1
|
Rocha MR, Castillo-Medina YK, de Lima Coelho BM, Rios LLL, Morgado-Diaz JA. Wnt/β-catenin pathway as a link between therapy resistance-driven epithelial-mesenchymal transition and stemness in colorectal cancer. Cell Biol Int 2025; 49:154-160. [PMID: 39707719 DOI: 10.1002/cbin.12270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/20/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
The high plasticity of cells undergoing epithelial-mesenchymal transition (EMT) promotes increased tumor heterogeneity, and its interaction with tumor-associated stromal cells appears to contribute to developing a stemness phenotype. Cells with these characteristics exhibit increased resistance to chemotherapy and radiotherapy, leading to disease relapse and metastasis. Here, we discuss the activation of the Wnt/β-catenin pathway in promoting EMT and stemness within the context of cellular resistance to these therapies. We discuss whether EMT and cancer stem cells (CSCs) function in conjunction, independently, or if a link is connecting their development. We further propose that this pathway is necessary to establish a connection between these two phenotypes. And suggest that it could hinder the rise of CSCs from treatment-induced EMT cells when inhibited. Understanding this cellular phenomenon might allow the development of new targeted therapies to improve clinical responses, particularly in colorectal cancer.
Collapse
Affiliation(s)
- Murilo Ramos Rocha
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Yuri Kelly Castillo-Medina
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | | | - Luidy Lucas Lopes Rios
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Jose Andres Morgado-Diaz
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Oelschläger L, Künstner A, Frey F, Leitner T, Leypoldt L, Reimer N, Gebauer N, Bastian L, Weisel K, Sailer VW, Röcken C, Klapper W, Konukiewitz B, Murga Penas EM, Forster M, Schub N, Ahmed HMM, Kirfel J, von Bubnoff NCC, Busch H, Khandanpour C. Whole-Exome Sequencing, Mutational Signature Analysis, and Outcome in Multiple Myeloma-A Pilot Study. Int J Mol Sci 2024; 25:13418. [PMID: 39769182 PMCID: PMC11680055 DOI: 10.3390/ijms252413418] [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: 10/31/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The complex and heterogeneous genomic landscape of multiple myeloma (MM) and many of its clinical and prognostic implications remains to be understood. In other cancers, such as breast cancer, using whole-exome sequencing (WES) and molecular signatures in clinical practice has revolutionized classification, prognostic prediction, and patient management. However, such integration is still in its early stages in MM. In this study, we analyzed WES data from 35 MM patients to identify potential mutational signatures and driver mutations correlated with clinical and cytogenetic characteristics. Our findings confirm the complex mutational spectrum and its impact on previously described ontogenetic and epigenetic pathways. They show TYW1 as a possible new potential driver gene and find no significant associations of mutational signatures with clinical findings. Further studies are needed to strengthen the role of mutational signatures in the clinical context of patients with MM to improve patient management.
Collapse
Affiliation(s)
- Lorenz Oelschläger
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany
| | - Friederike Frey
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| | - Theo Leitner
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| | - Lisa Leypoldt
- Department of Hematology, Oncology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20521 Hamburg, Germany
| | - Niklas Reimer
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| | - Lorenz Bastian
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany
- Division for Stem Cell Transplantation and Immunotherapy, University Hospital of Schleswig-Holstein, 24105 Kiel, Germany
| | - Katja Weisel
- Department of Hematology, Oncology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20521 Hamburg, Germany
| | - Verena-Wilbeth Sailer
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany
- Department of Pathology, University of Lübeck, 23538 Lübeck, Germany
| | - Christoph Röcken
- Department of Pathology, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany
| | - Wolfram Klapper
- Department of Pathology, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany
| | - Björn Konukiewitz
