1
|
Yuan R, Zhang J, Zhou J, Cong Q. Recent progress and future challenges in structure-based protein-protein interaction prediction. Mol Ther 2025; 33:2252-2268. [PMID: 40195117 DOI: 10.1016/j.ymthe.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
Protein-protein interactions (PPIs) play a fundamental role in cellular processes, and understanding these interactions is crucial for advances in both basic biological science and biomedical applications. This review presents an overview of recent progress in computational methods for modeling protein complexes and predicting PPIs based on 3D structures, focusing on the transformative role of artificial intelligence-based approaches. We further discuss the expanding biomedical applications of PPI research, including the elucidation of disease mechanisms, drug discovery, and therapeutic design. Despite these advances, significant challenges remain in predicting host-pathogen interactions, interactions between intrinsically disordered regions, and interactions related to immune responses. These challenges are worthwhile for future explorations and represent the frontier of research in this field.
Collapse
Affiliation(s)
- Rongqing Yuan
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jing Zhang
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jian Zhou
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
2
|
Ahmed SF, Anand J, Zhang W, Buetow L, Rishi L, Mitchell L, Bohlen J, Lilla S, Sibbet GJ, Nixon C, Patel A, Majorek KA, Zanivan S, Bustamante JC, Sidhu SS, Blyth K, Huang DT. Locking CBL TKBD in its native conformation presents a novel therapeutic opportunity in mutant CBL-dependent leukemia. Mol Ther 2025:S1525-0016(25)00361-2. [PMID: 40329529 DOI: 10.1016/j.ymthe.2025.04.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/28/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Casitas B-lineage lymphoma (CBL) is an E3 ubiquitin ligase critical for negatively regulating receptor protein tyrosine kinases (RTKs). Deleterious CBL mutants lose E3 activity, but act as adaptors that gain function to cause myeloproliferative neoplasms. Currently, there is no targeted treatment available for patients with CBL mutant-dependent disorders. By combining phage-display technology and structure-based optimization, we discovered CBLock, a nanomolar affinity peptide inhibitor, that binds the substrate-binding site of CBL's tyrosine kinase binding domain (TKBD). CBLock disrupts the interaction between CBL mutants and RTKs, thereby impairing RTK-mediated priming of adaptor function of CBL mutants and downstream signaling. Notably, CBLock binds TKBD without inducing conformational changes, thereby preserving its ligand-free native conformation. In contrast, when CBL binds RTK substrates, TKBD undergoes a conformational change. Maintaining the native CBL TKBD conformation was crucial for CBLock to inhibit proliferation, induce cell-cycle arrest, and promote apoptosis in leukemia cells harboring CBL mutations. In a mouse xenograft model of acute myeloid leukemia (AML), CBLock reduced tumor burden and improved survival rate. Moreover, CBLock inhibited the proliferation of cells derived from patients with CBL mutations. Therefore, inhibiting CBL TKBD in its native state presents a promising therapeutic opportunity in targeting mutant CBL-dependent leukemia.
Collapse
Affiliation(s)
- Syed Feroj Ahmed
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jayanthi Anand
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Wei Zhang
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, 50 Stone Road E, Guelph, ON N1G2W1, Canada
| | - Lori Buetow
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Loveena Rishi
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Louise Mitchell
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Sergio Lilla
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Gary J Sibbet
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Colin Nixon
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Amrita Patel
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Karolina A Majorek
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Sara Zanivan
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Jacinta C Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, 75015 Paris, France; Paris Cité University, Imagine Institute, 75015 Paris, France
| | - Sachdev S Sidhu
- School of Pharmacy, University of Waterloo, 10 Victoria Street S A, Kitchener, ON N2G1C5, Canada
| | - Karen Blyth
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Danny T Huang
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
3
|
Costa A, Breccia M. SOHO State of the Art Updates and Next Questions. Atypical Chronic Myeloid Leukemia: Pathogenesis, Diagnostic Challenges, and Therapeutic Strategies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00110-7. [PMID: 40288921 DOI: 10.1016/j.clml.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Atypical chronic myeloid leukemia (aCML) is a rare and challenging clonal hematopoietic disorder within the myelodysplastic/myeloproliferative neoplasm (MDS/MPN) spectrum. Over the past two decades, substantial progress has been made in understanding the genetic mechanisms driving aCML, revealing a complex and heterogeneous mutational landscape. Key ancestral mutations, such as ASXL1 and ETNK1, have been identified, providing a foundation for the pathogenesis and for the possible emergence of secondary abnormalities, particularly in epigenetic regulation (eg, SETBP1), and in splicing process (eg, SRSF2). These molecular insights have been integrated into current diagnostic classifications, refining disease characterization and offering potential targets for precision therapies. Despite these advances, significant clinical challenges persist due to the disease's rarity and the lack of randomized clinical trials. Therapeutic strategies remain inadequately defined, with allogeneic stem cell transplantation being the only curative option. This review provides an overview of the molecular, clinical, and therapeutic information that may pave the way for essential advancements in the proper management of this disease.
Collapse
Affiliation(s)
- Alessandro Costa
- Hematology Unit, Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy.
| |
Collapse
|
4
|
Zhang N, Liang Y, Meng YQ, Li YC, Lu X, Li L, Ye T. Analysis and identification of potential biomarkers for dysfunctional uterine bleeding. J Reprod Immunol 2025; 168:104427. [PMID: 39862473 DOI: 10.1016/j.jri.2025.104427] [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: 11/13/2023] [Revised: 12/04/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025]
Abstract
Clinical evidence increasingly suggests that traditional treatments for dysfunctional uterine bleeding (DUB) have limited success. In this study, blood samples from 10 DUB patients and 10 healthy controls were collected for transcriptome sequencing. Then, the differentially expressed genes (DEGs) were screened and crossed with the DUB-related module genes to obtain the target genes. These target genes were analyzed for functional enrichment. Further, the biomarkers were screened by protein-protein interaction (PPI) analysis and analyzed by the gene set enrichment analysis (GSEA) and ingenuity pathway analysis (IPA). To explore the pathogenesis of DUB, immune microenvironment analyses were also performed. Potential drugs targeting these biomarkers were predicted for clinical treatment. The expression of these biomarkers was validated using quantitative real-time polymerase chain reaction (qRT-PCR). The results showed that, a total of 754 target genes were found to be related to cell proliferation and senescence. Five biomarkers-CENPE, KIF11, PIK3R1, SMC3, and SMC4-were identified, all of which were down-regulated in the DUB group, and most of these findings were confirmed by qRT-PCR. Notably, CENPE expression showed a negative association with activated NK cells and a positive association with resting NK cells. In addition, 44 potential drugs were predicted for DUB treatment. In conclusion, five DUB biomarkers were identified, enhancing understanding of gene regulation in DUB and providing a theoretical foundation for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- N Zhang
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 50001, China
| | - Y Liang
- Department of Gynaecology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Y Q Meng
- Department of Gynaecology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Y C Li
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 50001, China
| | - X Lu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - L Li
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 50001, China.
| | - T Ye
- Department of Chinese Medicine Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 50001, China.
