (HCC). We used univariate Cox analysis to obtain transcription factors associated with CD8+ T cell exhaustion from The Cancer Genome Atlas dataset. Then, the prognostic signature for transcription factors basic leucine zipper ATF-like transcription factor, Eomesodermin, and T-box protein 21 regulating T cell exhaustion was constructed using LASSO Cox regression. The relative expression levels of the mRNA of the 3 transcription factors were detected by reverse transcription-quantitative polymerase chain reaction in 23 pairs of HCC and paracancer tissues, and verified internally in The Cancer Genome Atlas dataset and externally in the International Cancer Genome Consortium dataset. Cox regression analysis showed that risk score was an independent prognostic variable. The overall survival of the high-risk group was significantly lower than that of the low-risk group. The low-risk group had higher immune scores, matrix scores, and ESTIMATE scores, and significantly increased expression levels of most immune checkpoint genes in the low-risk group. Therefore, patients with lower risk scores benefit more from immunotherapy. The combination of the 3 transcription factors can evaluate the exhaustion state of CD8+ T cells in the TME, laying a foundation for evaluating the TME and immunotherapy efficacy in patients with HCC.
Collapse
Affiliation(s)
- Xi Jin
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kun Zhou
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Clinical Laboratory, Beidahuang Industry Group General Hospital, Harbin, China
| | - Rongzheng Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingbo Li
- Department of Anesthesiology Research Institute, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengrui Guo
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Han Qiao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Wu
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyang Cao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanglu Dong
- Department of Tumor Radiotherapy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuyun Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Lu X, Liu J, Feng L, Huang Y, Xu Y, Li C, Wang W, Kan Y, Yang J, Zhang M. BATF promotes tumor progression and association with FDG PET-derived parameters in colorectal cancer. J Transl Med 2024; 22:558. [PMID: 38862971 PMCID: PMC11165778 DOI: 10.1186/s12967-024-05367-5] [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: 01/15/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024] Open
Abstract
PURPOSE The purpose of the study was to evaluate the expression and function of basic leucine zipper ATF-like transcription factor (BATF) in colorectal cancer (CRC), and its correlation with 2-deoxy-2[18F]fluoro-D-glucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) parameters. METHODS The TIMER database, GEPIA database, TCGA, and GEO database were used to analyze the expression profile of BATF in human cancers. The reverse transcription‑quantitative PCR and western blot analyses were used to evaluate the mRNA level and protein expression in different CRC cell lines. The expression of BATF in SW620 and HCT116 cells was silenced and cell counting kit-8 assays and clonogenic assay were utilized to evaluate the role of BATF in CRC proliferation. The expression of tumor BATF and glucose transporter 1 (GLUT-1) were examined using immunohistochemical tools in 37 CRC patients undergoing preoperative 18F-FDG PET/CT imaging. The correlation between the PET/CT parameters and immunohistochemical result was evaluated. RESULTS In database, BATF was highly expressed in pan-cancer analyses, including CRC, and was associated with poor prognosis in CRC. In vitro, the results showed that knocking down of BATF expression could inhibit the proliferation of SW620 and HCT116 cells. In CRC patients, BATF expression was upregulated in tumor tissues compared with matched para-tumoral tissues, and was related with gender and Ki-67 levels. BATF expression was positively related to GLUT-1 expression and PET/CT parameters, including tumor size, maximum standard uptake value, metabolic tumor volume, and total lesion glycolysis. The multiple logistic analyses showed that SUVmax was an independent predictor of BATF expression. With 15.96 g/cm3 as the cutoff, sensitivity was 85.71%, specificity 82.61%, and area-under-the-curve 0.854. CONCLUSION BATF may be an oncogene associated with 18F-FDG PET/CT parameters in CRC. SUVmax may be an independent predictor of BATF expression.
Collapse
Affiliation(s)
- Xia Lu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jun Liu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yanfeng Xu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Cuicui Li
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yin Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Mingyu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
9
|
Zhang R, Miao J, Zhai M, Liu R, Li F, Xu X, Huang L, Wang T, Yang R, Yang R, Wang Y, He A, Wang J. BATF promotes extramedullary infiltration through TGF-β1/Smad/MMPs axis in acute myeloid leukemia. Mol Carcinog 2024; 63:1146-1159. [PMID: 38477642 DOI: 10.1002/mc.23715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/01/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
Acute myeloid leukemia (AML) is one of the most prevalent types of leukemia and is challenging to cure for most patients. Basic Leucine Zipper ATF-Like Transcription Factor (BATF) has been reported to participate in the development and progression of numerous tumors. However, its role in AML is largely unknown. In this study, the expression and prognostic value of BATF were examined in AML. Our results demonstrated that BATF expression was upregulated in AML patients, which was significantly correlated with poor clinical characteristics and survival. Afterward, functional experiments were performed after knocking down or overexpressing BATF by transfecting small interfering RNAs and overexpression plasmids into AML cells. Our findings revealed that BATF promoted the migratory and invasive abilities of AML cells in vitro and in vivo. Moreover, the target genes of BATF were searched from databases to explore the binding of BATF to the target gene using ChIP and luciferase assays. Notably, our observations validated that BATF is bound to the promoter region of TGF-β1, which could transcriptionally enhance the expression of TGF-β1 and activate the TGF-β1/Smad/MMPs signaling pathway. In summary, our study established the aberrantly high expression of BATF and its pro-migratory function via the TGF-β1-Smad2/3-MMP2/9 axis in AML, which provides novel insights into extramedullary infiltration of AML.
Collapse
Affiliation(s)
- Ru Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiyu Miao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Zhai
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fangmei Li
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuezhu Xu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lingjuan Huang
- Department of Geriatrics, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Ting Wang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Yang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruoyu Yang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yiwen Wang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
- Department of Tumor and Immunology in Precision Medical Institute, Xi'an Jiaotong University, Xi'an, China
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianli Wang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Key Laboratory of Hematological Diseases, Xi'an, China
| |
Collapse
|
10
|
Dabbaghipour R, Ahmadi E, Entezam M, Farzam OR, Sohrabi S, Jamali S, Sichani AS, Paydar H, Baradaran B. Concise review: The heterogenous roles of BATF3 in cancer oncogenesis and dendritic cells and T cells differentiation and function considering the importance of BATF3-dependent dendritic cells. Immunogenetics 2024; 76:75-91. [PMID: 38358555 DOI: 10.1007/s00251-024-01335-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2024]
Abstract
The transcription factor, known as basic leucine zipper ATF-like 3 (BATF3), is a crucial contributor to the development of conventional type 1 dendritic cells (cDC1), which is definitely required for priming CD8 + T cell-mediated immunity against intracellular pathogens and malignancies. In this respect, BATF3-dependent cDC1 can bring about immunological tolerance, an autoimmune response, graft immunity, and defense against infectious agents such as viruses, microbes, parasites, and fungi. Moreover, the important function of cDC1 in stimulating CD8 + T cells creates an excellent opportunity to develop a highly effective target for vaccination against intracellular pathogens and diseases. BATF3 has been clarified to control the development of CD8α+ and CD103+ DCs. The presence of BATF3-dependent cDC1 in the tumor microenvironment (TME) reinforces immunosurveillance and improves immunotherapy approaches, which can be beneficial for cancer immunotherapy. Additionally, BATF3 acts as a transcriptional inhibitor of Treg development by decreasing the expression of the transcription factor FOXP3. However, when overexpressed in CD8 + T cells, it can enhance their survival and facilitate their transition to a memory state. BATF3 induces Th9 cell differentiation by binding to the IL-9 promoter through a BATF3/IRF4 complex. One of the latest research findings is the oncogenic function of BATF3, which has been approved and illustrated in several biological processes of proliferation and invasion.
Collapse
Affiliation(s)
- Reza Dabbaghipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mona Entezam
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Jamali
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Saber Sichani
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Hadi Paydar
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Prutsch N, He S, Berezovskaya A, Durbin AD, Dharia NV, Maher KA, Matthews JD, Hare L, Turner SD, Stegmaier K, Kenner L, Merkel O, Look AT, Abraham BJ, Zimmerman MW. STAT3 couples activated tyrosine kinase signaling to the oncogenic core transcriptional regulatory circuitry of anaplastic large cell lymphoma. Cell Rep Med 2024; 5:101472. [PMID: 38508140 PMCID: PMC10983107 DOI: 10.1016/j.xcrm.2024.101472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/01/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Anaplastic large cell lymphoma (ALCL) is an aggressive, CD30+ T cell lymphoma of children and adults. ALK fusion transcripts or mutations in the JAK-STAT pathway are observed in most ALCL tumors, but the mechanisms underlying tumorigenesis are not fully understood. Here, we show that dysregulated STAT3 in ALCL cooccupies enhancers with master transcription factors BATF3, IRF4, and IKZF1 to form a core regulatory circuit that establishes and maintains the malignant cell state in ALCL. Critical downstream targets of this network in ALCL cells include the protooncogene MYC, which requires active STAT3 to facilitate high levels of MYC transcription. The core autoregulatory transcriptional circuitry activity is reinforced by MYC binding to the enhancer regions associated with STAT3 and each of the core regulatory transcription factors. Thus, activation of STAT3 provides the crucial link between aberrant tyrosine kinase signaling and the core transcriptional machinery that drives tumorigenesis and creates therapeutic vulnerabilities in ALCL.
Collapse
Affiliation(s)
- Nicole Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA
| | - Shuning He
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA
| | - Alla Berezovskaya
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA
| | - Adam D Durbin
- Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02141, USA
| | - Kelsey A Maher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jamie D Matthews
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Lucy Hare
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Department of Pediatric Oncology and Hematology, Addenbrooke's Hospital, Cambridge, UK
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02141, USA
| | - Lukas Kenner
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Olaf Merkel
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA.
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Mark W Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Wang J, Gao M, Cheng M, Luo J, Lu M, Xing X, Sun Y, Lu Y, Li X, Shi C, Wang J, Wang N, Yang W, Jiang Y, Huang H, Yang G, Zeng Y, Wang C, Cao X. Single-Cell Transcriptional Analysis of Lamina Propria Lymphocytes in the Jejunum Reveals Innate Lymphoid Cell-like Cells in Pigs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:130-142. [PMID: 37975680 DOI: 10.4049/jimmunol.2300463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Pigs are the most suitable model to study various therapeutic strategies and drugs for human beings, although knowledge about cell type-specific transcriptomes and heterogeneity is poorly available. Through single-cell RNA sequencing and flow cytometry analysis of the types in the jejunum of pigs, we found that innate lymphoid cells (ILCs) existed in the lamina propria lymphocytes (LPLs) of the jejunum. Then, through flow sorting of live/dead-lineage (Lin)-CD45+ cells and single-cell RNA sequencing, we found that ILCs in the porcine jejunum were mainly ILC3s, with a small number of NK cells, ILC1s, and ILC2s. ILCs coexpressed IL-7Rα, ID2, and other genes and differentially expressed RORC, GATA3, and other genes but did not express the CD3 gene. ILC3s can be divided into four subgroups, and genes such as CXCL8, CXCL2, IL-22, IL-17, and NCR2 are differentially expressed. To further detect and identify ILC3s, we verified the classification of ILCs in the porcine jejunum subgroup and the expression of related hallmark genes at the protein level by flow cytometry. For systematically characterizing ILCs in the porcine intestines, we combined our pig ILC dataset with publicly available human and mice ILC data and identified that the human and pig ILCs shared more common features than did those mouse ILCs in gene signatures and cell states. Our results showed in detail for the first time (to our knowledge) the gene expression of porcine jejunal ILCs, the subtype classification of ILCs, and the markers of various ILCs, which provide a basis for an in-depth exploration of porcine intestinal mucosal immunity.