- Department of Pathology, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany
| | - Eva Maria Murga Penas
- Institute of Human Genetics, University Hospital Schleswig-Holstein (UKSH)/Christian-Albrechts University Kiel (CAU), 24105 Kiel, Germany
| | - Michael Forster
- Institute of Clinical Molecular Biology, Christian-Albrechts University, 24105 Kiel, Germany
| | - Natalie Schub
- Division for Stem Cell Transplantation and Immunotherapy, University Hospital of Schleswig-Holstein, 24105 Kiel, Germany
| | - Helal M. M. Ahmed
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| | - Jutta Kirfel
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany
- Department of Pathology, University of Lübeck, 23538 Lübeck, Germany
| | - Nikolas Christian Cornelius von Bubnoff
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, 23538 Lübeck, Germany
| | - Cyrus Khandanpour
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein (UKSH), University Cancer Center Schleswig-Holstein (UCCSH), Campus Lübeck, 23538 Lübeck, Germany
| |
Collapse
|
3
|
Tsukanov VV, Tonkikh JL, Kasparov EV, Vasyutin AV. Inhibition of M2 tumor-associated macrophages polarization by modulating the Wnt/β-catenin pathway as a possible liver cancer therapy method. World J Gastroenterol 2024; 30:4399-4403. [PMID: 39494099 PMCID: PMC11525861 DOI: 10.3748/wjg.v30.i40.4399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/09/2024] [Accepted: 09/26/2024] [Indexed: 10/16/2024] Open
Abstract
The problem of liver cancer is becoming increasingly important due to the epidemic of metabolic diseases and persistent high alcohol consumption. This determines great attention to the development and improvement of methods for early diagnosis and treatment of liver cancer. Huang et al presented a study in the World Journal of Gastroenterology, in which they showed that the use of the traditional Chinese medicine Calculus bovis (CB) can suppress tumor growth in mice by inhibiting M2 tumor-associated macrophages (TAM) through modulating the activity of the Wnt/β-catenin pathway. The interaction of CB components with the Wnt/β-catenin pathway, M2 TAM polarization, and tumor dynamics were studied using network pharmacology, transcriptomics, and molecular docking. It is now generally accepted that the polarization of TAM and the differentiation of the functions of M1 and M2 phagocytes are of great importance for the progression of neoplasms. It is assumed that M2 TAM promote proliferation and migration of tumor cells. Attempts to medicinally influence the Wnt/β-catenin pathway in order to modulate phagocyte polarization now belong to one of the most promising areas of immunotherapy of oncological diseases. Undoubtedly, the work of the Chinese authors deserves attention and further development.
Collapse
Affiliation(s)
- Vladislav V Tsukanov
- Clinical Department of the Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| | - Julia L Tonkikh
- Clinical Department of the Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| | - Edward V Kasparov
- Clinical Department of the Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| | - Alexander V Vasyutin
- Clinical Department of the Digestive System Pathology of Adults and Children, Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, Krasnoyarsk 660022, Russia
| |
Collapse
|
4
|
Masci D, Puxeddu M, Silvestri R, La Regina G. Targeting CBP and p300: Emerging Anticancer Agents. Molecules 2024; 29:4524. [PMID: 39407454 PMCID: PMC11482477 DOI: 10.3390/molecules29194524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
CBP and p300 are versatile transcriptional co-activators that play essential roles in regulating a wide range of signaling pathways, including Wnt/β-catenin, p53, and HIF-1α. These co-activators influence various cellular processes such as proliferation, differentiation, apoptosis, and response to hypoxia, making them pivotal in normal physiology and disease progression. The Wnt/β-catenin signaling pathway, in particular, is crucial for cellular proliferation, differentiation, tissue homeostasis, and embryogenesis. Aberrant activation of this pathway is often associated with several types of cancer, such as colorectal tumor, prostate cancer, pancreatic and hepatocellular carcinomas. In recent years, significant efforts have been directed toward identifying and developing small molecules as novel anticancer agents capable of specifically inhibiting the interaction between β-catenin and the transcriptional co-activators CBP and p300, which are required for Wnt target gene expression and are