| |
Collapse
|
5
|
Tian P, Zheng J, Qiao K, Fan Y, Xu Y, Wu T, Chen S, Zhang Y, Zhang B, Ambrogio C, Wang H. scPharm: Identifying Pharmacological Subpopulations of Single Cells for Precision Medicine in Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412419. [PMID: 39560158 PMCID: PMC11727242 DOI: 10.1002/advs.202412419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/06/2024] [Indexed: 11/20/2024]
Abstract
Intratumour heterogeneity significantly hinders the efficacy of anticancer therapies. Compared with drug perturbation experiments, which yield pharmacological data at the bulk cell level, single-cell RNA sequencing (scRNA-seq) technology provides a means to capture molecular heterogeneity at single-cell resolution. Here, scPharm is introduced, a computational framework that integrates pharmacological profiles with scRNA-seq data to identify pharmacological subpopulations of cells within a tumour and prioritize tailored drugs. scPharm uses the normalized enrichment scores (NESs) determined from gene set enrichment analysis to assess the distribution of cell identity genes in drug response-determined gene lists. Based on the strong correlation between the NES and drug response at single-cell resolution, scPharm successfully identified the sensitive subpopulations in ER-positive and HER2-positive human breast cancer tissues, revealed dynamic changes in the resistant subpopulation of human PC9 cells treated with erlotinib, and expanded its ability to a mouse model. Its superior performance and computational efficiency are confirmed through comparative evaluations with other single-cell prediction tools. Additionally, scPharm predicted combination drug strategies by gauging compensation or booster effects between drugs and evaluated drug toxicity in healthy cells in the tumour microenvironment. Overall, scPharm provides a novel approach for precision medicine in cancers by revealing therapeutic heterogeneity at single-cell resolution.
Collapse
Affiliation(s)
- Peng Tian
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Jie Zheng
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Keying Qiao
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yuxiao Fan
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yue Xu
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Tao Wu
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Shuting Chen
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yinuo Zhang
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Bingyue Zhang
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health SciencesMolecular Biotechnology CenterUniversity of TorinoTorino10126Italy
| | - Haiyun Wang
- Research Center for Translational MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| |
Collapse
|
6
|
Bohlen J, Bagarić I, Vatovec T, Ogishi M, Ahmed SF, Cederholm A, Buetow L, Sobrino S, Le Floc’h C, Arango-Franco CA, Seabra L, Michelet M, Barzaghi F, Leardini D, Saettini F, Vendemini F, Baccelli F, Catala A, Gambineri E, Veltroni M, Aguilar de la Red Y, Rice GI, Consonni F, Berteloot L, Largeaud L, Conti F, Roullion C, Masson C, Bessot B, Seeleuthner Y, Le Voyer T, Rinchai D, Rosain J, Neehus AL, Erazo-Borrás L, Li H, Janda Z, Cho EJ, Muratore E, Soudée C, Lainé C, Delabesse E, Goulvestre C, Ma CS, Puel A, Tangye SG, André I, Bole-Feysot C, Abel L, Erlacher M, Zhang SY, Béziat V, Lagresle-Peyrou C, Six E, Pasquet M, Alsina L, Aiuti A, Zhang P, Crow YJ, Landegren N, Masetti R, Huang DT, Casanova JL, Bustamante J. Autoinflammation in patients with leukocytic CBL loss of heterozygosity is caused by constitutive ERK-mediated monocyte activation. J Clin Invest 2024; 134:e181604. [PMID: 39403923 PMCID: PMC11475086 DOI: 10.1172/jci181604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Patients heterozygous for germline CBL loss-of-function (LOF) variants can develop myeloid malignancy, autoinflammation, or both, if some or all of their leukocytes become homozygous for these variants through somatic loss of heterozygosity (LOH) via uniparental isodisomy. We observed an upregulation of the inflammatory gene expression signature in whole blood from these patients, mimicking monogenic inborn errors underlying autoinflammation. Remarkably, these patients had constitutively activated monocytes that secreted 10 to 100 times more inflammatory cytokines than those of healthy individuals and CBL LOF heterozygotes without LOH. CBL-LOH hematopoietic stem and progenitor cells (HSPCs) outgrew the other cells, accounting for the persistence of peripheral monocytes homozygous for the CBL LOF variant. ERK pathway activation was required for the excessive production of cytokines by both resting and stimulated CBL-LOF monocytes, as shown in monocytic cell lines. Finally, we found that about 1 in 10,000 individuals in the UK Biobank were heterozygous for CBL LOF variants and that these carriers were at high risk of hematological and inflammatory conditions.
Collapse
Affiliation(s)
- Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Ivan Bagarić
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Taja Vatovec
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Syed F. Ahmed
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lori Buetow
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Steicy Sobrino
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Laboratory of Chromatin and Gene Regulation during Development, Paris Cité University, INSERM U1163, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM U1163, Imagine Institute, Paris, France
| | - Corentin Le Floc’h
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Carlos A. Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Luis Seabra
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Marine Michelet
- Unit of Allergy and Pneumology, Children’s Hospital, Toulouse, France
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Leardini
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Francesco Saettini
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Francesco Baccelli
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Albert Catala
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Eleonora Gambineri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Marinella Veltroni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | | | - Gillian I. Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Filippo Consonni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
- “Mario Serio” Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Laureline Berteloot
- Department of Pediatric Imaging, Necker Hospital for Sick Children, Paris, France
- INSERM U1163, Paris, France
| | - Laetitia Largeaud
- Laboratory of Hematology, Hospital Center of the University of Toulouse, Toulouse, France
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Cécile Roullion
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Genomics Core Facility and
| | - Cécile Masson
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Bioinformatic Plateform, INSERM U1163 and INSERM US24/CNRS UAR3633, Paris Cité University, Paris, France
| | - Boris Bessot
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Saint-Louis Hospital, Paris, France
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children–AP-HP, Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Lucia Erazo-Borrás
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Hailun Li
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Zarah Janda
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - En-Jui Cho
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Edoardo Muratore
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Candice Lainé
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Eric Delabesse
- Department of Hematology, CHU and Centre de Recherche de Cancérologie de Toulouse, Paul-Sabatier University, Toulouse, France
| | | | - Cindy S. Ma
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, Australia
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, Australia
| | - Isabelle André
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Christine Bole-Feysot
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Genomics Core Facility and
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Chantal Lagresle-Peyrou
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Emmanuelle Six
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM U1163, Imagine Institute, Paris, France
| | - Marlène Pasquet
- Department of Pediatric Hematology and Oncology, Centre Hospitalo–Universitaire de Toulouse, Toulouse, France
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Yanick J. Crow
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Riccardo Masetti
- Unit of Allergy and Pneumology, Children’s Hospital, Toulouse, France
| | - Danny T. Huang
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Necker Hospital for Sick Children–AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children–AP-HP, Paris, France
| |
Collapse
|
7
|
Zabel KM, Rebbe R, Vasef M, Foucar C. Case of B-acute lymphoblastic leukaemia with t(1;19)(q23;p13.3) TCF3::PBX1 and co-occurring CBL mutation in an elderly patient. BMJ Case Rep 2024; 17:e260617. [PMID: 39266021 DOI: 10.1136/bcr-2024-260617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Abstract
The t(1;19) (q23;p13) TCF3::PBX1 is a well-described, recurring chromosomal abnormality in B-acute lymphoblastic leukaemia (B-ALL) that has historically been associated with a worse prognosis in paediatric patients. Gene expression profiling has demonstrated that TCF3::PBX1 results in a distinct subtype of B-ALL, leading to its recognition in the most recent WHO and ICC classifications. Though initially believed to be a poor prognostic sign in the adult population, emerging evidence suggests its presence may instead be intermediate or even favourable in B-ALL. However, adults with TCF3::PBX1 are typically younger and often qualify for treatment with paediatric-inspired regimens. Thus, the prognostic significance in this population remains unclear. This translocation appears to be very rare in older adults with B-ALL and its predictive and prognostic nature in this population is unknown. Herein, we explore a case of this translocation occurring in a patient in her 70s. She initially presented to the emergency department with abdominal pain and thrombocytopenia and was subsequently diagnosed with B-ALL. In addition to t(1;19) (q23;p13), a pathologic mutation in the CBL gene was identified. CBL mutations have been implicated in cancer progression and are mostly described in paediatric B-ALL. She was treated with modified Ph-negative EWALL induction (Vincristine, Idarubicin, dexamethasone) and achieved a complete remission. However, she subsequently experienced an early relapse and was refractory to targeted therapy with blinatumomab. After treatment with inotuzumab ozogamicin, she achieved a second complete remission. Unfortunately, she then suffered a central nervous system (CNS) relapse and passed away from complications of her disease. This case serves as an example of the heterogeneous nature of B-ALL. It demonstrates that patients with ostensibly favourable prognostic factors may experience poor response rates to traditional chemotherapy as well as targeted salvage agents. It also illustrates the challenges of treating B-ALL in the elderly population.