Collapse
Affiliation(s)
- Junhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ming Gao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Mingyang Cheng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jiawei Luo
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Mei Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xinyuan Xing
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yiyuan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xiaoxu Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wentao Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yanlong Jiang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China; and Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
13
|
Wang Y, Li Y, Wang C, Lio CWJ, Ma Q, Liu B. CEMIG: prediction of the cis-regulatory motif using the de Bruijn graph from ATAC-seq. Brief Bioinform 2023; 25:bbad505. [PMID: 38189539 PMCID: PMC10772951 DOI: 10.1093/bib/bbad505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/21/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024] Open
Abstract
Sequence motif discovery algorithms enhance the identification of novel deoxyribonucleic acid sequences with pivotal biological significance, especially transcription factor (TF)-binding motifs. The advent of assay for transposase-accessible chromatin using sequencing (ATAC-seq) has broadened the toolkit for motif characterization. Nonetheless, prevailing computational approaches have focused on delineating TF-binding footprints, with motif discovery receiving less attention. Herein, we present Cis rEgulatory Motif Influence using de Bruijn Graph (CEMIG), an algorithm leveraging de Bruijn and Hamming distance graph paradigms to predict and map motif sites. Assessment on 129 ATAC-seq datasets from the Cistrome Data Browser demonstrates CEMIG's exceptional performance, surpassing three established methodologies on four evaluative metrics. CEMIG accurately identifies both cell-type-specific and common TF motifs within GM12878 and K562 cell lines, demonstrating its comparative genomic capabilities in the identification of evolutionary conservation and cell-type specificity. In-depth transcriptional and functional genomic studies have validated the functional relevance of CEMIG-identified motifs across various cell types. CEMIG is available at https://github.com/OSU-BMBL/CEMIG, developed in C++ to ensure cross-platform compatibility with Linux, macOS and Windows operating systems.
Collapse
Affiliation(s)
- Yizhong Wang
- School of Mathematics, Shandong University, Jinan, 250100, China
| | - Yang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Chan-Wang Jerry Lio
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan, 250100, China
| |
Collapse
|
14
|
Zhang Y, Cheng K, Choi J. TCR Pathway Mutations in Mature T Cell Lymphomas. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1450-1458. [PMID: 37931208 PMCID: PMC10715708 DOI: 10.4049/jimmunol.2200682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 06/06/2023] [Indexed: 11/08/2023]
Abstract
Mature T cell lymphomas are heterogeneous neoplasms that are aggressive and resistant to treatment. Many of these cancers retain immunological properties of their cell of origin. They express cytokines, cytotoxic enzymes, and cell surface ligands normally induced by TCR signaling in untransformed T cells. Until recently, their molecular mechanisms were unclear. Recently, high-dimensional studies have transformed our understanding of their cellular and genetic characteristics. Somatic mutations in the TCR signaling pathway drive lymphomagenesis by disrupting autoinhibitory domains, increasing affinity to ligands, and/or inducing TCR-independent signaling. Collectively, most of these mutations augment signaling pathways downstream of the TCR. Emerging data suggest that these mutations not only drive proliferation but also determine lymphoma immunophenotypes. For example, RHOA mutations are sufficient to induce disease-relevant CD4+ T follicular helper cell phenotypes. In this review, we describe how mutations in the TCR signaling pathway elucidate lymphoma pathophysiology but also provide insights into broader T cell biology.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathleen Cheng
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jaehyuk Choi
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
15
|
Song Z, Wu W, Wei W, Xiao W, Lei M, Cai KQ, Huang DW, Jeong S, Zhang JP, Wang H, Kadin ME, Waldmann TA, Staudt LM, Nakagawa M, Yang Y. Analysis and therapeutic targeting of the IL-1R pathway in anaplastic large cell lymphoma. Blood 2023; 142:1297-1311. [PMID: 37339580 PMCID: PMC10613726 DOI: 10.1182/blood.2022019166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/11/2023] [Accepted: 06/01/2023] [Indexed: 06/22/2023] Open
Abstract
Anaplastic large cell lymphoma (ALCL), a subgroup of mature T-cell neoplasms with an aggressive clinical course, is characterized by elevated expression of CD30 and anaplastic cytology. To achieve a comprehensive understanding of the molecular characteristics of ALCL pathology and to identify therapeutic vulnerabilities, we applied genome-wide CRISPR library screenings to both anaplastic lymphoma kinase positive (ALK+) and primary cutaneous (pC) ALK- ALCLs and identified an unexpected role of the interleukin-1R (IL-1R) inflammatory pathway in supporting the viability of pC ALK- ALCL. Importantly, this pathway is activated by IL-1α in an autocrine manner, which is essential for the induction and maintenance of protumorigenic inflammatory responses in pC-ALCL cell lines and primary cases. Hyperactivation of the IL-1R pathway is promoted by the A20 loss-of-function mutation in the pC-ALCL lines we analyze and is regulated by the nonproteolytic protein ubiquitination network. Furthermore, the IL-1R pathway promotes JAK-STAT3 signaling activation in ALCLs lacking STAT3 gain-of-function mutation or ALK translocation and enhances the sensitivity of JAK inhibitors in these tumors in vitro and in vivo. Finally, the JAK2/IRAK1 dual inhibitor, pacritinib, exhibited strong activities against pC ALK- ALCL, where the IL-1R pathway is hyperactivated in the cell line and xenograft mouse model. Thus, our studies revealed critical insights into the essential roles of the IL-1R pathway in pC-ALCL and provided opportunities for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Zhihui Song
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Wenjun Wu
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Wei Wei
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Wenming Xiao
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD
| | - Michelle Lei
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Kathy Q. Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Subin Jeong
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Jing-Ping Zhang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Hongbo Wang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Marshall E. Kadin
- Department of Pathology and Laboratory Medicine, Brown University Alpert School of Medicine, Providence, RI
| | - Thomas A. Waldmann
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Masao Nakagawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Yibin Yang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|
16
|
Wu R, Lim MS. Updates in pathobiological aspects of anaplastic large cell lymphoma. Front Oncol 2023; 13:1241532. [PMID: 37810974 PMCID: PMC10556522 DOI: 10.3389/fonc.2023.1241532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Anaplastic large cell lymphomas (ALCL) encompass several distinct subtypes of mature T-cell neoplasms that are unified by the expression of CD30 and anaplastic cytomorphology. Identification of the cytogenetic abnormality t(2;5)(p23;q35) led to the subclassification of ALCLs into ALK+ ALCL and ALK- ALCL. According to the most recent World Health Organization (WHO) Classification of Haematolymphoid Tumours as well as the International Consensus Classification (ICC) of Mature Lymphoid Neoplasms, ALCLs encompass ALK+ ALCL, ALK- ALCL, and breast implant-associated ALCL (BI-ALCL). Approximately 80% of systemic ALCLs harbor rearrangement of ALK, with NPM1 being the most common partner gene, although many other fusion partner genes have been identified to date. ALK- ALCLs represent a heterogeneous group of lymphomas with distinct clinical, immunophenotypic, and genetic features. A subset harbor recurrent rearrangement of genes, including TYK2, DUSP22, and TP63, with a proportion for which genetic aberrations have yet to be characterized. Although primary cutaneous ALCL (pc-ALCL) is currently classified as a subtype of primary cutaneous T-cell lymphoma, due to the large anaplastic and pleomorphic morphology together with CD30 expression in the malignant cells, this review also discusses the pathobiological features of this disease entity. Genomic and proteomic studies have contributed significant knowledge elucidating novel signaling pathways that are implicated in ALCL pathogenesis and represent candidate targets of therapeutic interventions. This review aims to offer perspectives on recent insights regarding the pathobiological and genetic features of ALCL.
Collapse
Affiliation(s)
| | - Megan S. Lim
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
17
|
Lai P, Liu F, Liu X, Sun J, Wang Y. Differential molecular programs of cutaneous anaplastic large cell lymphoma and CD30-positive transformed mycosis fungoides. Front Immunol 2023; 14:1270365. [PMID: 37790936 PMCID: PMC10544577 DOI: 10.3389/fimmu.2023.1270365] [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: 07/31/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023] Open
Abstract
Background Discriminating between cutaneous anaplastic large cell lymphoma (cALCL) and CD30-positive transformed mycosis fungoides (CD30+ TMF) is challenging, particularly when they arise in the context of pre-existing mycosis fungoides. The development of molecular diagnostic tools was hampered by the rarity of both diseases and the limited understanding of their pathogenesis. Methods In this study, we established a cohort comprising 25 cALCL cases and 25 CD30+ TMF cases, with transcriptomic data obtained from 31 samples. We compared the clinicopathological information and investigated the gene expression profiling between these two entities. Furthermore, we developed an immunohistochemistry (IHC) algorithm to differentiate these two entities clinically. Results Our investigation revealed distinct clinicopathological features and unique gene expression programs associated with cALCL and CD30+ TMF. cALCL and CD30+ TMF displayed marked differences in gene expression patterns. Notably, CD30+ TMF demonstrated enrichment of T cell receptor signaling pathways and an exhausted T cell phenotype, accompanied by infiltration of B cells, dendritic cells, and neurons. In contrast, cALCL cells expressed high levels of HLA class II genes, polarized towards a Th17 phenotype, and exhibited neutrophil infiltration. An IHC algorithm with BATF3 and TCF7 staining emerged as potential diagnostic markers for identifying these two entities. Conclusions Our findings provide valuable insights into the differential molecular signatures associated with cALCL and CD30+ TMF, which contribute to their distinct clinicopathological behaviors. An appropriate IHC algorithm could be used as a potential diagnostic tool.
Collapse
Affiliation(s)
- Pan Lai
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Fengjie Liu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangjun Liu
- Department of Dermatology, Shandong University Qilu Hospital, Jinan, China
| | - Jingru Sun
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Yang Wang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| |
Collapse
|
18
|
Xiang C, Wu W, Fan M, Wang Z, Feng X, Liu C, Liu J, Liu G, Xia L, Si H, Gu Y, Liu N, Luo D, Wang Y, Ma D, Hu S, Liu H. Phosphorylated STAT3 as a potential diagnostic and predictive biomarker in ALK - ALCL vs. CD30 high PTCL, NOS. Front Immunol 2023; 14:1132834. [PMID: 37388733 PMCID: PMC10303105 DOI: 10.3389/fimmu.2023.1132834] [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: 12/28/2022] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
Aims The differential diagnosis between ALK-negative anaplastic large cell lymphoma (ALK- ALCL) and peripheral T-cell lymphoma, not otherwise specified (PTCL, NOS) with high expression of CD30 (CD30high) are essential. However, no reliable biomarker is available in daily practice except CD30. STAT3 is characteristically activated in ALCL. We aimed to investigate whether the status of STAT3 phosphorylation could help the differential diagnosis. Methods The status of phosphorylation of STAT3 was examined using two antibodies against pSTAT3-Y705 and pSTAT3-S727 by immunohistochemistry in ALK+ ALCL (n=33), ALK- ALCL (n=22) and PTCL, NOS (n=34). Ten PTCL, NOS with diffuse CD30 expression were defined as CD30high PTCL, NOS. Flowcytometric analysis were performed to evaluate the expression of pSTAT3-Y705/S727 in PTCL, NOS (n=3). Results The median H-scores of pSTAT3-Y705 and S727 were 280 and 260 in ALK+ ALCL, 250 and 240 in ALK- ALCL, and 45 and 75 in CD30high subgroup, respectively. Using H score of 145 as the cutoff value, pSTAT3-S727 alone distinguished between ALK- ALCL and CD30high PTCL, NOS with a sensitivity of 100% and specificity of 83%. Additionally, pSTAT3-S727, but not pSTAT3-Y705, was also expressed by background tumor-infiltrating lymphocytes (S727TILs) in PTCL, NOS. PTCL, NOS patients with high S727TILs H score had a favorable prognosis than those with no TILs (3-year OS rate: 43% vs. 0, p=0.013) or low S727TILs (3-year OS rate: 43% vs. 0, p=0.099). Flowcytometric analysis revealed that of the three patients investigated, two had enhanced pSTAT-S727 signals in neoplastic cell populations, and all three patients were negative for pSTAT3-Y705 expression in both tumor cells and background lymphocytes. Conclusions pSTAT3-Y705/S727 can be used to help distinguish ALK- ALCL from CD30high PTCL, NOS and pSTAT3-S727 expression by TILs predicts the prognosis of a subset of PTCL, NOS.