consequently involved in the regulation of tumor cell proliferation, migration, and invasion. This review summarizes the most significant and original research articles published from 2010 to date, found by means of a PubMed search, highlighting recent advancements in developing both specific and non-specific inhibitors of CBP/β-catenin and p300/β-catenin interactions. For a more comprehensive view, we have also explored the therapeutic potential of CBP/p300 bromodomain and histone acetyltransferase inhibitors in disrupting the transcriptional activation of genes involved in various signaling pathways related to cancer progression. By focusing on these therapeutic strategies, this review aims to offer a detailed overview of recent approaches in cancer treatment that selectively target CBP and p300, with particular emphasis on their roles in Wnt/β-catenin-driven oncogenesis.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| |
Collapse
|
5
|
Hedayati N, Mafi A, Farahani A, Hashemi M, Nabavi N, Alimohammadi M, Rahimzadeh P, Taheriazam A, Farahani N. The importance of the circRNA/Wnt axis in gliomas: Biological functions and clinical opportunities. Pathol Res Pract 2024; 261:155510. [PMID: 39116573 DOI: 10.1016/j.prp.2024.155510] [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: 05/25/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Gliomas are among the most common cancers in the central nervous system, arising through various signaling pathways. One significant pathway is Wnt signaling, a tightly regulated process that plays a crucial role in gliomagenesis and development. The current study aims to explore the relationship between circular RNAs (circRNAs) and the Wnt/β-catenin signaling pathway in gliomas, considering the growing recognition of circRNAs in disease pathogenesis. A comprehensive review of recent research was conducted to investigate the roles of circRNAs in gliomas, focusing on their expression patterns and interactions with the Wnt signaling pathway. The analysis included studies examining circRNAs' function as microRNA sponges and their impact on glioma biology. The findings reveal that circRNAs are differentially expressed in gliomas and significantly influence the occurrence, growth, and metastasis of these tumors. Specifically, circRNAs interact with the Wnt signaling pathway, affecting glioma development and progression. This interaction highlights the importance of circRNAs in glioma pathophysiology. Understanding the regulatory network involving circRNAs and Wnt signaling offers valuable insights into glioma pathophysiology. CircRNAs hold promise as diagnostic and prognostic biomarkers and may serve as targets for novel therapeutic strategies in glioma treatment.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aryan Farahani
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
6
|
Zhou X, Pan J, Chen L, Zhang S, Chen Y. DeepIMAGER: Deeply Analyzing Gene Regulatory Networks from scRNA-seq Data. Biomolecules 2024; 14:766. [PMID: 39062480 PMCID: PMC11274664 DOI: 10.3390/biom14070766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Understanding the dynamics of gene regulatory networks (GRNs) across diverse cell types poses a challenge yet holds immense value in unraveling the molecular mechanisms governing cellular processes. Current computational methods, which rely solely on expression changes from bulk RNA-seq and/or scRNA-seq data, often result in high rates of false positives and low precision. Here, we introduce an advanced computational tool, DeepIMAGER, for inferring cell-specific GRNs through deep learning and data integration. DeepIMAGER employs a supervised approach that transforms the co-expression patterns of gene pairs into image-like representations and leverages transcription factor (TF) binding information for model training. It is trained using comprehensive datasets that encompass scRNA-seq profiles and ChIP-seq data, capturing TF-gene pair information across various cell types. Comprehensive validations on six cell lines show DeepIMAGER exhibits superior performance in ten popular GRN inference tools and has remarkable robustness against dropout-zero events. DeepIMAGER was applied to scRNA-seq datasets of multiple myeloma (MM) and detected potential GRNs for TFs of RORC, MITF, and FOXD2 in MM dendritic cells. This technical innovation, combined with its capability to accurately decode GRNs from scRNA-seq, establishes DeepIMAGER as a valuable tool for unraveling complex regulatory networks in various cell types.