Collapse
Affiliation(s)
- Kenneth Matthew Zabel
- Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Ryan Rebbe
- Molecular/Genetic Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Mohammad Vasef
- University of New Mexico Health Sciences, Albuquerque, New Mexico, USA
| | - Charles Foucar
- Department of Hematology and Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
8
|
Li X, Li W, Zhang Y, Xu L, Song Y. Exploiting the potential of the ubiquitin-proteasome system in overcoming tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Genes Dis 2024; 11:101150. [PMID: 38947742 PMCID: PMC11214299 DOI: 10.1016/j.gendis.2023.101150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/15/2023] [Accepted: 09/01/2023] [Indexed: 07/02/2024] Open
Abstract
The advent of tyrosine kinase inhibitors (TKI) targeting BCR-ABL has drastically changed the treatment approach of chronic myeloid leukemia (CML), greatly prolonged the life of CML patients, and improved their prognosis. However, TKI resistance is still a major problem with CML patients, reducing the efficacy of treatment and their quality of life. TKI resistance is mainly divided into BCR-ABL-dependent and BCR-ABL-independent resistance. Now, the main clinical strategy addressing TKI resistance is to switch to newly developed TKIs. However, data have shown that these new drugs may cause serious adverse reactions and intolerance and cannot address all resistance mutations. Therefore, finding new therapeutic targets to overcome TKI resistance is crucial and the ubiquitin-proteasome system (UPS) has emerged as a focus. The UPS mediates the degradation of most proteins in organisms and controls a wide range of physiological processes. In recent years, the study of UPS in hematological malignant tumors has resulted in effective treatments, such as bortezomib in the treatment of multiple myeloma and mantle cell lymphoma. In CML, the components of UPS cooperate or antagonize the efficacy of TKI by directly or indirectly affecting the ubiquitination of BCR-ABL, interfering with CML-related signaling pathways, and negatively or positively affecting leukemia stem cells. Some of these molecules may help overcome TKI resistance and treat CML. In this review, the mechanism of TKI resistance is briefly described, the components of UPS are introduced, existing studies on UPS participating in TKI resistance are listed, and UPS as the therapeutic target and strategies are discussed.
Collapse
Affiliation(s)
- Xudong Li
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yanli Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Linping Xu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
9
|
Stellacci E, Carter JN, Pannone L, Stevenson D, Moslehi D, Venanzi S, Bernstein JA, Tartaglia M, Martinelli S. Immunological and hematological findings as major features in a patient with a new germline pathogenic CBL variant. Am J Med Genet A 2024; 194:e63627. [PMID: 38613168 PMCID: PMC11223960 DOI: 10.1002/ajmg.a.63627] [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: 02/01/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024]
Abstract
Casitas B-lineage lymphoma (CBL) encodes an adaptor protein with E3-ligase activity negatively controlling intracellular signaling downstream of receptor tyrosine kinases. Somatic CBL mutations play a driver role in a variety of cancers, particularly myeloid malignancies, whereas germline defects in the same gene underlie a RASopathy having clinical overlap with Noonan syndrome (NS) and predisposing to juvenile myelomonocytic leukemia and vasculitis. Other features of the disorder include cardiac defects, postnatal growth delay, cryptorchidism, facial dysmorphisms, and predisposition to develop autoimmune disorders. Here we report a novel CBL variant (c.1202G>T; p.Cys401Phe) occurring de novo in a subject with café-au-lait macules, feeding difficulties, mild dysmorphic features, psychomotor delay, autism spectrum disorder, thrombocytopenia, hepatosplenomegaly, and recurrent hypertransaminasemia. The identified variant affects an evolutionarily conserved residue located in the RING finger domain, a known mutational hot spot of both germline and somatic mutations. Functional studies documented enhanced EGF-induced ERK phosphorylation in transiently transfected COS1 cells. The present findings further support the association of pathogenic CBL variants with immunological and hematological manifestations in the context of a presentation with only minor findings reminiscent of NS or a clinically related RASopathy.
Collapse
Affiliation(s)
- Emilia Stellacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- These authors equally contributed to this work
| | - Jennefer N. Carter
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics - Medical Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- These authors equally contributed to this work
| | - Luca Pannone
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - David Stevenson
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics - Medical Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dorsa Moslehi
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
| | - Serenella Venanzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Jonathan A. Bernstein
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics - Medical Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
- These authors equally contributed to this work
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- These authors equally contributed to this work
| |
Collapse
|
10
|
Xu X, Zhang Y, Lu Y, Zhang X, Zhao C, Wang J, Guan Q, Feng Y, Gao M, Yu J, Song Z, Liu X, Golchehre Z, Li L, Ren W, Pan-Hammarström Q, Zhang H, Wang X. CD58 Alterations Govern Antitumor Immune Responses by Inducing PDL1 and IDO in Diffuse Large B-Cell Lymphoma. Cancer Res 2024; 84:2123-2140. [PMID: 38635903 DOI: 10.1158/0008-5472.can-23-2874] [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: 09/20/2023] [Revised: 01/27/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
Recurrent abnormalities in immune surveillance-related genes affect the progression of diffuse large B-cell lymphoma (DLBCL) and modulate the response to therapeutic interventions. CD58 interacts with the CD2 receptor on T cells and NK cells and is recurrently mutated and deleted in DLBCL, suggesting that it may play a role in regulating antitumor immunity. In this study, we comprehensively analyzed the genomic characteristics of CD58 through targeted next-generation sequencing, RNA sequencing (RNA-seq), whole-exome sequencing, and single-cell RNA-seq in patients with newly diagnosed DLBCL. The CD58 mutation rate was 9.1%, and the copy number loss rate was 44.7% among all enrolled patients with DLBCL. Notably, CD58 genetic alterations, along with low CD58 expression, significantly correlated with reduced rates of response to R-CHOP therapy and inferior progression-free survival and overall survival. Single-cell RNA-seq revealed that CD58 expression in tumor cells was negatively correlated with CD8+ T-cell exhaustion/dysfunction status. Insufficient T-cell activation resulting from CD58 alterations could not be attributed solely to CD2 signaling. CD58 inhibited the activity of the JAK2/STAT1 pathway by activating the LYN/CD22/SH2 domain-containing phosphatase 1 (SHP1) axis, thereby limiting PDL1 and IDO expression. Elevated PDL1 and IDO expression in CD58-deficient DLBCL cells led to immune evasion and tumor-intrinsic resistance to chimeric antigen receptor T-cell therapy. Direct activation of CD58-CD2 costimulatory signaling in combination with anti-PDL1 blockade or IDO inhibitor sensitized CD58-deficient DLBCL to chimeric antigen receptor T-cell therapy. Collectively, this work identified the multiple roles of CD58 in regulating antitumor immune responses in DLBCL. Significance: Loss of CD58 mediates immune evasion and therapy resistance in diffuse large B-cell lymphoma by upregulating PDL1 and IDO through LYN/CD22/SHP1 signaling, providing potential targets and therapeutic strategies to improve patient treatment.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- CD58 Antigens/genetics
- CD58 Antigens/metabolism
- Female
- Male
- Mutation
- Animals
- Middle Aged
- Mice
- Cell Line, Tumor
- Aged
Collapse
Affiliation(s)