Collapse
Affiliation(s)
- Chenxi Xiang
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Wanna Wu
- Department of Pathology, The First Affiliated Hospital and School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Meiting Fan
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Zhen Wang
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center and National Clinical Research Center For Cancer and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cuiling Liu
- Department of Pathology, School of Basic Medical Sciences and Third Hospital, Pekin University Health Science Center, Beijing, China
| | - Jia Liu
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guangzhen Liu
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lei Xia
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Haipeng Si
- Department of Pathology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Gu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Nian Liu
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Dan Luo
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yubo Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Dongshen Ma
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Shimin Hu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hui Liu
- Department of Pathology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Ribeiro ML, Sánchez Vinces S, Mondragon L, Roué G. Epigenetic targets in B- and T-cell lymphomas: latest developments. Ther Adv Hematol 2023; 14:20406207231173485. [PMID: 37273421 PMCID: PMC10236259 DOI: 10.1177/20406207231173485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
Non-Hodgkin's lymphomas (NHLs) comprise a diverse group of diseases, either of mature B-cell or of T-cell derivation, characterized by heterogeneous molecular features and clinical manifestations. While most of the patients are responsive to standard chemotherapy, immunotherapy, radiation and/or stem cell transplantation, relapsed and/or refractory cases still have a dismal outcome. Deep sequencing analysis have pointed out that epigenetic dysregulations, including mutations in epigenetic enzymes, such as chromatin modifiers and DNA methyltransferases (DNMTs), are prevalent in both B- cell and T-cell lymphomas. Accordingly, over the past decade, a large number of epigenetic-modifying agents have been developed and introduced into the clinical management of these entities, and a few specific inhibitors have already been approved for clinical use. Here we summarize the main epigenetic alterations described in B- and T-NHL, that further supported the clinical development of a selected set of epidrugs in determined diseases, including inhibitors of DNMTs, histone deacetylases (HDACs), and extra-terminal domain proteins (bromodomain and extra-terminal motif; BETs). Finally, we highlight the most promising future directions of research in this area, explaining how bioinformatics approaches can help to identify new epigenetic targets in B- and T-cell lymphoid neoplasms.
Collapse
Affiliation(s)
- Marcelo Lima Ribeiro
- Lymphoma Translational Group, Josep Carreras
Leukaemia Research Institute, Badalona, Spain
- Laboratory of Immunopharmacology and Molecular
Biology, Sao Francisco University Medical School, Braganca Paulista,
Brazil
| | - Salvador Sánchez Vinces
- Laboratory of Immunopharmacology and Molecular
Biology, Sao Francisco University Medical School, Braganca Paulista,
Brazil
| | - Laura Mondragon
- T Cell Lymphoma Group, Josep Carreras Leukaemia
Research Institute, IJC. Ctra de Can Ruti, Camí de les Escoles s/n, 08916
Badalona, Barcelona, Spain
| | - Gael Roué
- Lymphoma Translational Group, Josep Carreras
Leukaemia Research Institute, IJC. Ctra de Can Ruti, Camí de les Escoles
s/n, 08916 Badalona, Barcelona, Spain
| |
Collapse
|
20
|
Turi M, Anilkumar Sithara A, Hofmanová L, Žihala D, Radhakrishnan D, Vdovin A, Knápková S, Ševčíková T, Chyra Z, Jelínek T, Šimíček M, Gullà A, Anderson KC, Hájek R, Hrdinka M. Transcriptome Analysis of Diffuse Large B-Cell Lymphoma Cells Inducibly Expressing MyD88 L265P Mutation Identifies Upregulated CD44, LGALS3, NFKBIZ, and BATF as Downstream Targets of Oncogenic NF-κB Signaling. Int J Mol Sci 2023; 24:ijms24065623. [PMID: 36982699 PMCID: PMC10057398 DOI: 10.3390/ijms24065623] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
During innate immune responses, myeloid differentiation primary response 88 (MyD88) functions as a critical signaling adaptor protein integrating stimuli from toll-like receptors (TLR) and the interleukin-1 receptor (IL-1R) family and translates them into specific cellular outcomes. In B cells, somatic mutations in MyD88 trigger oncogenic NF-κB signaling independent of receptor stimulation, which leads to the development of B-cell malignancies. However, the exact molecular mechanisms and downstream signaling targets remain unresolved. We established an inducible system to introduce MyD88 to lymphoma cell lines and performed transcriptomic analysis (RNA-seq) to identify genes differentially expressed by MyD88 bearing the L265P oncogenic mutation. We show that MyD88L265P activates NF-κB signaling and upregulates genes that might contribute to lymphomagenesis, including CD44, LGALS3 (coding Galectin-3), NFKBIZ (coding IkBƺ), and BATF. Moreover, we demonstrate that CD44 can serve as a marker of the activated B-cell (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) and that CD44 expression is correlated with overall survival in DLBCL patients. Our results shed new light on the downstream outcomes of MyD88L265P oncogenic signaling that might be involved in cellular transformation and provide novel therapeutical targets.
Collapse
Affiliation(s)
- Marcello Turi
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Anjana Anilkumar Sithara
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Lucie Hofmanová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - David Žihala
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Dhwani Radhakrishnan
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Alexander Vdovin
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Sofija Knápková
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Tereza Ševčíková
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Zuzana Chyra
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Michal Šimíček
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Annamaria Gullà
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Kenneth Carl Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Matouš Hrdinka
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
- Correspondence:
| |
Collapse
|
21
|
Gao Y, Hu S, Li R, Jin S, Liu F, Liu X, Li Y, Yan Y, Liu W, Gong J, Yang S, Tu P, Shen L, Bai F, Wang Y. Hyperprogression of cutaneous T cell lymphoma after anti-PD-1 treatment. JCI Insight 2023; 8:164793. [PMID: 36649072 PMCID: PMC9977500 DOI: 10.1172/jci.insight.164793] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUNDImmune checkpoint blockade is an emerging treatment for T cell non-Hodgkin's lymphoma (T-NHL), but some patients with T-NHL have experienced hyperprogression with undetermined mechanisms upon anti-PD-1 therapy.METHODSSingle-cell RNA-Seq, whole-genome sequencing, whole-exome sequencing, and functional assays were performed on primary malignant T cells from a patient with advanced cutaneous T cell lymphoma who experienced hyperprogression upon anti-PD-1 treatment.RESULTSThe patient was enrolled in a clinical trial of anti-PD-1 therapy and experienced disease hyperprogression. Single-cell RNA-Seq revealed that PD-1 blockade elicited a remarkable activation and proliferation of the CD4+ malignant T cells, which showed functional PD-1 expression and an exhausted status. Further analyses identified somatic amplification of PRKCQ in the malignant T cells. PRKCQ encodes PKCθ; PKCθ is a key player in the T cell activation/NF-κB pathway. PRKCQ amplification led to high expressions of PKCθ and p-PKCθ (T538) on the malignant T cells, resulting in an oncogenic activation of the T cell receptor (TCR) signaling pathway. PD-1 blockade in this patient released this signaling, derepressed the proliferation of malignant T cells, and resulted in disease hyperprogression.CONCLUSIONOur study provides real-world clinical evidence that PD-1 acts as a tumor suppressor for malignant T cells with oncogenic TCR activation.TRIAL REGISTRATIONClinicalTrials.gov (NCT03809767).FUNDINGThe National Natural Science Foundation of China (81922058), the National Science Fund for Distinguished Young Scholars (T2125002), the National Science and Technology Major Project (2019YFC1315702), the National Youth Top-Notch Talent Support Program (283812), and the Peking University Clinical Medicine plus X Youth Project (PKU2019LCXQ012) supported this work.
Collapse
Affiliation(s)
- Yumei Gao
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Simeng Hu
- Biomedical Pioneering Innovation Center (BIOPIC), and School of Life Sciences, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, China
| | - Ruoyan Li
- Biomedical Pioneering Innovation Center (BIOPIC), and School of Life Sciences, Peking University, Beijing, China.,Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Shanzhao Jin
- Biomedical Pioneering Innovation Center (BIOPIC), and School of Life Sciences, Peking University, Beijing, China.,BioMap Beijing Intelligence Technology Limited, Block C Information Center Haidian District, Beijing, China
| | - Fengjie Liu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Xiangjun Liu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Yingyi Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Yicen Yan
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Weiping Liu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, and
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Shuxia Yang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Ping Tu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), and School of Life Sciences, Peking University, Beijing, China.,Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China.,Center for Translational Cancer Research, Peking University First Hospital, Beijing, China
| | - Yang Wang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China.,National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| |
Collapse
|
22
|
Analysis and therapeutic targeting of the EP300 and CREBBP acetyltransferases in anaplastic large cell lymphoma and Hodgkin lymphoma. Leukemia 2023; 37:396-407. [PMID: 36456744 PMCID: PMC9949602 DOI: 10.1038/s41375-022-01774-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022]
Abstract
Anaplastic large cell lymphoma (ALCL) and classical Hodgkin lymphoma (HL) share a similar cytological and high surface expression of CD30, and novel therapeutic strategies are needed. The EP300 and CREBBP acetyltransferases play essential roles in the pathogenesis of non-Hodgkin B cell lymphoma, but their functions in ALCL and HL are unknown. In the current study, we investigated the physiological roles of EP300 and CREBBP in both ALCL and HL, and exploited the therapeutic potential of EP300/CREBBP small molecule inhibitors that target either the HAT or bromodomain activities. Our studies demonstrated distinct roles for EP300 and CREBBP in supporting the viability of ALCL and HL, which was bolstered by the transcriptome analyses. Specifically, EP300 but not CREBBP directly modulated the expression of oncogenic MYC/IRF4 network, surface receptor CD30, immunoregulatory cytokines IL10 and LTA, and immune checkpoint protein PD-L1. Importantly, EP300/CREBBP HAT inhibitor A-485 and bromodomain inhibitor CPI-637 exhibited strong activities against ALCL and HL in vitro and in xenograft mouse models, and inhibited PD-L1 mediated tumor immune escape. Thus, our studies revealed critical insights into the physiological roles of EP300/CREBBP in these lymphomas, and provided opportunities for developing novel strategies for both targeted and immune therapies.
Collapse
|
23
|
Li Y, Ge J, Zhao X, Xu M, Gou M, Xie B, Huang J, Sun Q, Sun L, Bai X, Tan S, Wang X, Dong C. Cell autonomous expression of BCL6 is required to maintain lineage identity of mouse CCR6+ ILC3s. J Exp Med 2023; 220:213808. [PMID: 36651876 PMCID: PMC9856750 DOI: 10.1084/jem.20220440] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 11/04/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Innate lymphoid cells (ILC) are similar to T helper (Th) cells in expression of cytokines and transcription factors. For example, RORγt is the lineage-specific transcription factor for both ILC3 and Th17 cells. However, the ILC counterpart for BCL6-expressing T follicular helper (Tfh) cells has not been defined. Here, we report that in the ILC compartment, BCL6 is selectively co-expressed with not only CXCR5 but also RORγt and CCR6 in ILC3 from multiple tissues. BCL6-deficient ILC3 produces enhanced levels of IL-17A and IL-22. More importantly, phenotypic and single-cell ATAC-seq analysis show that absence of BCL6 in mature ILC3 increases the numbers of ILC1 and transitional cells co-expressing ILC3 and ILC1 marker genes. A lineage-tracing experiment further reveals BCL6+ ILC3 to ILC1 trans-differentiation under steady state. Finally, microbiota promote BCL6 expression in colonic CCR6+ ILC3 and thus reinforce their stability. Collectively, our data have demonstrated that CCR6+ ILC3 have both Th17 and Tfh programs and that BCL6 expression in these cells functions to maintain their lineage identity.
Collapse
Affiliation(s)
- Yuling Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China,Tsinghua University-Peking University Center for Life Sciences, Tsinghua University, Beijing, China
| | - Jing Ge
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Miao Xu
- Broad institute of MIT and Harvard, Cambridge, MA, USA
| | - Mengting Gou
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Bowen Xie
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Jinling Huang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Qinli Sun
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Lin Sun
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Xue Bai
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Sangnee Tan
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohu Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China,Tsinghua University-Peking University Center for Life Sciences, Tsinghua University, Beijing, China,Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China,Research Unit of Immune Regulation and Immune Diseases of Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China,Correspondence to Chen Dong:
| |
Collapse
|
24
|
Huo X, Guo T, Wang K, Yao B, Li D, Li H, Chen W, Wang L, Wu Z. Methylation-based reclassification and risk stratification of skull-base chordomas. Front Oncol 2022; 12:960005. [PMID: 36439461 PMCID: PMC9691996 DOI: 10.3389/fonc.2022.960005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/11/2022] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Skull-base chordomas are rare malignant bone cancers originating from the remnant of the notochord. Survival is variable, and clinical or molecular factors cannot reliably predict their outcomes. This study therefore identified epigenetic subtypes that defined new chordoma epigenetic profiles and their corresponding characteristics. METHODS Methylation profiles of 46 chordoma-resected neoplasms between 2008 and 2014, along with clinical information, were collected. K-means consensus clustering and principal component analysis were used to identify and validate the clusters. Single-sample gene set enrichment analysis, methylCIBERSORT algorithm, and copy number analysis were used to identify the characteristics of the clusters. RESULTS Unsupervised clustering analysis confirmed two clusters with a progression-free survival difference. Gene set enrichment analysis indicated that the early and late estrogen response pathways and the hypoxia pathway were activated whereas the inflammatory and interferon gamma responses were suppressed. Forty-six potential therapeutic targets corresponding to differentially methylated sites were identified from chordoma patients. Subgroups with a worse outcome were characterized by low immune cell infiltration, higher tumor purity, and higher stemness indices. Moreover, copy number amplifications mostly occurred in cluster 1 tumors and the high-risk group. Additionally, the presence of a CCNE1 deletion was exclusively found in the group of chordoma patients with better outcome, whereas RB1 and CDKN2A/2B deletions were mainly found in the group of chordoma patients with worse outcome. CONCLUSIONS Chordoma prognostic epigenetic subtypes were identified, and their corresponding characteristics were found to be variable.