Collapse
Affiliation(s)
- Xiguo Zhou
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Jingyi Pan
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Liang Chen
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Shaoqiang Zhang
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Yong Chen
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
7
|
Li B, Jin K, Liu Z, Su X, Xu Z, Liu G, Xu J, Liu H, Chang Y, Wang Y, Zhu Y, Wang Z, Xu L, Zhang W. Integrating molecular subtype and CD8 + T cells infiltration to predict treatment response and survival in muscle-invasive bladder cancer. Cancer Immunol Immunother 2024; 73:66. [PMID: 38430246 PMCID: PMC10908619 DOI: 10.1007/s00262-024-03651-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/03/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Luminal and Basal are the primary intrinsic subtypes of muscle-invasive bladder cancer (MIBC). The presence of CD8+ T cells infiltration holds significant immunological relevance, potentially influencing the efficacy of antitumor responses. This study aims to synergize the influence of molecular subtypes and CD8+ T cells infiltration in MIBC. METHODS This study included 889 patients with MIBC from Zhongshan Hospital, The Cancer Genome Atlas, IMvigor210 and NCT03179943 cohorts. We classified the patients into four distinct groups, based on the interplay of molecular subtypes and CD8+ T cells and probed into the clinical implications of these subgroups in MIBC. RESULTS Among patients with Luminal-CD8+Thigh tumors, the confluence of elevated tumor mutational burden and PD-L1 expression correlated with a heightened potential for positive responses to immunotherapy. In contrast, patients featured by Luminal-CD8+Tlow displayed a proclivity for deriving clinical advantages from innovative targeted interventions. The Basal-CD8+Tlow subgroup exhibited the least favorable three-year overall survival outcome, whereas their Basal-CD8+Thigh counterparts exhibited a heightened responsiveness to chemotherapy. CONCLUSIONS We emphasized the significant role of immune-molecular subtypes in shaping therapeutic approaches for MIBC. This insight establishes a foundation to refine the process of selecting subtype-specific treatments, thereby advancing personalized interventions for patients.
Collapse
Affiliation(s)
- Bingyu Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaifeng Jin
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaopei Liu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaohe Su
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziyue Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ge Liu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jingtong Xu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hailong Liu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yiwei Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijuan Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Yin P, Zhou X. Potential Clinical Role of LncRNA miR503HG in Multiple Myeloma and its Effect on the Proliferation and Adhesion of Myeloma Cells. Indian J Hematol Blood Transfus 2024; 40:43-51. [PMID: 38312189 PMCID: PMC10830954 DOI: 10.1007/s12288-023-01658-x] [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: 01/03/2023] [Accepted: 04/10/2023] [Indexed: 02/06/2024] Open
Abstract
This study mainly explored the role of lncRNA miR503HG in multiple myeloma and the potential downstream regulatory mechanism affecting disease. Real-time quantitative polymerase chain reaction was used to measure the expression levels of miR503HG and miR-103. A cell counting kit-8 assay was performed to detect cell viability. The concentrations of adhesion-related factors (MUC-1, VCAM-1, ICAM-1) were determined using enzyme-linked immunosorbent assay. The targeting relationship between miR503HG and miR-103 was detected by dual-luciferase reporter assay. The miR503HG expression in peripheral blood of multiple myeloma patients was lower than that of normal healthy individuals and associated with ISS stage and worse overall survival. miR-103 was identified as the downstream target of miR503HG. Upregulation of miR503HG could inhibit cell proliferation and adhesion of multiple myeloma cell lines, which could partially reverse the inhibition of adhesion and proliferation by high expression of miR-103. lncRNA miR503HG expression was downregulated in multiple myeloma and had potential diagnostic/prognostic value. MiR503HG exerts a molecular sponge effect on miR-103 and affects its expression, thus achieving the inhibitory effect on multiple myeloma.