- Xiyue Xu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yidan Zhang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yaxiao Lu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiaoyan Zhang
- State Key Laboratory of Experimental Hematology and Division of Pediatric Blood Diseases Center, Institute of Hematology and Blood Diseases Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Tianjin, China
| | - Cuicui Zhao
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of VIP Ward, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jiesong Wang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Lymphoma and Head and Neck Oncology, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, China
| | - Qingpei Guan
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Critical Care Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Yingfang Feng
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Meng Gao
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jingwei Yu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zheng Song
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xia Liu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zahra Golchehre
- Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Lanfang Li
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Weicheng Ren
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | | | - Huilai Zhang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xianhuo Wang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
11
|
Liu L, Zhu Y, Lan J, Chu L, Li W, Xue C. Association between CBL gene polymorphism and susceptibility of microscopic polyangiitis in a Chinese population: A case-control analysis. Cytokine 2024; 179:156596. [PMID: 38669907 DOI: 10.1016/j.cyto.2024.156596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
OBJECTIVE To assess whether Casitas B-lineage lymphoma (CBL) gene polymorphism influences the risk of microscopic polyangiitis (MPA) in Chinese populations. METHODS In total, 266 MPA patients and 297 healthy controls were recruited for a case-control study. Five CBL SNPs were genotyped using multiplex polymerase chain reaction and high-throughput sequencing. The relationship between SNPs and the risk of MPA under different genetic models was evaluated by SNPstats. SNP-SNP interaction was analyzed by generalized multifactor dimensionality reduction (GMDR). Finally, the association between CBL SNPs and treatment effects were assessed. RESULTS The results showed that CBL rs2276083 was associated with decreasing MPA risk under dominant (OR: 0.53; p = 0.014) and recessive models (OR: 0.52; p = 0.0034). Stratification analysis indicated that rs2276083 and rs2509671 in age < 60 years, rs2276083 in female or in Han population were protective factors for MPA. The CBL haplotype (A-A-G-C-T) was associated with an increased risk of MPA. GMDR suggested that CBL rs2276083, phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PI3KCA) rs1607237, and autophagy-related gene 7 (ATG7) rs7549008 might interact with each other in MPA development (p = 0.0107). CBL rs1047417 with AG genotype and rs11217234 with AG genotype had better clinical treatment effects than other two genotypes (p = 0.048 and p = 0.025, respectively). CONCLUSION The genetic polymorphism of CBL had a potential association with the risk of MPA and clinical treatment effects in Guangxi population in China.
Collapse
Affiliation(s)
- Liu Liu
- The Second Affiliated Hospital of Guangxi Medical University, Department of Nephrology, Nanning Guangxi, 530007, China; The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang Hunan, 421001, China
| | - Yan Zhu
- The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang Hunan, 421001, China
| | - Jingjing Lan
- The Second Affiliated Hospital of Guangxi Medical University, Department of Nephrology, Nanning Guangxi, 530007, China
| | - Liepeng Chu
- The Second Affiliated Hospital of Guangxi Medical University, Department of Nephrology, Nanning Guangxi, 530007, China
| | - Wei Li
- The Second Affiliated Hospital of Guangxi Medical University, Department of Nephrology, Nanning Guangxi, 530007, China
| | - Chao Xue
- The Second Affiliated Hospital of Guangxi Medical University, Department of Nephrology, Nanning Guangxi, 530007, China.
| |
Collapse
|
12
|
Guo H, Zhou C, Zheng M, Zhang J, Wu H, He Q, Ding L, Yang B. Insights into the role of derailed endocytic trafficking pathway in cancer: From the perspective of cancer hallmarks. Pharmacol Res 2024; 201:107084. [PMID: 38295915 DOI: 10.1016/j.phrs.2024.107084] [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: 10/19/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
The endocytic trafficking pathway is a highly organized cellular program responsible for the regulation of membrane components and uptake of extracellular substances. Molecules internalized into the cell through endocytosis will be sorted for degradation or recycled back to membrane, which is determined by a series of sorting events. Many receptors, enzymes, and transporters on the membrane are strictly regulated by endocytic trafficking process, and thus the endocytic pathway has a profound effect on cellular homeostasis. However, the endocytic trafficking process is typically dysregulated in cancers, which leads to the aberrant retention of receptor tyrosine kinases and immunosuppressive molecules on cell membrane, the loss of adhesion protein, as well as excessive uptake of nutrients. Therefore, hijacking endocytic trafficking pathway is an important approach for tumor cells to obtain advantages of proliferation and invasion, and to evade immune attack. Here, we summarize how dysregulated endocytic trafficking process triggers tumorigenesis and progression from the perspective of several typical cancer hallmarks. The impact of endocytic trafficking pathway to cancer therapy efficacy is also discussed.
Collapse
Affiliation(s)
- Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China; Cancer Center of Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Medicine, Hangzhou City University, Hangzhou 310015, China; The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China.
| |
Collapse
|
13
|
Feng X, Yang L, Liu X, Liu M, Liu L, Liu J, Luo J. Long non-coding RNA small nucleolar RNA host gene 29 drives chronic myeloid leukemia progression via microRNA-483-3p/Casitas B-lineage Lymphoma axis-mediated activation of the phosphoinositide 3-kinase/Akt pathway. Med Oncol 2024; 41:60. [PMID: 38252204 DOI: 10.1007/s12032-023-02287-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024]
Abstract
The aberrant expression of the long non-coding RNA (lncRNA) Small Nucleolar RNA Host Gene 29 (SNHG29) has been associated with various human cancers. However, the role of SNHG29 in chronic myeloid leukemia (CML) remains elusive. Therefore, this study aimed to investigate the function of SNHG29 in CML and unveil its potential underlying mechanisms. Herein, peripheral blood samples from 44 CML patients and 17 healthy subjects were collected. The expressions of SNHG29, microRNA-483-3p (miR-483-3p), and Casitas B-lineage Lymphoma (CBL) were measured using quantitative polymerase chain reaction (qPCR) or Western Blot. Cell viability, apoptosis, and cell cycle progression were evaluated using the Cell Counting Kit-8 assay, 5-ethynyl-2'-deoxyuridine incorporation, and flow cytometry, respectively. Western Blot analysis was employed to assess protein expressions related to cellular proliferation, apoptosis, and oncogenesis. RNA immunoprecipitation and dual-luciferase reporter assays were utilized to verify the interactions among SNHG29, miR-483-3p, and CBL. SNHG29 was significantly overexpressed in both blood samples of CML patients and CML cell lines. In CML, increased expression of SNHG29 was positively correlated with clinical staging, and patients with high SNHG29 expression had poorer survival outcomes. Functionally, knocking down SNHG29 effectively inhibited CML cell proliferation and promoted apoptosis. Mechanistically, SNHG29 acted as a competing endogenous RNA for miR-483-3p to modulate CBL expression, thereby activating the Phosphoinositide 3-Kinase/Akt signaling pathway and mediating CML progression. In summary, these findings reveal that SNHG29 promotes tumorigenesis in CML, offering a potential therapeutic strategy for CML treatment.