Collapse
Affiliation(s)
- Xulei Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Tengxian Guo
- Department of Neurosurgery, Beijing Fengtai Hospital, Beijing, China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Bohan Yao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Huan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Wei Chen
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Liang Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
25
|
Kuehnle N, Gottwein E. Druggable host gene dependencies in primary effusion lymphoma. Curr Opin Virol 2022; 56:101270. [PMID: 36182745 PMCID: PMC10043043 DOI: 10.1016/j.coviro.2022.101270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) causes primary effusion lymphoma (PEL). Here, we review what is known about human gene essentiality in PEL-derived cell lines. We provide an updated list of PEL-specific human gene dependencies, based on the improved definition of core essential genes across human cancer types. The requirements of PEL cell lines for interferon regulatory factor 4 (IRF4), basic leukine zipper ATF-like transcription factor (BATF), G1/S cyclin D2 (CCND2), CASP8 and FADD like apoptosis regulator (CFLAR), MCL1 apoptosis regulator (MCL1), and murine double minute 2 (MDM2) have been confirmed experimentally. KSHV co-opts IRF4 and BATF to drive superenhancer (SE)-mediated expression of IRF4 itself, MYC, and CCND2. IRF4 dependency of SE-mediated gene expression is shared with Epstein-Barr virus-transformed lymphoblastoid cell lines (LCLs) and human T-cell leukemia virus type 1-transformed adult T-cell leukemia/lymphoma (ATLL) cell lines, as well as several B-cell lymphomas of nonviral etiology. LCLs and ATLL cell lines similarly share dependencies on CCND2 and CFLAR with PEL, but also have distinct gene dependencies. Genetic dependencies could be exploited for therapeutic intervention in PEL and other cancers.
Collapse
Affiliation(s)
- Neil Kuehnle
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Eva Gottwein
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
26
|
Arnold PR, Wen M, Zhang L, Ying Y, Xiao X, Chu X, Wang G, Zhang X, Mao Z, Zhang A, Hamilton DJ, Chen W, Li XC. Suppression of FOXP3 expression by the AP-1 family transcription factor BATF3 requires partnering with IRF4. Front Immunol 2022; 13:966364. [PMID: 36090981 PMCID: PMC9452699 DOI: 10.3389/fimmu.2022.966364] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
FOXP3 is the lineage-defining transcription factor for Tregs, a cell type critical to immune tolerance, but the mechanisms that control FOXP3 expression in Tregs remain incompletely defined, particularly as it relates to signals downstream of TCR and CD28 signaling. Herein, we studied the role of IRF4 and BATF3, two transcription factors upregulated upon T cell activation, to the conversion of conventional CD4+ T cells to FOXP3+ T cells (iTregs) in vitro. We found that IRF4 must partner with BATF3 to bind to a regulatory region in the Foxp3 locus where they cooperatively repress FOXP3 expression and iTreg induction. In addition, we found that interactions of these transcription factors are necessary for glycolytic reprogramming of activated T cells that is antagonistic to FOXP3 expression and stability. As a result, Irf4 KO iTregs show increased demethylation of the critical CNS2 region in the Foxp3 locus. Together, our findings provide important insights how BATF3 and IRF4 interactions integrate activating signals to control CD4+ cell fate decisions and govern Foxp3 expression.
Collapse
Affiliation(s)
- Preston R. Arnold
- College of Medicine, Texas A&M Health, Bryan, TX, United States
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Mou Wen
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Lei Zhang
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Yuanlin Ying
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Xiang Xiao
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Xiufeng Chu
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Guangchuan Wang
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Xiaolong Zhang
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Zhuyun Mao
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
| | - Aijun Zhang
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, TX, United States
- Center for Bioenergetics, Houston Methodist Hospital and Research Institute, Texas Medical Center, Houston, TX, United States
- Weill Cornell Medical College of Cornell University, New York, NY, United States
| | - Dale J. Hamilton
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, TX, United States
- Center for Bioenergetics, Houston Methodist Hospital and Research Institute, Texas Medical Center, Houston, TX, United States
- Weill Cornell Medical College of Cornell University, New York, NY, United States
| | - Wenhao Chen
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY, United States
| | - Xian C. Li
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Texas Medical Center, Houston, TX, United States
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY, United States
- *Correspondence: Xian C. Li,
| |
Collapse
|
27
|
Liang HC. IL-2/IL-2R signaling and IL-2Rα-targeted therapy in anaplastic large cell lymphoma. PATHOLOGIE (HEIDELBERG, GERMANY) 2022; 43:25-30. [PMID: 36094651 DOI: 10.1007/s00292-022-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
Anaplastic large cell lymphoma (ALCL) is a CD30-positive non-Hodgkin's T‑cell lymphoma. Despite the implementation of CD30 antibody-drug conjugate-targeted therapy into front-line treatment regimens, the prognosis of some subtypes of the disease remains unsatisfactory. In the relapsed/refractory setting, effective second-line treatment options are still lacking. However, it has been reported that blockade of direct downstream targets of activator protein‑1 (AP-1) transcription factors, which are highly dysregulated in ALCL, results in complete and sustained remission in late-stage relapsed/refractory anaplastic lymphoma kinase (ALK)-positive ALCL patients. Moreover, it has been identified that involvement of the BATF3/AP‑1 module promotes lymphomagenesis via oncogenic BATF3/IL-2/IL-2R signaling through hyperphosphorylation of ERK1/2, STAT1, and STAT5 in ALCL cells regardless of their ALK status. Therefore, targeting BATF3/IL-2/IL-2R signaling may represent a novel therapeutic alternative for ALCL patients.
Collapse
Affiliation(s)
- Huan-Chang Liang
- Human Oncology & Pathogenesis Program (HOPP), Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
28
|
Long Noncoding RNA BCYRN1 Recruits BATF to Promote TM4SF1 Upregulation and Enhance HCC Cell Proliferation and Invasion. DISEASE MARKERS 2022; 2022:1561607. [PMID: 35730016 PMCID: PMC9206761 DOI: 10.1155/2022/1561607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/29/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common form of cancer for which a subset of reliable clinical biomarkers has been defined. However, other factors including long noncoding RNAs (lncRNAs) can also regulate HCC development. This study was thus designed to understand how the lncRNA Brain cytoplasmic RNA 1 (BCYRN1) modulates HCC progression. Bioinformatics approaches were used to identify genes, lncRNAs, and transcription factors that were differentially expressed in the context of HCC, after which the relative expression of BCYRN1 in HCC and control tissues was assessed via qPCR. The ability of BCYRN1 to bind the transcription factor BATF was further evaluated in an RNA immunoprecipitation (RIP) assay, while chromatin immunoprecipitation (ChIP) was used to gauge the binding of the TM4SF1 promoter by BATF. Luciferase reporter assays were also used to assess the association between BCYRN1 and the TM4SF1 promoter. Subsequent loss- and gain-of-function assays were then conducted to explore the effects of altering BCYRN1 expression levels on the proliferative, invasive, and migratory activity of HCC cells. BCYRN1 upregulation was associated with poorer clinical outcomes in HCC patients, and knocking down this lncRNA impaired HCC cell migration and invasion. From a mechanistic perspective, BATF was recruited to the TM4SF1 promoter by BCYRN1, and reducing the expression of this lncRNA was sufficient to constrain xenograft tumor growth in mice. These results highlight BCYRN1 as a putative therapeutic target in HCC tumors.
Collapse
|
29
|
Jia C, Ma Y, Wang M, Liu W, Tang F, Chen J. Evidence of Omics, Immune Infiltration, and Pharmacogenomics for BATF in a Pan-Cancer Cohort. Front Mol Biosci 2022; 9:844721. [PMID: 35573731 PMCID: PMC9098817 DOI: 10.3389/fmolb.2022.844721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Cytotoxic CD8+ T-cell exhaustion is the major barrier for immunotherapy in tumors. Recent studies have reported that the basic leucine zipper activating transcription factor–like transcription factor (BATF) is responsible for countering cytotoxic CD8+ T-cell exhaustion. Nevertheless, the expression and roles of BATF in tumors have been poorly explored. Methods: In the present study, we conducted a multi-omics analysis, including gene expression, methylation status, DNA alterations, pharmacogenomics, and survival status based on data from the Cancer Genome Atlas (TCGA) database to discern expression patterns and prognostic roles of BATF in tumors. We also explored potential roles of BATF in a pan-cancer cohort by performing immune infiltration, Gene Ontology (GO) enrichment, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. In vitro assay was also performed to explore roles of BATF in tumor cells. Results: We found that BATF was aberrantly upregulated in 27 types of tumors with respect to the corresponding normal tissues. Abnormal BATF expression in tumors predicted survival times of patients in a tissue-dependent manner. The results of GO, immune infiltration, and KEGG analysis revealed that increased BATF expression in tumors participated in modulating immune cell infiltration via immune-related pathways. BATF expression could also predict immunotherapeutic and chemotherapy responses in cancers. Moreover, knockdown of BATF suppresses tumor cell viability. Conclusion: Our present study reports the vital roles of BATF in tumors and provides a theoretical basis for targeting BATF therapy.
Collapse
|
30
|
López C, Schleussner N, Bernhart SH, Kleinheinz K, Sungalee S, Sczakiel HL, Kretzmer H, Toprak UH, Glaser S, Wagener R, Ammerpohl O, Bens S, Giefing M, González Sánchez JC, Apic G, Hübschmann D, Janz M, Kreuz M, Mottok A, Müller JM, Seufert J, Hoffmann S, Korbel JO, Russell RB, Schüle R, Trümper L, Klapper W, Radlwimmer B, Lichter P, Küppers R, Schlesner M, Mathas S, Siebert R. Focal structural variants revealed by whole genome sequencing disrupt the histone demethylase KDM4C in B-cell lymphomas. Haematologica 2022; 108:543-554. [PMID: 35522148 PMCID: PMC9890021 DOI: 10.3324/haematol.2021.280005] [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: 09/13/2021] [Indexed: 02/03/2023] Open
Abstract
Histone methylation-modifiers, such as EZH2 and KMT2D, are recurrently altered in B-cell lymphomas. To comprehensively describe the landscape of alterations affecting genes encoding histone methylation-modifiers in lymphomagenesis we investigated whole genome and transcriptome data of 186 mature B-cell lymphomas sequenced in the ICGC MMML-Seq project. Besides confirming common alterations of KMT2D (47% of cases), EZH2 (17%), SETD1B (5%), PRDM9 (4%), KMT2C (4%), and SETD2 (4%), also identified by prior exome or RNA-sequencing studies, we here found recurrent alterations to KDM4C in chromosome 9p24, encoding a histone demethylase. Focal structural variation was the main mechanism of KDM4C alterations, and was independent from 9p24 amplification. We also identified KDM4C alterations in lymphoma cell lines including a focal homozygous deletion in a classical Hodgkin lymphoma cell line. By integrating RNA-sequencing and genome sequencing data we predict that KDM4C structural variants result in loss-offunction. By functional reconstitution studies in cell lines, we provide evidence that KDM4C can act as a tumor suppressor. Thus, we show that identification of structural variants in whole genome sequencing data adds to the comprehensive description of the mutational landscape of lymphomas and, moreover, establish KDM4C as a putative tumor suppressive gene recurrently altered in subsets of B-cell derived lymphomas.