Collapse
Affiliation(s)
- Pingling Yin
- Department of Hematology, Affiliated Hospital of Gansu Medical College, No. 296, Kongtong East Road, Kongtong District, Pingliang, Gansu, 744000 China
| | - Xiang Zhou
- Department of Hematology, Affiliated Hospital of Gansu Medical College, No. 296, Kongtong East Road, Kongtong District, Pingliang, Gansu, 744000 China
| |
Collapse
|
9
|
Kozalak G, Koşar A. Autophagy-related mechanisms for treatment of multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:838-857. [PMID: 38239705 PMCID: PMC10792488 DOI: 10.20517/cdr.2023.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Multiple myeloma (MM) is a type of hematological cancer that occurs when B cells become malignant. Various drugs such as proteasome inhibitors, immunomodulators, and compounds that cause DNA damage can be used in the treatment of MM. Autophagy, a type 2 cell death mechanism, plays a crucial role in determining the fate of B cells, either promoting their survival or inducing cell death. Therefore, autophagy can either facilitate the progression or hinder the treatment of MM disease. In this review, autophagy mechanisms that may be effective in MM cells were covered and evaluated within the contexts of unfolded protein response (UPR), bone marrow microenvironment (BMME), drug resistance, hypoxia, DNA repair and transcriptional regulation, and apoptosis. The genes that are effective in each mechanism and research efforts on this subject were discussed in detail. Signaling pathways targeted by new drugs to benefit from autophagy in MM disease were covered. The efficacy of drugs that regulate autophagy in MM was examined, and clinical trials on this subject were included. Consequently, among the autophagy mechanisms that are effective in MM, the most suitable ones to be used in the treatment were expressed. The importance of 3D models and microfluidic systems for the discovery of new drugs for autophagy and personalized treatment was emphasized. Ultimately, this review aims to provide a comprehensive overview of MM disease, encompassing autophagy mechanisms, drugs, clinical studies, and further studies.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
10
|
Liu T, Zhao J, Zhang X, Wang Y, Wang W, Song J. Wnt pathway in bone: knowledge structure and hot spots from 1993 to 2022. Front Physiol 2023; 14:1279423. [PMID: 38033331 PMCID: PMC10687587 DOI: 10.3389/fphys.2023.1279423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Background: The role of the Wnt pathway in bone and its targets in skeletal disease has garnered interest, but the field lacks a systematic analysis of research. This paper presents a bibliometric study of publications related to the Wnt signaling pathway in bone to describe the current state of study and predict future outlooks. Methods: All relevant articles and reviews from 1993 to 2022 were collected from the Web of Science Core Collection (WoSCC). Bibliometric analysis and visualization were performed using CiteSpace 6.1 R3, VOSviewer 1.6.15, and the Online Analysis Platform of Literature Metrology (http://bibliometric.com/). Results: A total of 7,184 papers were retrieved, authored by 28,443 researchers from 89 countries/regions and published in 261 academic journals. The annual publication numbers peaked in 2021. China and United States are the leading countries, with the University of California and Harvard University as the most active institutions. Wang, Yang is the most prolific author. Bone has the most published research, while Proceedings of the National Academy of Sciences of the United States is the most cited journal on average. The main keywords include expression, Wnt, osteoporosis, bone, and osteogenic differentiation. Current and developing research hotspots focus on bone mass, sclerostin antibody, multiple myeloma, and cartilage development. Conclusion: This paper provides new insights for researchers to delve into the mechanisms of Wnt and bone related diseases and translate into clinical studies. It reveals the development and future research trends in Wnt and skeletal-related studies.
Collapse
Affiliation(s)
| | | | | | | | - Wei Wang
- The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jidong Song
- The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
11
|
Zheng B, Yi K, Zhang Y, Pang T, Zhou J, He J, Lan H, Xian H, Li R. Multi-omics analysis of multiple myeloma patients with differential response to first-line treatment. Clin Exp Med 2023; 23:3833-3846. [PMID: 37515690 DOI: 10.1007/s10238-023-01148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023]
Abstract
The genome backgrounds of multiple myeloma (MM) would affect the efficacy of specific treatment. However, the mutational and transcriptional landscapes in MM patients with differential response to first-line treatment remains unclear. We collected paired whole-exome sequencing (WES) and transcriptomic data of over 200 MM cases from MMRF-COMPASS project. R package, maftools was applied to analyze the somatic mutations and mutational signatures across MM samples. Differential expressed genes (DEG) was calculated using R package, DESeq2. The feature selection of the predictive model was determined by LASSO regression. In silico analysis revealed newly discovered recurrent mutated genes such as TTN, MUC16. TP53 mutation was observed more frequent in nonCR (complete remission) group with poor prognosis. DNA repair-associated mutational signatures were enriched in CR patients. Transcriptomic profiling showed that the activity of NF-kappa B and TGF-β pathways was suppressed in CR patients. A transcriptome-based response predictive model was constructed and showed promising predictive accuracy in MM patients receiving first-line treatment. Our study delineated distinctive mutational and transcriptional landscapes in MM patients with differential response to first-line treatment. Furthermore, we constructed a 20-gene predictive model which showed promising accuracy in predicting treatment response in newly diagnosed MM patients.