Collapse
Affiliation(s)
- XueFeng Feng
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China
| | - Lin Yang
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China
| | - Xiaojun Liu
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China
| | - Menghan Liu
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China
| | - Lu Liu
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China
| | - Jing Liu
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China
| | - JianMin Luo
- Department of Second Ward of Hematology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang City, 050000, Hebei, China.
| |
Collapse
|
14
|
Yan JH, Liao KQ, Yao L, Chen JL, Xiong LF, Tao XZ. LncRNA AL645608.3 mediates malignant progression of acute myeloid leukemia. Am J Transl Res 2024; 16:342-355. [PMID: 38322561 PMCID: PMC10839383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024]
Abstract
OBJECTIVE To investigate the role of lncRNA AL645608.3 in the malignant progression of acute myeloid leukemia (AML) cells and explore relevant molecular mechanisms. METHODS The expression level of AL645608.3 was measured in AML cell lines (THP-1, HL-60, KG-1, and AML-193) via real-time quantitative polymerase chain reaction (RT-qPCR). Small hairpin RNA (shRNA) and open reading frame of AL645608.3 were cloned into lentiviral vectors and were infected into THP-1 and AML-193 cells. The expression of casitas B-lineage lymphoma (CBL), interferon regulatory factor 6 (IRF6), and interferon beta 1 (IFNB1) was detected through RT-qPCR, and western blot. Co-immunoprecipitation (Co-IP) on IRF6 was conducted. Matrix metalloprotease-9 (MMP-9) activity was evaluated via gelatin zymography assay. RESULTS LncRNA AL645608.3 was expressed in the four AML cell lines (THP-1, HL-60, KG-1, and AML-193). Silencing AL645608.3 mitigated the expression of IRF6 and IFNB1 but elevated the expression of CBL in THP-1 cells. Oppositely, AL645608.3 overexpression up-regulated the expression of IRF6 and IFNB1 but decreased the expression of CBL in AML-193 cells. Co-IP results proved that AL645608.3 could directly mediate IRF6 activity in THP-1 and AML-193 cells. MMP-9 activity was decreased by AL645608.3 knockdown and was improved by AL645608.3 overexpression in AML-193 cells. CONCLUSION AL645608.3 is expressed in different AML cell lines, and mediates the expression of CBL, IRF6, IFNB1, and MMP-9. These findings might deepen our comprehension of the molecular mechanisms underlying AML.
Collapse
Affiliation(s)
- Jin-Hua Yan
- School of Information Management, Jiangxi University of Finance and EconomicsNanchang 330013, Jiangxi, China
| | - Kai-Qiong Liao
- Department of Hematology, The First Hospital of NanchangNanchang 330008, Jiangxi, China
| | - Ling Yao
- Department of Gastroenterology, The First Hospital of NanchangNanchang 330008, Jiangxi, China
| | - Jian-Lan Chen
- Department of Hematology, The First Hospital of NanchangNanchang 330008, Jiangxi, China
| | - Li-Fang Xiong
- Department of Hematology, The First Hospital of NanchangNanchang 330008, Jiangxi, China
| | - Xu-Zhang Tao
- Department of Nuclear Medicine, The First Hospital of NanchangNanchang 330008, Jiangxi, China
| |
Collapse
|
15
|
Liongue C, Ward AC. Myeloproliferative Neoplasms: Diseases Mediated by Chronic Activation of Signal Transducer and Activator of Transcription (STAT) Proteins. Cancers (Basel) 2024; 16:313. [PMID: 38254802 PMCID: PMC10813624 DOI: 10.3390/cancers16020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are hematopoietic diseases characterized by the clonal expansion of single or multiple lineages of differentiated myeloid cells that accumulate in the blood and bone marrow. MPNs are grouped into distinct categories based on key clinical presentations and distinctive mutational hallmarks. These include chronic myeloid leukemia (CML), which is strongly associated with the signature BCR::ABL1 gene translocation, polycythemia vera (PV), essential thrombocythemia (ET), and primary (idiopathic) myelofibrosis (PMF), typically accompanied by molecular alterations in the JAK2, MPL, or CALR genes. There are also rarer forms such as chronic neutrophilic leukemia (CNL), which involves mutations in the CSF3R gene. However, rather than focusing on the differences between these alternate disease categories, this review aims to present a unifying molecular etiology in which these overlapping diseases are best understood as disruptions of normal hematopoietic signaling: specifically, the chronic activation of signaling pathways, particularly involving signal transducer and activator of transcription (STAT) transcription factors, most notably STAT5B, leading to the sustained stimulation of myelopoiesis, which underpins the various disease sequalae.
Collapse
Affiliation(s)
- Clifford Liongue
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia;
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia;
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
16
|
Liang H, Yin G, Shi G, Liu Z, Liu X, Li J. Echinacoside regulates PI3K/AKT/HIF-1α/VEGF cross signaling axis in proliferation and apoptosis of breast cancer. Anal Biochem 2024; 684:115360. [PMID: 37865269 DOI: 10.1016/j.ab.2023.115360] [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: 06/29/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
CONTEXT Echinacoside (ECH) is a natural anti-cancer compound and is of great value in cancer treatment. However, the mechanism underlying this effect on breast cancer (BC) was unclear. OBJECTIVE To explore the mechanism of ECH treating BC by network pharmacology and experimental validation. MATERIALS & METHODS Several databases were searched to screen potential targets of ECH and obtain information on targets related to BC. STRING was applied to construct a Protein-protein interaction (PPI) network. DAVID was applied for Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Gene Expression Profiling Interactive Analysis (GEPIA) was searched for the relationship between the expression profile and overall survival of major targets in normal breast and BC tissues. Finally, the results of network pharmacology analysis were validated by experiments. RESULTS Seventeen targets of ECH overlapped with targets in BC. Ten hub targets were determined through PPI. By GO and KEGG analysis 15 entries and 25 pathways were obtained, in which phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), hypoxia inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) played greater roles. Validation of key targets in the GEPIA database showed that PIK3R1 and PIK3CD remained consistent with the results of the study. Experiments in vitro showed ECH inhibited proliferation, induced apoptosis and reduced mRNA levels and protein expression of PI3K, AKT, hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA) in MCF-7 cells. Furthermore, experiments in vivo revealed that ECH significantly reduced tumor growth, promoted apoptosis and decreased the related mRNA levels and protein expression, suggesting ECH works on BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. DISCUSSION & CONCLUSION In summary, ECH played an important role in anti-BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. Furthermore, ECH had multi-target and multi-pathway effects, which may be a promising natural compound for treating BC.