Collapse
Affiliation(s)
- Cristina López
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany,*CL and NS contributed equally as co-first authors
| | - Nikolai Schleussner
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany,*CL and NS contributed equally as co-first authors
| | - Stephan H. Bernhart
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany,Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Kortine Kleinheinz
- Department for Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and Bioquant, University of Heidelberg, Heidelberg, Germany
| | | | - Henrike L. Sczakiel
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany
| | - Helene Kretzmer
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany,Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany,Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Umut H. Toprak
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), Heidelberg, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany,Hopp-Children’s Cancer Center at the NCT Heidelberg (KiTZ), Division of Neuroblastoma Genomics (B087), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Rabea Wagener
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
| | - Ole Ammerpohl
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
| | - Susanne Bens
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany
| | - Maciej Giefing
- Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Gordana Apic
- BioQuant and Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Heidelberg, Germany
| | - Daniel Hübschmann
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Department for Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and Bioquant, University of Heidelberg, Heidelberg, Germany,German Cancer Consortium (DKTK), Heidelberg, Germany,Heidelberg Institute of Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Martin Janz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany
| | - Markus Kreuz
- Institute for Medical Informatics Statistics and Epidemiology, Leipzig, Germany
| | - Anja Mottok
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Judith M. Müller
- Klinik fur Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Julian Seufert
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steve Hoffmann
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany,Transcriptome Bioinformatics, LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany,Leibniz Institute on Ageing-Fritz Lipmann Institute (FLI), Computational Biology, Jena, Germany
| | - Jan O. Korbel
- EMBL Heidelberg, Genome Biology Unit, Heidelberg,, Germany
| | - Robert B. Russell
- BioQuant and Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Heidelberg, Germany
| | - Roland Schüle
- Klinik fur Urologie und Zentrale Klinische Forschung, Klinikum der Albert-Ludwigs-Universität Freiburg, Freiburg, Germany,BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Lorenz Trümper
- Department of Hematology and Oncology, Georg-August-University of Göttingen, Göttingen, Germany
| | - Wolfram Klapper
- Hematopathology Section, Christian-Albrechts-University, Kiel, Germany
| | - Bernhard Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Küppers
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany, and German Cancer Consortium (DKTK)
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics (B240), German Cancer Research Center (DKFZ), Heidelberg, Germany,Biomedical Informatics, Data Mining and Data Analytics, Augsburg University, Augsburg, Germany
| | - Stephan Mathas
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany, and Experimental and Clinical Research Center, a joint cooperation between the MDC and the Charité, Berlin, Germany,SM and RS contributed equally as co-senior authors
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany,Institute of Human Genetics, Christian-Albrechts-University, Kiel, Germany,SM and RS contributed equally as co-senior authors
| |
Collapse
|
31
|
Resistance to Targeted Agents Used to Treat Paediatric ALK-Positive ALCL. Cancers (Basel) 2021; 13:cancers13236003. [PMID: 34885113 PMCID: PMC8656581 DOI: 10.3390/cancers13236003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary In general, the non-Hodgkin lymphoma (NHL), anaplastic large cell lymphoma (ALCL) diagnosed in childhood has a good survival outcome when treated with multi-agent chemotherapy. However, side effects of treatment are common, and outcomes are poorer after relapse, which occurs in up to 30% of cases. New drugs are required that are more effective and have fewer side effects. Targeted therapies are potential solutions to these problems, however, the development of resistance may limit their impact. This review summarises the potential resistance mechanisms to these targeted therapies. Abstract Non-Hodgkin lymphoma (NHL) is the third most common malignancy diagnosed in children. The vast majority of paediatric NHL are either Burkitt lymphoma (BL), diffuse large B-cell lymphoma (DLBCL), anaplastic large cell lymphoma (ALCL), or lymphoblastic lymphoma (LL). Multi-agent chemotherapy is used to treat all of these types of NHL, and survival is over 90% but the chemotherapy regimens are intensive, and outcomes are generally poor if relapse occurs. Therefore, targeted therapies are of interest as potential solutions to these problems. However, the major problem with all targeted agents is the development of resistance. Mechanisms of resistance are not well understood, but increased knowledge will facilitate optimal management strategies through improving our understanding of when to select each targeted agent, and when a combinatorial approach may be helpful. This review summarises currently available knowledge regarding resistance to targeted therapies used in paediatric anaplastic lymphoma kinase (ALK)-positive ALCL. Specifically, we outline where gaps in knowledge exist, and further investigation is required in order to find a solution to the clinical problem of drug resistance in ALCL.
Collapse
|
32
|
Nagel S, Meyer C. Establishment of the TBX-code reveals aberrantly activated T-box gene TBX3 in Hodgkin lymphoma. PLoS One 2021; 16:e0259674. [PMID: 34807923 PMCID: PMC8608327 DOI: 10.1371/journal.pone.0259674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/22/2021] [Indexed: 11/23/2022] Open
Abstract
T-box genes encode transcription factors which control basic processes in development of several tissues including cell differentiation in the hematopoietic system. Here, we analyzed the physiological activities of all 17 human T-box genes in early hematopoiesis and in lymphopoiesis including developing and mature B-cells, T-cells, natural killer (NK)-cells and innate lymphoid cells. The resultant expression pattern comprised six genes, namely EOMES, MGA, TBX1, TBX10, TBX19 and TBX21. We termed this gene signature TBX-code which enables discrimination of normal and aberrant activities of T-box genes in lymphoid malignancies. Accordingly, expression analysis of T-box genes in Hodgkin lymphoma (HL) patients using a public profiling dataset revealed overexpression of EOMES, TBX1, TBX2, TBX3, TBX10, TBX19, TBX21 and TBXT while MGA showed aberrant downregulation. Analysis of T-cell acute lymphoid leukemia patients indicated aberrant overexpression of six T-box genes while no deregulated T-box genes were detected in anaplastic large cell lymphoma patients. As a paradigm we focused on TBX3 which was ectopically activated in about 6% of HL patients analyzed. Normally, TBX3 is expressed in tissues like lung, adrenal gland and retina but not in hematopoiesis. HL cell line KM-H2 expressed enhanced TBX3 levels and was used as an in vitro model to identify upstream regulators and downstream targets in this malignancy. Genomic studies of this cell line showed focal amplification of the TBX3 locus at 12q24 which may underlie its aberrant expression. In addition, promoter analysis and comparative expression profiling of HL cell lines followed by knockdown experiments revealed overexpressed transcription factors E2F4 and FOXC1 and chromatin modulator KDM2B as functional activators. Furthermore, we identified repressed target genes of TBX3 in HL including CDKN2A, NFKBIB and CD19, indicating its respective oncogenic function in proliferation, NFkB-signaling and B-cell differentiation. Taken together, we have revealed a lymphoid TBX-code and used it to identify an aberrant network around deregulated T-box gene TBX3 in HL which promotes hallmark aberrations of this disease. These findings provide a framework for future studies to evaluate deregulated T-box genes in lymphoid malignancies.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ–German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- * E-mail:
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ–German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
33
|
Nagel S. The Role of NKL Homeobox Genes in T-Cell Malignancies. Biomedicines 2021; 9:biomedicines9111676. [PMID: 34829904 PMCID: PMC8615965 DOI: 10.3390/biomedicines9111676] [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: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Homeobox genes encode transcription factors controlling basic developmental processes. The homeodomain is encoded by the homeobox and mediates sequence-specific DNA binding and interaction with cofactors, thus operating as a basic regulatory platform. Similarities in their homeobox sequences serve to arrange these genes in classes and subclasses, including NKL homeobox genes. In accordance with their normal functions, deregulated homeobox genes contribute to carcinogenesis along with hematopoietic malignancies. We have recently described the physiological expression of eleven NKL homeobox genes in the course of hematopoiesis and termed this gene expression pattern NKL-code. Due to the developmental impact of NKL homeobox genes these data suggest a key role for their activity in the normal regulation of hematopoietic cell differentiation including T-cells. On the other hand, aberrant overexpression of NKL-code members or ectopical activation of non-code members has been frequently reported in lymphoid and myeloid leukemia/lymphoma, demonstrating their oncogenic impact in the hematopoietic compartment. Here, we provide an overview of the NKL-code in normal hematopoiesis and discuss the oncogenic role of deregulated NKL homeobox genes in T-cell malignancies.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| |
Collapse
|
34
|
Roma S, Carpen L, Raveane A, Bertolini F. The Dual Role of Innate Lymphoid and Natural Killer Cells in Cancer. from Phenotype to Single-Cell Transcriptomics, Functions and Clinical Uses. Cancers (Basel) 2021; 13:cancers13205042. [PMID: 34680190 PMCID: PMC8533946 DOI: 10.3390/cancers13205042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Innate lymphoid cells (ILCs), a family of innate immune cells including natural killers (NKs), play a multitude of roles in first-line cancer control, in escape from immunity and in cancer progression. In this review, we summarize preclinical and clinical data on ILCs and NK cells concerning their phenotype, function and clinical applications in cellular therapy trials. We also describe how single-cell transcriptome sequencing has been used and forecast how it will be used to better understand ILC and NK involvement in cancer control and progression as well as their therapeutic potential. Abstract The role of innate lymphoid cells (ILCs), including natural killer (NK) cells, is pivotal in inflammatory modulation and cancer. Natural killer cell activity and count have been demonstrated to be regulated by the expression of activating and inhibitory receptors together with and as a consequence of different stimuli. The great majority of NK cell populations have an anti-tumor activity due to their cytotoxicity, and for this reason have been used for cellular therapies in cancer patients. On the other hand, the recently classified helper ILCs are fundamentally involved in inflammation and they can be either helpful or harmful in cancer development and progression. Tissue niche seems to play an important role in modulating ILC function and conversion, as observed at the transcriptional level. In the past, these cell populations have been classified by the presence of specific cellular receptor markers; more recently, due to the advent of single-cell RNA sequencing (scRNA-seq), it has been possible to also explore them at the transcriptomic level. In this article we review studies on ILC (and NK cell) classification, function and their involvement in cancer. We also summarize the potential application of NK cells in cancer therapy and give an overview of the most recent studies involving ILCs and NKs at scRNA-seq, focusing on cancer. Finally, we provide a resource for those who wish to start single-cell transcriptomic analysis on the context of these innate lymphoid cell populations.
Collapse
|
35
|
Wurster KD, Costanza M, Kreher S, Glaser S, Lamprecht B, Schleussner N, Anagnostopoulos I, Hummel M, Jöhrens K, Stein H, Molina A, Diepstra A, Gillissen B, Köchert K, Siebert R, Merkel O, Kenner L, Janz M, Mathas S. Aberrant Expression of and Cell Death Induction by Engagement of the MHC-II Chaperone CD74 in Anaplastic Large Cell Lymphoma (ALCL). Cancers (Basel) 2021; 13:cancers13195012. [PMID: 34638496 PMCID: PMC8507667 DOI: 10.3390/cancers13195012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Anaplastic large cell lymphoma (ALCL) is a lymphoid malignancy considered to be derived from T cells. Currently, two types of systemic ALCL are distinguished: anaplastic lymphoma kinase (ALK)-positive and ALK-negative ALCL. Although ALK+ and ALK− ALCL differ at the genomic and molecular levels, various key biological and molecular features are highly similar between both entities. We have developed the concept that both ALCL entities share a common principle of pathogenesis. In support of this concept, we here describe a common deregulation of CD74, which is usually not expressed in T cells, in ALCL. Ligation of CD74 induces cell death of ALCL cells in various conditions, and an anti-CD74-directed antibody-drug conjugate efficiently kills ALCL cell lines. Furthermore, we reveal expression of the proto-oncogene and known CD74 interaction partner MET in a fraction of ALCL cases. These data give insights into ALCL pathogenesis and might help to develop new treatment strategies for ALCL. Abstract In 50–60% of cases, systemic anaplastic large cell lymphoma (ALCL) is characterized by the t(2;5)(p23;q35) or one of its variants, considered to be causative for anaplastic lymphoma kinase (ALK)-positive (ALK+) ALCL. Key pathogenic events in ALK-negative (ALK−) ALCL are less well defined. We have previously shown that deregulation of oncogenic genes surrounding the chromosomal breakpoints on 2p and 5q is a unifying feature of both ALK+ and ALK− ALCL and predisposes for occurrence of t(2;5). Here, we report that the invariant chain of the MHC-II complex CD74 or li, which is encoded on 5q32, can act as signaling molecule, and whose expression in lymphoid cells is usually restricted to B cells, is aberrantly expressed in T cell-derived ALCL. Accordingly, ALCL shows an altered DNA methylation pattern of the CD74 locus compared to benign T cells. Functionally, CD74 ligation induces cell death of ALCL cells. Furthermore, CD74 engagement enhances the cytotoxic effects of conventional chemotherapeutics in ALCL cell lines, as well as the action of the ALK-inhibitor crizotinib in ALK+ ALCL or of CD95 death-receptor signaling in ALK− ALCL. Additionally, a subset of ALCL cases expresses the proto-oncogene MET, which can form signaling complexes together with CD74. Finally, we demonstrate that the CD74-targeting antibody-drug conjugate STRO-001 efficiently and specifically kills CD74-positive ALCL cell lines in vitro. Taken together, these findings enabled us to demonstrate aberrant CD74-expression in ALCL cells, which might serve as tool for the development of new treatment strategies for this lymphoma entity.