Collapse
Affiliation(s)
- Bo Zheng
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China.
| | - Ke Yi
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Yajun Zhang
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Tongfang Pang
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Jieyi Zhou
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Jie He
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Hongyan Lan
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Hongming Xian
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China
| | - Rong Li
- Nuclear Radiation Injury Protection and Treatment Department, Navy Medical Center of PLA, Naval Medical University, Huaihai West Road No. 338, Shanghai, 200050, China.
| |
Collapse
|
12
|
Xu J, Wang Y, Li P, Chen C, Jiang Z, Wang X, Liu P. PRUNE1 (located on chromosome 1q21.3) promotes multiple myeloma with 1q21 Gain by enhancing the links between purine and mitochondrion. Br J Haematol 2023; 203:599-613. [PMID: 37666675 DOI: 10.1111/bjh.19088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/05/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023]
Abstract
Patients with multiple myeloma (MM) with chromosome 1q21 Gain (1q21+) are clinically and biologically heterogeneous. 1q21+ in the real world actually reflects the prognosis for gain/amplification of the CKS1B gene. In this study, we found that the copy number of prune exopolyphosphatase 1 (PRUNE1), located on chromosome 1q21.3, could further stratify the prognosis of MM patients with 1q21+. Using selected reaction monitoring/multiple reaction monitoring (SRM/MRM) analysis, liquid chromatography-tandem mass spectrometry (LC-MS/MS), transmission electron microscopy (TEM), confocal fluorescence microscopy, calculation of adenosine triphosphate (ATP), intracellular reactive oxygen species (ROS) and mitochondrial oxygen consumption rates (OCRs), we demonstrated for the first time that PRUNE1 promotes the proliferation and invasion of MM cells by stimulating purine metabolism, purine synthesis enzymes and mitochondrial functions, enhancing links between purinosomes and mitochondria. SOX11 was identified as a transcription factor for PRUNE1. Through integrated analysis of the transcriptome and proteome, CD73 was determined to be the downstream target of PRUNE1. Furthermore, it has been determined that dipyridamole can effectively suppress the proliferation of MM cells with high-expression levels of PRUNE1 in vitro and in vivo. These findings provide insights into disease-causing mechanisms and new therapeutic targets for MM patients with 1q21+.
Collapse
Affiliation(s)
- Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yawen Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Panpan Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihong Jiang
- Department of Hematology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Xiaona Wang
- Department of Hematology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| |
Collapse
|
13
|
Yang X, Du Y, Luo L, Xu X, Xiong S, Yang X, Guo L, Liang T. Deciphering the Enigmatic Influence: Non-Coding RNAs Orchestrating Wnt/β-Catenin Signaling Pathway in Tumor Progression. Int J Mol Sci 2023; 24:13909. [PMID: 37762212 PMCID: PMC10530696 DOI: 10.3390/ijms241813909] [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: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Dysregulated expression of specific non-coding RNAs (ncRNAs) has been strongly linked to tumorigenesis, cancer progression, and therapeutic resistance. These ncRNAs can act as either oncogenes or tumor suppressors, thereby serving as valuable diagnostic and prognostic markers. Numerous studies have implicated the participation of ncRNAs in the regulation of diverse signaling pathways, including the pivotal Wnt/β-catenin signaling pathway that is widely acknowledged for its pivotal role in embryogenesis, cellular proliferation, and tumor biology control. Recent emerging evidence has shed light on the capacity of ncRNAs to interact with key components of the Wnt/β-catenin signaling pathway, thereby modulating the expression of Wnt target genes in cancer cells. Notably, the activity of this pathway can reciprocally influence the expression levels of ncRNAs. However, comprehensive analysis investigating the specific ncRNAs associated with the Wnt/β-catenin signaling pathway and their intricate interactions in cancer remains elusive. Based on these noteworthy findings, this review aims to unravel the intricate associations between ncRNAs and the Wnt/β-catenin signaling pathway during cancer initiation, progression, and their potential implications for therapeutic interventions. Additionally, we provide a comprehensive overview of the characteristics of ncRNAs and the Wnt/β-catenin signaling pathway, accompanied by a thorough discussion of their functional roles in tumor biology. Targeting ncRNAs and molecules associated with the Wnt/β-catenin signaling pathway may emerge as a promising and effective therapeutic strategy in future cancer treatments.