Collapse
Affiliation(s)
- Hongyi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guangxi Shi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Zhiyong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Xiaofei Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Jingwei Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
17
|
Wang HY, Liu Y, Deng L, Jiang K, Yang XH, Wu XY, Guo KH, Wang F. Clinical significance of genetic profiling based on different anatomic sites in patients with mucosal melanoma who received or did not receive immune checkpoint inhibitors. Cancer Cell Int 2023; 23:187. [PMID: 37649078 PMCID: PMC10469937 DOI: 10.1186/s12935-023-03032-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND To date, data on the efficacy of targeted therapies for mucosal melanoma (MM) are limited. In this study, we analyzed genetic alterations according to the primary site of origin, which could provide clues for targeted therapy for MM. METHODS We conducted a retrospective cohort study of 112 patients with MM. Targeted sequencing was performed to analyze genetic aberrations. Kaplan-Meier analysis was conducted with the log-rank test to compare the significance among subgroups. RESULTS In total, 112 patients with MM were included according to the anatomic sites: 38 (33.9%) in the head and neck, 22 (19.6%) in the genitourinary tract, 21 (18.8%) in the anorectum, 19 (17.0%) in the esophagus, 10 (8.9%) in the uvea, and 2 (1.8%) in the small bowel. The most significantly mutated genes included BRAF (17%), KIT (15%), RAS (15%), TP53 (13%), NF1 (12%), SF3B1 (11%), GNA11 (7%), GNAQ (5%), and FBXW7 (4%). A large number of chromosomal structural variants was found. The anatomic sites of esophagus and small bowel were independent risk factors for progression-free survival (PFS, hazard ratio [HR] 4.78, 95% confidence interval [CI] 2.42-9.45, P < 0.0001) and overall survival (OS, HR 5.26, 95% CI 2.51-11.03, P < 0.0001). Casitas B-lineage lymphoma (CBL) mutants showed significantly poorer PFS and OS. In contrast, MM patients who received immune checkpoint inhibitors (ICIs) had a significantly more favorable OS (HR 0.39, 95% CI 0.20-0.75, P = 0.008). CONCLUSIONS Our findings reveal the genetic features of patients with MM, mainly across six anatomic sites, offering a potential avenue for targeted therapies.
Collapse
Affiliation(s)
- Hai-Yun Wang
- Department of Pathology, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, National Children's Medical Center for South Central Region, 510623, Guangzhou, P. R. China
- Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, National Children's Medical Center for South Central Region, 510623, Guangzhou, P. R. China
| | - Ye Liu
- Shenzhen Hospital, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 518116, Shenzhen, China
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, P. R. China
| | - Ling Deng
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, P. R. China
| | - Kuntai Jiang
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, P. R. China
| | - Xin-Hua Yang
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, P. R. China
| | - Xiao-Yan Wu
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, P. R. China
| | - Kai-Hua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road, 510080, Guangzhou, P. R. China.
| | - Fang Wang
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, P. R. China.
| |
Collapse
|
18
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
19
|
宋 筱, 肖 斌, 陆 景, 张 文, 李 锦, 竹 昕, 孙 朝, 李 林. [CBL inhibits proliferation and invasion of breast cancer cells by ubiquitylation-mediated degradation of NCK2]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1594-1603. [PMID: 36504051 PMCID: PMC9742774 DOI: 10.12122/j.issn.1673-4254.2022.11.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To observe the effects of Casitas B lymphoma (CBL) protein on proliferation, migration and invasion of breast cancer cells and explore its mechanism of action. METHODS Cultured breast cancer cell lines MDA-MB-231 and MCF7A were transfected with a CBL-overexpressing plasmid and a specific siRNA targeting CBL (siRNA-CBL), respectively, and the changes in cell proliferation, migration and invasion were examined using colony-forming assay, cell counting kit-8 (CCK-8), scratch test and Transwell assay. Flow cytometry and Western blotting were performed to examine the effects of CBL overexpression on cell cycle and epithelial-mesenchymal transition (EMT) of MDA-MB-231 cells, and the changes in the number of filamentous pseudopodia were observed by rhodamine- labeled phalloidin staining of the cytoskeleton. IP-mass spectrometry identified NCK2 as the interacting proteins of CBL, and their interaction was verified by immunoprecipitation and immunofluorescence co-localization experiments in HEK-293T cells transfected with the plasmids for overexpression of CBL, NCK2, or both. Cycloheximide tracking and ubiquitination assays were used for assessing the effects of CBL on stability and ubiquitination of NCK2 protein in MDA-MB-231 cells; CCK-8 and Transwell assays were used to determine the effect of NCK2 overexpression on CBL-mediated proliferation and migration of the cells. RESULTS The proliferation, migration and invasion were significantly suppressed in MDA-MB-231 cells overexpressing CBL (P < 0.05) and significantly enhanced in MCF7 cells with CBL silencing (P < 0.01). Silencing of CBL promoted G1/S transition in MCF7 cells (P < 0.05). Overexpression of CBL significantly decreased the expressions of CDK2/4 (P < 0.01), cyclinA2/B1/D1/D3/E2 (P < 0.05), Snail, N-cadherin, claudin-1 (P < 0.05), and upregulated the expression of E-cadherin (P < 0.05). CBL silencing upregulated the expressions of CDK2/4/6 (P < 0.05), cyclin A2/B1/D1/D3/E2 (P < 0.05), Snail, vimentin, and claudin-1 (P < 0.05) and down-regulated E-cadherin expression (P < 0.05). CBL overexpression obviously reduced the number of filamentous pseudopodia in MDA-MB-231 cells, and the reverse changes were observed in MCF7 cells with CBL silencing. In MDA-MB-231 cells, CBL overexpression lowered NCK2 protein stability (P < 0.05) and promoted its ubiquitin-mediated degradation (P < 0.01). Overexpression of NCK2 obviously reversed CBL-mediated inhibition of cell proliferation and migration (P < 0.01). CONCLUSION CBL can inhibit the proliferation, migration and invasion of breast cancer cells through ubiquitination-mediated degradation of NCK2.
Collapse
Affiliation(s)
- 筱羽 宋
- 南方医科大学检验与生物技术学院,广东 广州 510515School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - 斌 肖
- 广州医科大学附属第六医院,清远市人民医院检验医学部,广东 清远 511500Department of Laboratory Medicine, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511500, China
| | - 景润 陆
- 贵阳市第一人民医院医学检验科,贵州 贵阳 550002Department of Laboratory Medicine, First People's Hospital of Guiyang, Guiyang 550002, China
| | - 文武 张
- 广州医科大学附属第六医院,清远市人民医院检验医学部,广东 清远 511500Department of Laboratory Medicine, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511500, China
| | - 锦潮 李
- 中国人民解放军南部战区总医院检验科,广东 广州 510010Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou 510010, China
| | - 昕 竹
- 广州医科大学附属第六医院,清远市人民医院检验医学部,广东 清远 511500Department of Laboratory Medicine, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511500, China
| | - 朝晖 孙
- 中国人民解放军南部战区总医院检验科,广东 广州 510010Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou 510010, China
| | - 林海 李
- 南方医科大学检验与生物技术学院,广东 广州 510515School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
- 广州医科大学附属第六医院,清远市人民医院检验医学部,广东 清远 511500Department of Laboratory Medicine, Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511500, China
| |
Collapse
|
20
|
TRPV1 Is a Potential Tumor Suppressor for Its Negative Association with Tumor Proliferation and Positive Association with Antitumor Immune Responses in Pan-Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6964550. [PMID: 36304985 PMCID: PMC9596243 DOI: 10.1155/2022/6964550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022]
Abstract
Background Although numerous studies have shown that the expression and activation of TRPV1 have an important role in cancer development, a comprehensive exploration of associations between TRPV1 expression and tumor proliferation, microenvironment, and clinical outcomes in pan-cancer remains insufficient. Methods From The Cancer Genome Atlas (TCGA) program, we downloaded multiomics data of ten cancer cohorts and investigated the correlations between TRPV1 expression and immune signatures' enrichment, stromal content, genomic features, oncogenic signaling, and clinical features in these cancer cohorts and pan-cancer. Results Elevated expression of TRPV1 correlated with better clinical outcomes in pan-cancer and diverse cancer types. In multiple cancer types, TRPV1 expression correlated negatively with the expression of tumor proliferation marker genes (MKI67 and RACGAP1), proliferation scores, cell cycle scores, stemness scores, epithelial-mesenchymal transition scores, oncogenic pathways' enrichment, tumor immunosuppressive signals, intratumor heterogeneity, homologous recombination deficiency, tumor mutation burden, and stromal content. Moreover, TRPV1 expression was downregulated in late-stage versus early-stage tumors. In breast cancer, bladder cancer, and low-grade glioma, TRPV1 expression was more inferior in invasive than in noninvasive subtypes. Pathway analysis showed that the enrichment of cancer-associated pathways correlated inversely with TRPV1 expression levels. Conclusion TRPV1 upregulation correlates with decreased tumor proliferation, tumor driver gene expression, genomic instability, and tumor immunosuppressive signals in various cancers. Our results provide new understanding of the role of TRPV1 in both cancer biology and clinical practice.