Collapse
Affiliation(s)
- Kathrin D. Wurster
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Mariantonia Costanza
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Stephan Kreher
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University, Ulm University Medical Center, 89081 Ulm, Germany; (S.G.); (R.S.)
| | - Björn Lamprecht
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Nikolai Schleussner
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Ioannis Anagnostopoulos
- Institute of Pathology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany; (I.A.); (K.J.)
| | - Michael Hummel
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute of Pathology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany; (I.A.); (K.J.)
| | - Korinna Jöhrens
- Institute of Pathology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany; (I.A.); (K.J.)
| | | | - Arturo Molina
- Sutro Biopharma, South San Francisco, CA 94080, USA;
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands;
| | - Bernd Gillissen
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Karl Köchert
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University, Ulm University Medical Center, 89081 Ulm, Germany; (S.G.); (R.S.)
| | - Olaf Merkel
- Unit of Experimental and Laboratory Animal Pathology, Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (O.M.); (L.K.)
- European Research Initiative on ALK-related malignancies (ERIA), 1090 Vienna, Austria
| | - Lukas Kenner
- Unit of Experimental and Laboratory Animal Pathology, Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (O.M.); (L.K.)
- European Research Initiative on ALK-related malignancies (ERIA), 1090 Vienna, Austria
| | - Martin Janz
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Stephan Mathas
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- European Research Initiative on ALK-related malignancies (ERIA), 1090 Vienna, Austria
- Correspondence: ; Tel.: +49-30-94062863; Fax: +49-30-94063124
| |
Collapse
|
36
|
Liang HC, Costanza M, Prutsch N, Zimmerman MW, Gurnhofer E, Montes-Mojarro IA, Abraham BJ, Prokoph N, Stoiber S, Tangermann S, Lobello C, Oppelt J, Anagnostopoulos I, Hielscher T, Pervez S, Klapper W, Zammarchi F, Silva DA, Garcia KC, Baker D, Janz M, Schleussner N, Fend F, Pospíšilová Š, Janiková A, Wallwitz J, Stoiber D, Simonitsch-Klupp I, Cerroni L, Pileri S, de Leval L, Sibon D, Fataccioli V, Gaulard P, Assaf C, Knörr F, Damm-Welk C, Woessmann W, Turner SD, Look AT, Mathas S, Kenner L, Merkel O. Super-enhancer-based identification of a BATF3/IL-2R-module reveals vulnerabilities in anaplastic large cell lymphoma. Nat Commun 2021; 12:5577. [PMID: 34552066 PMCID: PMC8458384 DOI: 10.1038/s41467-021-25379-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
Anaplastic large cell lymphoma (ALCL), an aggressive CD30-positive T-cell lymphoma, comprises systemic anaplastic lymphoma kinase (ALK)-positive, and ALK-negative, primary cutaneous and breast implant-associated ALCL. Prognosis of some ALCL subgroups is still unsatisfactory, and already in second line effective treatment options are lacking. To identify genes defining ALCL cell state and dependencies, we here characterize super-enhancer regions by genome-wide H3K27ac ChIP-seq. In addition to known ALCL key regulators, the AP-1-member BATF3 and IL-2 receptor (IL2R)-components are among the top hits. Specific and high-level IL2R expression in ALCL correlates with BATF3 expression. Confirming a regulatory link, IL-2R-expression decreases following BATF3 knockout, and BATF3 is recruited to IL2R regulatory regions. Functionally, IL-2, IL-15 and Neo-2/15, a hyper-stable IL-2/IL-15 mimic, accelerate ALCL growth and activate STAT1, STAT5 and ERK1/2. In line, strong IL-2Rα-expression in ALCL patients is linked to more aggressive clinical presentation. Finally, an IL-2Rα-targeting antibody-drug conjugate efficiently kills ALCL cells in vitro and in vivo. Our results highlight the importance of the BATF3/IL-2R-module for ALCL biology and identify IL-2Rα-targeting as a promising treatment strategy for ALCL. Anaplastic large cell lymphoma (ALCL) is an aggressive T-cell lymphoma often with poor prognosis. To identify genes defining ALCL cell state and dependencies, the authors here characterize ALCL-specific super-enhancers and describe the BATF3/IL-2R−module as a therapeutic opportunity for ALCL.
Collapse
Affiliation(s)
- Huan-Chang Liang
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.,European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK
| | - Mariantonia Costanza
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany
| | - Nicole Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Mark W Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Elisabeth Gurnhofer
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Ivonne A Montes-Mojarro
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Institute of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nina Prokoph
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Stefan Stoiber
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory (CDL) for Applied Metabolomics, Medical University of Vienna, Vienna, Austria
| | - Simone Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Cosimo Lobello
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Jan Oppelt
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | | | - Thomas Hielscher
- German Cancer Consortium (DKTK) German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shahid Pervez
- Department of Pathology and Laboratory Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | | | - Daniel-Adriano Silva
- Institute for Protein Design, University of Washington, Seattle, WA, USA.,Department of Biochemistry, University of Washington, Seattle, WA, USA.,Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA, USA.,Department of Biochemistry, University of Washington, Seattle, WA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Martin Janz
- Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany
| | - Nikolai Schleussner
- Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany
| | - Falko Fend
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Institute of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Šárka Pospíšilová
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic.,Department of Internal Medicine-Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Andrea Janiková
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Department of Internal Medicine-Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Jacqueline Wallwitz
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Dagmar Stoiber
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Ingrid Simonitsch-Klupp
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Lorenzo Cerroni
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Stefano Pileri
- Division of Haematopathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital (CHUV) and Lausanne University, Lausanne, Switzerland
| | - David Sibon
- Hematology Department, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, and Institut Necker-Enfants Malades, INSERM UMR1151 (Normal and pathological lymphoid differentiation), Université de Paris, Paris, France
| | - Virginie Fataccioli
- Department of Pathology, Henri Mondor University Hospital, AP-HP, INSERM U955, University Paris East, Créteil, France
| | - Philippe Gaulard
- Department of Pathology, Henri Mondor University Hospital, AP-HP, INSERM U955, University Paris East, Créteil, France
| | - Chalid Assaf
- Department of Dermatology, HELIOS Hospital Krefeld, Krefeld, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Knörr
- Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Damm-Welk
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Wilhelm Woessmann
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Suzanne D Turner
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Stephan Mathas
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK. .,Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany. .,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany. .,German Cancer Consortium (DKTK) German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Lukas Kenner
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria. .,European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK. .,Christian Doppler Laboratory (CDL) for Applied Metabolomics, Medical University of Vienna, Vienna, Austria. .,Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria. .,Center for Biomarker Research in Medicine (CBMed) Core Lab 2, Medical University of Vienna, Vienna, Austria.
| | - Olaf Merkel
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria. .,European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.
| |
Collapse
|
37
|
Merlio JP, Kadin ME. Cytokines, Genetic Lesions and Signaling Pathways in Anaplastic Large Cell Lymphomas. Cancers (Basel) 2021; 13:4256. [PMID: 34503066 PMCID: PMC8428234 DOI: 10.3390/cancers13174256] [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: 07/22/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022] Open
Abstract
ALCL is a tumor of activated T cells and possibly innate lymphoid cells with several subtypes according to clinical presentation and genetic lesions. On one hand, the expression of transcription factors and cytokine receptors triggers signaling pathways. On the other hand, ALCL tumor cells also produce many proteins including chemokines, cytokines and growth factors that affect patient symptoms. Examples are accumulation of granulocytes stimulated by IL-8, IL-17, IL-9 and IL-13; epidermal hyperplasia and psoriasis-like skin lesions due to IL-22; and fever and weight loss in response to IL-6 and IFN-γ. In this review, we focus on the biology of the main ALCL subtypes as the identification of signaling pathways and ALCL-derived cytokines offers opportunities for targeted therapies.
Collapse
Affiliation(s)
- Jean-Philippe Merlio
- Tumor Biology and Tumor Bank Laboratory, Centre Hospitalier et Universitaire de Bordeaux, 33600 Pessac, France
- INSERM U1053, University Bordeaux, 33000 Bordeaux, France
| | - Marshall E. Kadin
- Department of Pathology and Laboratory Medicine, Brown University Alpert School of Medicine, Providence, RI 02903, USA
- Department of Dermatology, Boston University, Boston, MA 02215, USA
| |
Collapse
|
38
|
Yuan X, Rasul F, Nashan B, Sun C. Innate lymphoid cells and cancer: Role in tumor progression and inhibition. Eur J Immunol 2021; 51:2188-2205. [PMID: 34189723 PMCID: PMC8457100 DOI: 10.1002/eji.202049033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/12/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILCs), a critical component of the immune system, have recently been nominated as emerging players associated with tumor progression and inhibition. ILCs are classified into five groups: natural killer (NK) cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer (LTis) cells. NK cells and ILC1s are mainly involved in antitumor activities due to their cytotoxic and cytokine production capabilities, respectively. The current understanding of the heterogeneous behavior of ILC2s and ILC3s in tumors is limited and incomplete. Mostly, their dual roles are modulated by their resident tissues, released cytokines, cancer types, and plasticity. Based on overlap RORγt and cytokine expression, the LTi cells were previously considered part of the ILC3s ontogeny, which are essential for the formation of the secondary lymphoid organs during embryogenesis. Indeed, these facts highlight the urgency in understanding the respective mechanisms that shape the phenotypes and responses of ILCs, either on the repressive or proliferative side in the tumor microenvironment (TME). This review aims to provide an updated view of ILCs biology with respect to tumorigenesis, including a description of ILC plasticity, their interaction with other immune cells and communication with components of the TME. Taken together, targeting ILCs for cancer immunotherapy could be a promising approach against tumors that needs to be further study.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Faiz Rasul
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Björn Nashan
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Cheng Sun
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P. R. China
| |
Collapse
|
39
|
Usefulness of BATF3 Immunohistochemistry in Diagnosing Classical Hodgkin Lymphoma. Diagnostics (Basel) 2021; 11:diagnostics11061123. [PMID: 34202976 PMCID: PMC8234195 DOI: 10.3390/diagnostics11061123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
It is well recognized that the AP-1 transcription factor BATF3 is constitutively expressed in Hodgkin/Reed-Sternberg (HRS) cells, but its potential as a diagnostic marker for classical Hodgkin lymphoma (cHL) has not yet been addressed. In this study, we performed immunohistochemistry and analyzed the BATF3 expression in lymphoma cells on 218 lymphoma samples belonging to 14 different lymphoma entities. We observed varying degrees of BATF3 expression in nearly half of the cases (n = 100) with BATF3 expression being a constitutive feature of cHL (n = 53) and anaplastic large cell lymphoma (ALCL). By scoring BATF3 expression (BATF3-score) we observed constitutively high BATF3-scores in cHL and ALCL and low to moderate BATF3-scores in all other entities examined. Western blot analysis confirmed BATF3 protein expression in cell lysates from cHL cell lines (n = 7). Thus, BATF3 can be considered a useful IHC marker for the diagnosis of cHL as it is highly sensitive and sufficiently specific when analyzed by BATF3-scoring.
Collapse
|
40
|
Huang H, Fu J, Zhang L, Xu J, Li D, Onwuka JU, Zhang D, Zhao L, Sun S, Zhu L, Zheng T, Jia C, Cui B, Zhao Y. Integrative Analysis of Identifying Methylation-Driven Genes Signature Predicts Prognosis in Colorectal Carcinoma. Front Oncol 2021; 11:629860. [PMID: 34178621 PMCID: PMC8231008 DOI: 10.3389/fonc.2021.629860] [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: 11/16/2020] [Accepted: 05/24/2021] [Indexed: 01/20/2023] Open
Abstract
Background Aberrant DNA methylation is a critical regulator of gene expression and plays a crucial role in the occurrence, progression, and prognosis of colorectal cancer (CRC). We aimed to identify methylation-driven genes by integrative epigenetic and transcriptomic analysis to predict the prognosis of CRC patients. Methods Methylation-driven genes were selected for CRC using a MethylMix algorithm and LASSO regression screening strategy, and were further used to construct a prognostic risk-assessment model. The Cancer Genome Atlas (TCGA) database was obtained as the training set for both the screening of methylation-driven genes and the effect of genes signature on CRC prognosis. Then, the prognostic genes signature was validated in three independent expression arrays of CRC data from Gene Expression Omnibus (GEO). Results We identified 143 methylation-driven genes, of which the combination of BATF, PHYHIPL, RBP1, and PNPLA4 expression levels was screened as a better prognostic model with the best area under the curve (AUC) (AUC = 0.876). Compared with patients in the low-risk group, CRC patients in the high-risk group had significantly poorer overall survival in the training set (HR = 2.184, 95% CI: 1.404–3.396, P < 0.001). Similar results were observed in the validation set. Moreover, VanderWeele’s mediation analysis indicated that the effect of methylation on prognosis was mediated by the levels of their expression (HRindirect = 1.473, P = 0.001, Proportion mediated, 69.10%). Conclusions We identified a four-gene prognostic signature by integrative analysis and developed a risk-assessment model that is significantly associated with patients’ survival. Methylation-driven genes might be a potential prognostic signature for CRC patients.