Collapse
Affiliation(s)
- Xinbing Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Yajing Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Lulu Luo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Xinru Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Shizheng Xiong
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Xueni Yang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| |
Collapse
|
14
|
Mansoor A, Kamran H, Akhter A, Seno R, Torlakovic EE, Roshan TM, Shabani-Rad MT, Elyamany G, Minoo P, Stewart D. Identification of Potential Therapeutic Targets for Plasmablastic Lymphoma Through Gene Expression Analysis: Insights into RAS and Wnt Signaling Pathways. Mod Pathol 2023; 36:100198. [PMID: 37105495 DOI: 10.1016/j.modpat.2023.100198] [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: 01/09/2023] [Revised: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Plasmablastic lymphoma (PBL) is a rare and aggressive B-cell lymphoma with overlapping characteristics with diffuse large B-cell lymphoma (DLBCL) and multiple myeloma. Hyperactive Wnt signaling derails homeostasis and promotes oncogenesis and chemoresistance in DLBCL and multiple myeloma. Evidence suggests active cross-talk between the Wnt and RAS pathways impacting metastasis in solid cancers in which combined targeted therapies show effective results. Recent genomic studies in PBL demonstrated a high frequency of mutations linked with the RAS signaling pathway. However, the role of RAS and Wnt signaling pathway molecule expression in PBL remained unknown. We examined the expression of Wnt and RAS pathway-related genes in a well-curated cohort of PBL. Because activated B cells are considered immediate precursors of plasmablasts in B cell development, we compared this data with activated B-cell type DLBCL (ABC-DLBCL) patients, employing NanoString transcriptome analysis (770 genes). Hierarchical clustering revealed distinctive differential gene expression between PBL and ABC-DLBCL. Gene set enrichment analysis labeled the RAS signaling pathway as the most enriched (37 genes) in PBL, including upregulating critical genes, such as NRAS, RAF1, SHC1, and SOS1. Wnt pathway genes were also enriched (n = 22) by gene set enrichment analysis. Molecules linked with Wnt signaling activation, such as ligands or targets (FZD3, FZD7, c-MYC, WNT5A, WNT5B, and WNT10B), were elevated in PBL. Our data also showed that, unlike ABC-DLBCL, the deranged Wnt signaling activity in PBL was not linked with hyperactive nuclear factor κB and B-cell receptor signaling. In divergence, Wnt signaling inhibitors (CXXC4, SFRP2, and DKK1) also showed overexpression in PBL. The high expression of RAS signaling molecules reported may indicate linkage with gain-in-function RAS mutations. In addition, high expression of Wnt and RAS signaling molecules may pave pathways to explore benefiting from combined targeted therapies, as reported in solid cancer, to improve prognosis in PBL patients.
Collapse
Affiliation(s)
- Adnan Mansoor
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada.
| | - Hamza Kamran
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada
| | - Ariz Akhter
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada
| | - Rommel Seno
- Department of Pathology & Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Emina E Torlakovic
- Department of Pathology & Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Tariq Mahmood Roshan
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada
| | - Meer-Taher Shabani-Rad
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada
| | - Ghaleb Elyamany
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada
| | - Parham Minoo
- Department of Pathology & Laboratory Medicine, University of Calgary, and Alberta Precision Laboratories (APL), Calgary, Alberta, Canada
| | - Douglas Stewart
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| |
Collapse
|
15
|
Enukashvily NI, Belik LA, Semenova NY, Kostroma II, Motyko EV, Gritsaev SV, Bessmeltsev SS, Sidorkevich SV, Martynkevich IS. Transcription of WNT Genes in Hematopoietic Niche's Mesenchymal Stem Cells in Multiple Myeloma Patients with Different Responses to Treatment. Genes (Basel) 2023; 14:genes14051097. [PMID: 37239457 DOI: 10.3390/genes14051097] [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: 02/10/2023] [Revised: 04/20/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are involved in bone tissue remodeling due to their ability to differentiate into osteoblasts and to influence osteoclasts' activity. Multiple myeloma (MM) is associated with bone resorption. During disease progression, MSCs acquire a tumor-associated phenotype, losing their osteogenic potential. The process is associated with impaired osteoblasts/osteoclasts balance. The WNT signaling pathway plays a major role in maintaining the balance. In MM, it functions in an aberrant way. It is not known yet whether the WNT pathway is restored in patients' bone narrow after treatment. The aim of the study was to compare the level of WNT family gene transcription in the bone marrow MSCs of healthy donors and MM patients before and after therapy. The study included healthy donors (n = 3), primary patients (n = 3) and patients with different response status to therapy (bortezomib-containing induction regimens) (n = 12). The transcription of the WNT and CTNNB1 (encoding β-catenin) genes was accessed using qPCR. The mRNA quantity of ten WNT genes, as well as CTNNB1 mRNA encoding β-catenin, a key mediator in canonical signaling, was evaluated. The observed differences between the groups of patients indicated that aberrant functioning of the WNT pathway was retained after treatment. The differences that we detected for WNT2B, WNT9B and CTNNB1 suggested their possible application as prognostic molecular markers.