Collapse
|
21
|
Integrated Machine Learning and Single-Sample Gene Set Enrichment Analysis Identifies a TGF-Beta Signaling Pathway Derived Score in Headneck Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:3140263. [PMID: 36090900 PMCID: PMC9458367 DOI: 10.1155/2022/3140263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
Background The TGF-β signaling pathway is clinically predictive of pan-cancer. Nevertheless, its clinical prognosis and regulation of immune microenvironment (TME) characteristics as well as the prediction of immunotherapy efficacy need to be further elucidated in head and neck squamous cell carcinoma. Method At first, we summarized TGF-β related genes from previous published articles, used ssGSEA to establish the TGF-β risk score. Considering the complexity of its clinical application, we improved it with the LASSO-COX algorithm to construct the model. In addition, we explored the predictive efficacy of TGF-β risk score in the observation of TME phenotype and immunotherapy effect. Finally, the potency of TGF-β risk score in adjusting precise treatment of HNSC was evaluated. Results We systematically established TGF-β risk score with multi-level predictive ability. TGF-β risk score was employed to predict the tumor microenvironment status, which was negatively associated with NK cells but positively related to macrophages and fibroblasts. It reveals that patients with high TGF-β risk score predict “cold” TME status. In addition, higher risk scores indicate higher sensitivity to immunotherapy. Conclusion We first construct and validate TGF-β characteristics that can predict immune microenvironment phenotypes and immunotherapeutic effect in multiple datasets. Noteworthy, TGF-β risk score is helpful for individualized precise treatment of patients with the head and neck squamous cell carcinoma.
Collapse
|
22
|
Shen H, Xu X, Fu Z, Xu C, Wang Y. The interactions of CAP and LYN with the insulin signaling transducer CBL play an important role in polycystic ovary syndrome. Metabolism 2022; 131:155164. [PMID: 35217034 DOI: 10.1016/j.metabol.2022.155164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/28/2022] [Accepted: 02/13/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a hormonal disorder characterized by hyperandrogenism, ovulatory dysfunction, and insulin resistance. Evidence suggests that aberrations in insulin signaling-associated pathways may underlie PCOS pathogenesis. Our aim was to investigate the molecular mechanisms underlying PCOS and associated insulin resistance using in silico analyses, in vitro cell models, and in vivo murine models. METHODS R-based bioinformatics analysis was performed on granulosa cell microarray data from three human cohorts: healthy control, PCOS patients without insulin resistance, and PCOS patients with insulin resistance. Transgenic human granulosa cell models were utilized for in vitro studies. Transgenic murine models of dehydroepiandrosterone (DHEA)-induced PCOS were utilized for in vivo studies. RESULTS Sorbin and SH3 Domain Containing 1 (SORBS1), the parent gene of the insulin receptor-associated Casitas B-lineage lymphoma protein (CBL)-associated protein (CAP), is a key downregulated gene in PCOS patients with insulin resistance. CAP binding to CBL reduced CBLY731 phosphorylation, CBL-phosphoinositide 3-kinase (PI3K) p85α interactivity, protein kinase B (Akt)S473 phosphorylation, and NFκB-induced inflammatory marker expression but enhanced CRKII-mediated membrane GLUT4 translocation in granulosa cells. In contrast, the tyrosine kinase Lck/Yes-Related Novel Protein (LYN) is upregulated in PCOS patients with insulin resistance. LYN binding to CBL enhanced CBLY731 phosphorylation, CBL-PI3K p85α interactivity, AktS473 phosphorylation, and NFκB-induced inflammatory marker expression but did not impact membrane GLUT4 translocation. In PCOS mice, Cap overexpression, Cap transactivation by metformin, or enhancing Cbl-CrkII binding improved insulin sensitivity and ovarian dysfunction (i.e., estrous cycle disruption, cyst-like follicle formation, and sex hormone dysregulation). In contrast, Lyn knockdown, Lyn inhibition by PP2, or CBL-PI3K p85α blockade improved only ovarian dysfunction. Cbl3YF phosphomutant overexpression (which enhances Cbl-CrkII binding but blocks Cbl-PI3K p85α binding) ameliorated both ovarian dysfunction and insulin resistance. CONCLUSIONS The interactions of CAP and LYN with CBL, and the resulting effects on CBL phosphorylation and activity, may play an important role in PCOS pathogenesis. Targeting these players may be a viable therapeutic strategy for PCOS.
Collapse
Affiliation(s)
- Haoran Shen
- Department of Gynecology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200011, PR China.
| | - Xiao Xu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhongpeng Fu
- Department of Ultrasonography, Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Chengjie Xu
- Department of Intelligence Science, Obstetrics & Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Yao Wang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai 200011, PR China.
| |
Collapse
|
23
|
Xiao D, Hu X, Peng M, Deng J, Zhou S, Xu S, Wu J, Yang X. Inhibitory role of proguanil on the growth of bladder cancer via enhancing EGFR degradation and inhibiting its downstream signaling pathway to induce autophagy. Cell Death Dis 2022; 13:499. [PMID: 35614042 PMCID: PMC9132982 DOI: 10.1038/s41419-022-04937-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 12/14/2022]
Abstract
A major reason for the high mortality of patients with bladder cancer (BC) is that chemotherapy and surgery are only effective for very limited patients. Thus, developing novel treatment options becomes an urgent need for improving clinical outcomes and the quality of life for BC patients. Here, we demonstrated that proguanil significantly inhibited the growth of BC in vitro and in vivo. Importantly, our results indicated that the sensitivity of BC cells to proguanil is positively correlated with the expression of epidermal growth factor receptor (EGFR). Mechanistically, proguanil specifically targeted EGFR and promoted EGFR binding to Caveolin-1, enhanced its endocytosis in a Clathrin-independent manner, and then recruited c-Cbl to promote EGFR ubiquitination and degradation through the lysosomal pathway. Further studies suggested that proguanil induced autophagy by destabilizing EGFR and inhibiting its downstream signaling pathway. Thus, this study reveals the novel mechanism of proguanil on anticancer activity and implies the potential benefits of this drug in the treatment of BC.
Collapse
Affiliation(s)
- Di Xiao
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Xin Hu
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Mei Peng
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Jun Deng
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Sichun Zhou
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Simeng Xu
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Jingtao Wu
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China
| | - Xiaoping Yang
- grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan China ,grid.411427.50000 0001 0089 3695Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, Hunan Normal University, Changsha, Hunan China
| |
Collapse
|
24
|
Molecular Pathogenesis of Myeloproliferative Neoplasms: From Molecular Landscape to Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23094573. [PMID: 35562964 PMCID: PMC9100530 DOI: 10.3390/ijms23094573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 12/27/2022] Open
Abstract
Despite distinct clinical entities, the myeloproliferative neoplasms (MPN) share morphological similarities, propensity to thrombotic events and leukemic evolution, and a complex molecular pathogenesis. Well-known driver mutations, JAK2, MPL and CALR, determining constitutive activation of JAK-STAT signaling pathway are the hallmark of MPN pathogenesis. Recent data in MPN patients identified the presence of co-occurrence somatic mutations associated with epigenetic regulation, messenger RNA splicing, transcriptional mechanism, signal transduction, and DNA repair mechanism. The integration of genetic information within clinical setting is already improving patient management in terms of disease monitoring and prognostic information on disease progression. Even the current therapeutic approaches are limited in disease-modifying activity, the expanding insight into the genetic basis of MPN poses novel candidates for targeted therapeutic approaches. This review aims to explore the molecular landscape of MPN, providing a comprehensive overview of the role of drive mutations and additional mutations, their impact on pathogenesis as well as their prognostic value, and how they may have future implications in therapeutic management.