Collapse
Affiliation(s)
- Hao Huang
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Jinming Fu
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Lei Zhang
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Jing Xu
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Dapeng Li
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Justina Ucheojor Onwuka
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Ding Zhang
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Liyuan Zhao
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Simin Sun
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Lin Zhu
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Ting Zheng
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Chenyang Jia
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| | - Binbin Cui
- Department of Colorectal Surgery, The Third Hospital of Harbin Medical University, Harbin, China
| | - Yashuang Zhao
- Department of Epidemiology, Public Health School of Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Zhang Z, Lin M, Wang J, Yang F, Yang P, Liu Y, Chen Z, Zheng Y. Calycosin inhibits breast cancer cell migration and invasion by suppressing EMT via BATF/TGF-β1. Aging (Albany NY) 2021; 13:16009-16023. [PMID: 34096887 PMCID: PMC8266341 DOI: 10.18632/aging.203093] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/29/2021] [Indexed: 12/16/2022]
Abstract
In this study, we investigated the effects of calycosin on breast cancer cell progression and their underlying mechanisms. Calycosin dose- and time-dependently inhibited proliferation, migration, and invasion by T47D and MCF-7 breast cancer cells by downregulating basic leucine zipper ATF-like transcription factor (BATF) expression. Moreover, BATF promoted breast cancer cell migration and invasiveness by increasing TGFβ1 mRNA and protein levels. Bioinformatics analysis, dual luciferase reporter assays, and chromatin immunoprecipitation assays confirmed the presence of BATF-binding sites in the promoter sequence of TGFβ1 gene. Calycosin treatment inhibited epithelial-mesenchymal transition (EMT) of breast cancer cells by significantly increasing E-cadherin levels and decreasing N-cadherin, Vimentin, CD147, MMP-2, and MMP-9 levels through downregulation of BATF and TGFβ1. TGFβ1 knockdown reduced the migration and invasiveness of BATF-overexpressing breast cancer cells, whereas incubation with TGFβ1 enhanced the migration and invasiveness of calycosin-treated breast cancer cells. Our findings demonstrated that calycosin inhibited EMT and progression of breast cancer cells by suppressing BATF/TGFβ1 signaling. This suggests calycosin would be a promising therapeutic option for breast cancer patients.
Collapse
Affiliation(s)
- Zhenxia Zhang
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, Guangdong, China
| | - Min Lin
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, Guangdong, China
| | - Junli Wang
- Center of Reproductive Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Fenglian Yang
- School of Pharmacy, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Peikui Yang
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, Guangdong, China
| | - Yaqun Liu
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, Guangdong, China
| | - Zikai Chen
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, Guangdong, China
| | - Yuzhong Zheng
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, Guangdong, China
| |
Collapse
|
42
|
Li WX, Dai SX, An SQ, Sun T, Liu J, Wang J, Liu LG, Xun Y, Yang H, Fan LX, Zhang XL, Liao WQ, You H, Tamagnone L, Liu F, Huang JF, Liu D. Transcriptome integration analysis and specific diagnosis model construction for Hodgkin's lymphoma, diffuse large B-cell lymphoma, and mantle cell lymphoma. Aging (Albany NY) 2021; 13:11833-11859. [PMID: 33885377 PMCID: PMC8109084 DOI: 10.18632/aging.202882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/02/2021] [Indexed: 01/20/2023]
Abstract
Transcriptome differences between Hodgkin's lymphoma (HL), diffuse large B-cell lymphoma (DLBCL), and mantle cell lymphoma (MCL), which are all derived from B cell, remained unclear. This study aimed to construct lymphoma-specific diagnostic models by screening lymphoma marker genes. Transcriptome data of HL, DLBCL, and MCL were obtained from public databases. Lymphoma marker genes were screened by comparing cases and controls as well as the intergroup differences among lymphomas. A total of 9 HL marker genes, 7 DLBCL marker genes, and 4 MCL marker genes were screened in this study. Most HL marker genes were upregulated, whereas DLBCL and MCL marker genes were downregulated compared to controls. The optimal HL-specific diagnostic model contains one marker gene (MYH2) with an AUC of 0.901. The optimal DLBCL-specific diagnostic model contains 7 marker genes (LIPF, CCDC144B, PRO2964, PHF1, SFTPA2, NTS, and HP) with an AUC of 0.951. The optimal MCL-specific diagnostic model contains 3 marker genes (IGLV3-19, IGKV4-1, and PRB3) with an AUC of 0.843. The present study reveals the transcriptome data-based differences between HL, DLBCL, and MCL, when combined with other clinical markers, may help the clinical diagnosis and prognosis.
Collapse
Affiliation(s)
- Wen-Xing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Shao-Xing Dai
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - San-Qi An
- Biosafety Level-3 Laboratory, Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Tingting Sun
- National School of Development, Peking University, Beijing 100871, China
| | - Justin Liu
- Department of Statistics, University of California, Riverside, CA 92521, USA
| | - Jun Wang
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | | | - Yang Xun
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Hua Yang
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Li-Xia Fan
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xiao-Li Zhang
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Wan-Qin Liao
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Hua You
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Luca Tamagnone
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fang Liu
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jing-Fei Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dahai Liu
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| |
Collapse
|
43
|
IL10RA modulates crizotinib sensitivity in NPM1-ALK+ anaplastic large cell lymphoma. Blood 2021; 136:1657-1669. [PMID: 32573700 DOI: 10.1182/blood.2019003793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/19/2020] [Indexed: 02/08/2023] Open
Abstract
Anaplastic large cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by a hyperactive anaplastic lymphoma kinase (ALK) fusion protein. ALK inhibitors, such as crizotinib, provide alternatives to standard chemotherapy with reduced toxicity and side effects. Children with lymphomas driven by nucleophosmin 1 (NPM1)-ALK fusion proteins achieved an objective response rate to ALK inhibition therapy of 54% to 90% in clinical trials; however, a subset of patients progressed within the first 3 months of treatment. The mechanism for the development of ALK inhibitor resistance is unknown. Through genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) activation and knockout screens in ALCL cell lines, combined with RNA sequencing data derived from ALK inhibitor-relapsed patient tumors, we show that resistance to ALK inhibition by crizotinib in ALCL can be driven by aberrant upregulation of interleukin 10 receptor subunit alpha (IL10RA). Elevated IL10RA expression rewires the STAT3 signaling pathway, bypassing otherwise critical phosphorylation by NPM1-ALK. IL-10RA expression does not correlate with response to standard chemotherapy in pediatric patients, suggesting that a combination of crizotinib and chemotherapy could prevent ALK inhibitor resistance-specific relapse.
Collapse
|
44
|
Yan T, Shen C, Jiang P, Yu C, Guo F, Tian X, Zhu X, Lu S, Han B, Zhong M, Chen J, Liu Q, Chen Y, Zhang J, Hong J, Chen H, Fang JY. Risk SNP-induced lncRNA-SLCC1 drives colorectal cancer through activating glycolysis signaling. Signal Transduct Target Ther 2021; 6:70. [PMID: 33602893 PMCID: PMC7892549 DOI: 10.1038/s41392-020-00446-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/16/2020] [Accepted: 12/03/2020] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play key roles in colorectal carcinogenesis. Here, we aimed to identify the risk SNP-induced lncRNAs and to investigate their roles in colorectal carcinogenesis. First, we identified rs6695584 as the causative SNP in 1q41 locus. The A>G mutation of rs6695584 created a protein-binding motif of BATF, altered the enhancer activity, and subsequently activated lncSLCC1 expression. Further validation in two independent CRC cohorts confirmed the upregulation of lncSLCC1 in CRC tissues, and revealed that increased lncSLCC1 expression was associated with poor survival in CRC patients. Mechanistically, lncRNA-SLCC1 interacted with AHR and transcriptionally activated HK2 expression, the crucial enzyme in glucose metabolism, thereby driving the glycolysis pathway and accelerating CRC tumor growth. The functional assays revealed that lncSLCC1 induced glycolysis activation and tumor growth in CRC mediated by HK2. In addition, HK2 was upregulated in colorectal cancer tissues and positively correlated with lncSLCC1 expression and patient survival. Taken together, our findings reveal a risk SNP-mediated oncogene lncRNA-SLCC1 promotes CRC through activating the glycolysis pathway.
Collapse
Affiliation(s)
- Tingting Yan
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Chaoqin Shen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Penglei Jiang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Chenyang Yu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Fangfang Guo
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Xianglong Tian
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Xiaoqiang Zhu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Shiyuan Lu
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Bingshe Han
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Ming Zhong
- Division of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Jinxian Chen
- Division of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Yingxuan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China
| | - Junfang Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, Shanghai, 201306, China.
| | - Jie Hong
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China.
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China.
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Middle Shandong Road, 200001, Shanghai, China.
| |
Collapse
|
45
|
Congras A, Hoareau-Aveilla C, Caillet N, Tosolini M, Villarese P, Cieslak A, Rodriguez L, Asnafi V, Macintyre E, Egger G, Brousset P, Lamant L, Meggetto F. ALK-transformed mature T lymphocytes restore early thymus progenitor features. J Clin Invest 2021; 130:6395-6408. [PMID: 33141118 DOI: 10.1172/jci134990] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
Anaplastic large cell lymphoma (ALCL) is a mature T cell neoplasm that often expresses the CD4+ T cell surface marker. It usually harbors the t(2;5) (p23;q35) translocation, leading to the ectopic expression of NPM-ALK, a chimeric tyrosine kinase. We demonstrated that in vitro transduction of normal human CD4+ T lymphocytes with NPM-ALK results in their immortalization and malignant transformation. The tumor cells displayed morphological and immunophenotypical characteristics of primary patient-derived anaplastic large cell lymphomas. Cell growth, proliferation, and survival were strictly dependent on NPM-ALK activity and include activation of the key factors STAT3 and DNMT1 and expression of CD30 (the hallmark of anaplastic large-cell lymphoma). Implantation of NPM-ALK-transformed CD4+ T lymphocytes into immunodeficient mice resulted in the formation of tumors indistinguishable from patients' anaplastic large cell lymphomas. Integration of "Omic" data revealed that NPM-ALK-transformed CD4+ T lymphocytes and primary NPM-ALK+ ALCL biopsies share similarities with early T cell precursors. Of note, these NPM-ALK+ lymphoma cells overexpress stem cell regulators (OCT4, SOX2, and NANOG) and HIF2A, which is known to affect hematopoietic precursor differentiation and NPM-ALK+ cell growth. Altogether, for the first time our findings suggest that NPM-ALK could restore progenitor-like features in mature CD30+ peripheral CD4+ T cells, in keeping with a thymic progenitor-like pattern.