Collapse
|
25
|
Genomic and Epigenomic Landscape of Juvenile Myelomonocytic Leukemia. Cancers (Basel) 2022; 14:cancers14051335. [PMID: 35267643 PMCID: PMC8909150 DOI: 10.3390/cancers14051335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myelodysplastic/myeloproliferative neoplasm characterized by the constitutive activation of the RAS pathway. In spite of the recent progresses in the molecular characterization of JMML, this disease is still a clinical challenge due to its heterogeneity, difficult diagnosis, poor prognosis, and the lack of curative treatment options other than hematopoietic stem cell transplantation (HSCT). In this review, we will provide a detailed overview of the genetic and epigenetic alterations occurring in JMML, and discuss their clinical relevance in terms of disease prognosis and risk of relapse after HSCT. We will also present the most recent advances on novel preclinical and clinical therapeutic approaches directed against JMML molecular targets. Finally, we will outline future research perspectives to further explore the oncogenic mechanism driving JMML leukemogenesis and progression, with special attention to the application of single-cell next-generation sequencing technologies. Abstract Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm of early childhood. Most of JMML patients experience an aggressive clinical course of the disease and require hematopoietic stem cell transplantation, which is currently the only curative treatment. JMML is characterized by RAS signaling hyperactivation, which is mainly driven by mutations in one of five genes of the RAS pathway, including PTPN11, KRAS, NRAS, NF1, and CBL. These driving mutations define different disease subtypes with specific clinico-biological features. Secondary mutations affecting other genes inside and outside the RAS pathway contribute to JMML pathogenesis and are associated with a poorer prognosis. In addition to these genetic alterations, JMML commonly presents aberrant epigenetic profiles that strongly correlate with the clinical outcome of the patients. This observation led to the recent publication of an international JMML stratification consensus, which defines three JMML clinical groups based on DNA methylation status. Although the characterization of the genomic and epigenomic landscapes in JMML has significantly contributed to better understand the molecular mechanisms driving the disease, our knowledge on JMML origin, cell identity, and intratumor and interpatient heterogeneity is still scarce. The application of new single-cell sequencing technologies will be critical to address these questions in the future.
Collapse
|
26
|
Role of CBL Mutations in Cancer and Non-Malignant Phenotype. Cancers (Basel) 2022; 14:cancers14030839. [PMID: 35159106 PMCID: PMC8833995 DOI: 10.3390/cancers14030839] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary CBL mutations are progressively being described as involved in different clinical manifestations. Somatic CBL mutations can be found in different type of cancer. The clinical spectrum of germline mutations configures the so-called CBL syndrome, a cancer-predisposing condition that includes multisystemic involvement characterized by variable phenotypic expression and expressivity. In this review we provide an up-to-date review of the clinical manifestation of CBL mutations and of the molecular mechanisms in which CBL exerts its pathogenic role. Abstract CBL plays a key role in different cell pathways, mainly related to cancer onset and progression, hematopoietic development and T cell receptor regulation. Somatic CBL mutations have been reported in a variety of malignancies, ranging from acute myeloid leukemia to lung cancer. Growing evidence have defined the clinical spectrum of germline CBL mutations configuring the so-called CBL syndrome; a cancer-predisposing condition that also includes multisystemic involvement characterized by variable phenotypic expression and expressivity. This review provides a comprehensive overview of the molecular mechanisms in which CBL exerts its function and describes the clinical manifestation of CBL mutations in humans.
Collapse
|
27
|
Renneville A, Patnaik MM, Chan O, Padron E, Solary E. Increasing recognition and emerging therapies argue for dedicated clinical trials in chronic myelomonocytic leukemia. Leukemia 2021; 35:2739-2751. [PMID: 34175902 DOI: 10.1038/s41375-021-01330-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023]
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN). Median overall survival of this aggressive myeloid malignancy is only 2-3 years, with a 15-30% risk of acute leukemic transformation. The paucity of clinical trials specifically designed for CMML has made therapeutic management of CMML patients challenging. As a result, treatment paradigms for CMML patients are largely borrowed from MDS and MPN. The standard of care still relies on hydroxyurea, hypomethylating agents (HMA), and allogeneic stem cell transplantation, this latter option remaining the only potentially curative therapy. To date, approved drugs for CMML treatment are HMA, including azacitidine, decitabine, and more recently the oral combination of decitabine and cedazuridine. However, HMA treatment does not meaningfully alter the natural course of this disease. New treatment approaches for improving CMML-associated cytopenias or targeting the CMML malignant clone are emerging. More than 25 therapeutic agents are currently being evaluated in phase 1 or phase 2 clinical trials for CMML and other myeloid malignancies, often in combination with a HMA backbone. Several novel agents, such as sotatercept, ruxolitinib, lenzilumab, and tagraxofusp have shown promising clinical efficacy in CMML. Current evidence supports the idea that effective treatment in CMML will likely require combination therapy targeting multiple pathways, which emphasizes the need for additional new therapeutic options. This review focuses on recent therapeutic advances and innovative treatment strategies in CMML, including global and molecularly targeted approaches. We also discuss what may help to make progress in the design of rationally derived and disease-modifying therapies for CMML.
Collapse
Affiliation(s)
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Onyee Chan
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Eric Solary
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France. .,Faculty of Medicine, Université Paris-Sud, Le Kremlin-Bicêtre, France. .,Department of Hematology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
28
|
Hua H, Zhang H, Chen J, Wang J, Liu J, Jiang Y. Targeting Akt in cancer for precision therapy. J Hematol Oncol 2021; 14:128. [PMID: 34419139 PMCID: PMC8379749 DOI: 10.1186/s13045-021-01137-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Biomarkers-guided precision therapeutics has revolutionized the clinical development and administration of molecular-targeted anticancer agents. Tailored precision cancer therapy exhibits better response rate compared to unselective treatment. Protein kinases have critical roles in cell signaling, metabolism, proliferation, survival and migration. Aberrant activation of protein kinases is critical for tumor growth and progression. Hence, protein kinases are key targets for molecular targeted cancer therapy. The serine/threonine kinase Akt is frequently activated in various types of cancer. Activation of Akt promotes tumor progression and drug resistance. Since the first Akt inhibitor was reported in 2000, many Akt inhibitors have been developed and evaluated in either early or late stage of clinical trials, which take advantage of liquid biopsy and genomic or molecular profiling to realize personalized cancer therapy. Two inhibitors, capivasertib and ipatasertib, are being tested in phase III clinical trials for cancer therapy. Here, we highlight recent progress of Akt signaling pathway, review the up-to-date data from clinical studies of Akt inhibitors and discuss the potential biomarkers that may help personalized treatment of cancer with Akt inhibitors. In addition, we also discuss how Akt may confer the vulnerability of cancer cells to some kinds of anticancer agents.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingzhu Chen
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jieya Liu
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangfu Jiang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|