Collapse
Affiliation(s)
- Annabelle Congras
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Equipe Labellisée LIGUE 2017, Toulouse, France
| | - Coralie Hoareau-Aveilla
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Equipe Labellisée LIGUE 2017, Toulouse, France
| | - Nina Caillet
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Equipe Labellisée LIGUE 2017, Toulouse, France
| | - Marie Tosolini
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Pôle Technologique du CRCT, Plateau Bioinformatique, Toulouse, France
| | - Patrick Villarese
- Hematology and INSERM1151, Institut Necker-Enfants Malades, University Sorbonne Paris Cité at Descartes and Assistance Publique-Hopitaux de Paris, Paris, France
| | - Agata Cieslak
- Hematology and INSERM1151, Institut Necker-Enfants Malades, University Sorbonne Paris Cité at Descartes and Assistance Publique-Hopitaux de Paris, Paris, France
| | - Laura Rodriguez
- Etablissement Français du Sang, Nouvelle Aquitaine, INSERM U1035, Université de Bordeaux, Bordeaux, France
| | - Vahid Asnafi
- Hematology and INSERM1151, Institut Necker-Enfants Malades, University Sorbonne Paris Cité at Descartes and Assistance Publique-Hopitaux de Paris, Paris, France
| | - Elisabeth Macintyre
- Hematology and INSERM1151, Institut Necker-Enfants Malades, University Sorbonne Paris Cité at Descartes and Assistance Publique-Hopitaux de Paris, Paris, France
| | - Gerda Egger
- Department of Pathology, Medical University Vienna, Vienna, Austria.,Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Pierre Brousset
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Equipe Labellisée LIGUE 2017, Toulouse, France.,Institut Carnot Lymphome, Toulouse, France.,Laboratoire d'Excellence Toulouse Cancer and after Cancer (Labex TOUCAN), Toulouse, France
| | - Laurence Lamant
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Equipe Labellisée LIGUE 2017, Toulouse, France.,Institut Carnot Lymphome, Toulouse, France.,Laboratoire d'Excellence Toulouse Cancer and after Cancer (Labex TOUCAN), Toulouse, France.,European Research Initiative on ALK-Related Malignancies, Cambridge, United Kingdom, Vienna, Austria, and Toulouse, France
| | - Fabienne Meggetto
- INSERM, UMR1037 CRCT, F-31000, Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000, Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Equipe Labellisée LIGUE 2017, Toulouse, France.,Hematology and INSERM1151, Institut Necker-Enfants Malades, University Sorbonne Paris Cité at Descartes and Assistance Publique-Hopitaux de Paris, Paris, France.,Institut Carnot Lymphome, Toulouse, France.,Laboratoire d'Excellence Toulouse Cancer and after Cancer (Labex TOUCAN), Toulouse, France.,European Research Initiative on ALK-Related Malignancies, Cambridge, United Kingdom, Vienna, Austria, and Toulouse, France
| |
Collapse
|
46
|
Redl E, Sheibani-Tezerji R, Cardona CDJ, Hamminger P, Timelthaler G, Hassler MR, Zrimšek M, Lagger S, Dillinger T, Hofbauer L, Draganić K, Tiefenbacher A, Kothmayer M, Dietz CH, Ramsahoye BH, Kenner L, Bock C, Seiser C, Ellmeier W, Schweikert G, Egger G. Requirement of DNMT1 to orchestrate epigenomic reprogramming for NPM-ALK-driven lymphomagenesis. Life Sci Alliance 2021; 4:e202000794. [PMID: 33310759 PMCID: PMC7768196 DOI: 10.26508/lsa.202000794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Malignant transformation depends on genetic and epigenetic events that result in a burst of deregulated gene expression and chromatin changes. To dissect the sequence of events in this process, we used a T-cell-specific lymphoma model based on the human oncogenic nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) translocation. We find that transformation of T cells shifts thymic cell populations to an undifferentiated immunophenotype, which occurs only after a period of latency, accompanied by induction of the MYC-NOTCH1 axis and deregulation of key epigenetic enzymes. We discover aberrant DNA methylation patterns, overlapping with regulatory regions, plus a high degree of epigenetic heterogeneity between individual tumors. In addition, ALK-positive tumors show a loss of associated methylation patterns of neighboring CpG sites. Notably, deletion of the maintenance DNA methyltransferase DNMT1 completely abrogates lymphomagenesis in this model, despite oncogenic signaling through NPM-ALK, suggesting that faithful maintenance of tumor-specific methylation through DNMT1 is essential for sustained proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Elisa Redl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | | | - Patricia Hamminger
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerald Timelthaler
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Melanie Rosalia Hassler
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Maša Zrimšek
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sabine Lagger
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Dillinger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Lorena Hofbauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Kristina Draganić
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Andreas Tiefenbacher
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Michael Kothmayer
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Charles H Dietz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bernard H Ramsahoye
- Centre for Genetic and Experimental Medicine, Institute of Genomic and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), CoreLab 2, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christian Seiser
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Schweikert
- Max Planck Institute for Intelligent Systems, Tübingen, Germany
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| |
Collapse
|
47
|
Wu Z, Nicoll M, Ingham RJ. AP-1 family transcription factors: a diverse family of proteins that regulate varied cellular activities in classical hodgkin lymphoma and ALK+ ALCL. Exp Hematol Oncol 2021; 10:4. [PMID: 33413671 PMCID: PMC7792353 DOI: 10.1186/s40164-020-00197-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/07/2023] Open
Abstract
Classical Hodgkin lymphoma (cHL) and anaplastic lymphoma kinase-positive, anaplastic large cell lymphoma (ALK+ ALCL) are B and T cell lymphomas respectively, which express the tumour necrosis factor receptor superfamily member, CD30. Another feature shared by cHL and ALK+ ALCL is the aberrant expression of multiple members of the activator protein-1 (AP-1) family of transcription factors which includes proteins of the Jun, Fos, ATF, and Maf subfamilies. In this review, we highlight the varied roles these proteins play in the pathobiology of these lymphomas including promoting proliferation, suppressing apoptosis, and evading the host immune response. In addition, we discuss factors contributing to the elevated expression of these transcription factors in cHL and ALK+ ALCL. Finally, we examine therapeutic strategies for these lymphomas that exploit AP-1 transcriptional targets or the signalling pathways they regulate.
Collapse
Affiliation(s)
- Zuoqiao Wu
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.17063.330000 0001 2157 2938Present Address: Department of Medicine, University of Toronto, Toronto, Canada
| | - Mary Nicoll
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.14709.3b0000 0004 1936 8649Present Address: Department of Biology, McGill University, Montreal, Canada
| | - Robert J. Ingham
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| |
Collapse
|
48
|
Moon DJ, Deva AK. Adverse Events Associated with Breast Implants: The Role of Bacterial Infection and Biofilm. Clin Plast Surg 2021; 48:101-108. [PMID: 33220897 DOI: 10.1016/j.cps.2020.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- David J Moon
- Department of Plastic and Reconstructive Surgery, Macquarie University, Suite 301, Level 3, 2 Technology place, New South Wales 2109, Australia
| | - Anand K Deva
- Department of Plastic and Reconstructive Surgery, Macquarie University, Suite 301, Level 3, 2 Technology place, New South Wales 2109, Australia; Integrated Specialist Healthcare Education and Research Foundation, Miranda, New South Wales, 2228, Australia.
| |
Collapse
|
49
|
Alsagaby SA, Vijayakumar R, Premanathan M, Mickymaray S, Alturaiki W, Al-Baradie RS, AlGhamdi S, Aziz MA, Alhumaydhi FA, Alzahrani FA, Alwashmi AS, Al Abdulmonem W, Alharbi NK, Pepper C. Transcriptomics-Based Characterization of the Toxicity of ZnO Nanoparticles Against Chronic Myeloid Leukemia Cells. Int J Nanomedicine 2020; 15:7901-7921. [PMID: 33116508 PMCID: PMC7568638 DOI: 10.2147/ijn.s261636] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Zinc oxide nanoparticles (ZnO NPs) have recently attracted attention as potential anti-cancer agents. To the best of our knowledge, the toxicity of ZnO NPs against human chronic myeloid leukemia cells (K562 cell line) has not been studied using transcriptomics approach. OBJECTIVE The goals of this study were to evaluate the capability of ZnO NPs to induce apoptosis in human chronic myeloid leukemia cells (K562 cells) and to investigate the putative mechanisms of action. METHODS We used viability assay and flowcytometry coupled with Annexin V-FITC and propidium iodide to investigate the toxicity of ZnO NPs on K562 cells and normal peripheral blood mononuclear cells. Next we utilized a DNA microarray-based transcriptomics approach to characterize the ZnO NPs-induced changes in the transcriptome of K562 cells. RESULTS ZnO NPs exerted a selective toxicity (mainly by apoptosis) on the leukemic cells (p≤0.005) and altered their transcriptome; 429 differentially expressed genes (DEGs) with fold change (FC)≥4 and p≤0.008 with corrected p≤0.05 were identified in K562 cells post treatment with ZnO NPs. The over-expressed genes were implicated in "response to zinc", "response to toxic substance" and "negative regulation of growth" (corrected p≤0.05). In contrast, the repressed genes positively regulated "cell proliferation", "cell migration", "cell adhesion", "receptor signaling pathway via JAK-STAT" and "phosphatidylinositol 3-kinase signaling" (corrected p≤0.05). Lowering the FC to ≥1.5 with p≤0.05 and corrected p≤0.1 showed that ZnO NPs over-expressed the anti-oxidant defense system, drove K562 cells to undergo mitochondrial-dependent apoptosis, and targeted NF-κB pathway. CONCLUSION Taken together, our findings support the earlier studies that reported anti-cancer activity of ZnO NPs and revealed possible molecular mechanisms employed by ZnO NPs to induce apoptosis in K562 cells.
Collapse
Affiliation(s)
- Suliman A Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah11932, Saudi Arabia
| | - Rajendran Vijayakumar
- Department of Biology, College of Sciences, Majmaah University, Majmaah11932, Saudi Arabia
| | - Mariappan Premanathan
- Department of Biology, College of Sciences, Majmaah University, Majmaah11932, Saudi Arabia
| | - Suresh Mickymaray
- Department of Biology, College of Sciences, Majmaah University, Majmaah11932, Saudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah11932, Saudi Arabia
| | - Raid S Al-Baradie
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah11932, Saudi Arabia
| | - Saleh AlGhamdi
- Clinical Research Department, Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh11426, Saudi Arabia
| | - Mohammad A Aziz
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh11426, Saudi Arabia
- Colorectal Cancer Research Program, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Faisal A Alzahrani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah21589, Saudi Arabia
| | - Ameen S Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Naif Khalaf Alharbi
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh11426, Saudi Arabia
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Chris Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
50
|
Nagel S, Pommerenke C, MacLeod RAF, Meyer C, Kaufmann M, Drexler HG. The NKL-code for innate lymphoid cells reveals deregulated expression of NKL homeobox genes HHEX and HLX in anaplastic large cell lymphoma (ALCL). Oncotarget 2020; 11:3208-3226. [PMID: 32922661 PMCID: PMC7456612 DOI: 10.18632/oncotarget.27683] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/07/2020] [Indexed: 12/26/2022] Open
Abstract
NKL homeobox genes encode developmental transcription factors and display an NKL-code according to their physiological expression pattern in hematopoiesis. Here, we analyzed public transcriptome data from primary innate lymphoid cells (ILCs) for NKL homeobox gene activities and found that ILC3 expressed exclusively HHEX while in ILC1 and ILC2 these genes were silenced. Deregulation of the NKL-code promotes hematopoietic malignancies, including anaplastic large cell lymphoma (ALCL) which reportedly may derive from ILC3. Accordingly, we analyzed NKL homeobox gene activities in ALCL cell lines and investigated their role in this malignancy. Transcriptome analyses demonstrated low expression levels of HHEX but powerfully activated HLX. Forced expression of HHEX in ALCL cell lines induced genes involved in apoptosis and ILC3 differentiation, indicating tumor suppressor activity. ALCL associated NPM1-ALK and JAK-STAT3-signalling drove enhanced expression of HLX while discounting HHEX. Genomic profiling revealed copy number gains at the loci of HLX and STAT3 in addition to genes encoding both STAT3 regulators (AURKA, BCL3, JAK3, KPNB1, NAMPT, NFAT5, PIM3, ROCK1, SIX1, TPX2, WWOX) and targets (BATF3, IRF4, miR135b, miR21, RORC). Transcriptome data of ALCL cell lines showed absence of STAT3 mutations while MGA was mutated and downregulated, encoding a novel potential STAT3 repressor. Furthermore, enhanced IL17F-signalling activated HLX while TGFbeta-signalling inhibited HHEX expression. Taken together, our data extend the scope of the NKL-code for ILCs and spotlight aberrant expression of NKL homeobox gene HLX in ALCL. HLX represents a direct target of ALCL hallmark factor STAT3 and deregulates cell survival and differentiation in this malignancy.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz Institute, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Claudia Pommerenke
- Department of Human and Animal Cell Lines, Leibniz Institute, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Roderick A F MacLeod
- Department of Human and Animal Cell Lines, Leibniz Institute, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz Institute, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Maren Kaufmann
- Department of Human and Animal Cell Lines, Leibniz Institute, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Hans G Drexler
- Department of Human and Animal Cell Lines, Leibniz Institute, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|