Collapse
Affiliation(s)
- Natella I Enukashvily
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
- Cell Technologies Lab., North-Western State Medical University named after I.I. Mechnikov, 191015 St. Peterburg, Russia
| | - Liubov A Belik
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
- Cell Technology Center Pokrovsky, 199066 St. Petersburg, Russia
| | - Natalia Yu Semenova
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| | - Ivan I Kostroma
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| | - Ekaterina V Motyko
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| | - Sergey V Gritsaev
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| | - Stanislav S Bessmeltsev
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| | - Sergey V Sidorkevich
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| | - Irina S Martynkevich
- Russian Research Institute of Hematology and Transfusiology, FMBA of Russian Federation, 191024 St. Petersburg, Russia
| |
Collapse
|
16
|
Zhu M, Sun Y, Bai H, Wang Y, Yang B, Wang Q, Kuang H. Effects of saponins from Chinese herbal medicines on signal transduction pathways in cancer: A review. Front Pharmacol 2023; 14:1159985. [PMID: 37063281 PMCID: PMC10090286 DOI: 10.3389/fphar.2023.1159985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Cancer poses a serious threat to human health, and the search for safe and effective drugs for its treatment has aroused interest and become a long-term goal. Traditional Chinese herbal medicine (TCM), an ancient science with unique anti-cancer advantages, has achieved outstanding results in long-term clinical practice. Accumulating evidence shows that saponins are key bioactive components in TCM and have great research and development applications for their significant role in the treatment of cancer. Saponins are a class of glycosides comprising nonpolar triterpenes or sterols attached to hydrophilic oligosaccharide groups that exert antitumor effects by targeting the NF-κB, PI3Ks-Akt-mTOR, MAPK, Wnt-β-catenin, JAK-STAT3, APMK, p53, and EGFR signaling pathways. Presently, few advances have been made in physiological and pathological studies on the effect of saponins on signal transduction pathways involved in cancer treatment. This paper reviews the phytochemistry and extraction methods of saponins of TCM and their effects on signal transduction pathways in cancer. It aims to provide theoretical support for in-depth studies on the anticancer effects of saponins.
Collapse
Affiliation(s)
- Mingtao Zhu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Haodong Bai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yimeng Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Qiuhong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Qiuhong Wang, ; Haixue Kuang,
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
- *Correspondence: Qiuhong Wang, ; Haixue Kuang,
| |
Collapse
|
17
|
Molecular Features of the Mesenchymal and Osteoblastic Cells in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232415448. [PMID: 36555090 PMCID: PMC9779562 DOI: 10.3390/ijms232415448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a monoclonal gammopathy characterized by biological heterogeneity and unregulated proliferation of plasma cells (PCs) in bone marrow (BM). MM is a multistep process based on genomic instability, epigenetic dysregulation and a tight cross-talk with the BM microenvironment that plays a pivotal role supporting the proliferation, survival, drug-resistance and homing of PCs. The BM microenvironment consists of a hematopoietic and a non-hematopoietic compartment, which cooperate to create a tumor environment. Among the non-hematopoietic component, mesenchymal stromal cells (MSCs) and osteoblasts (OBs) appear transcriptionally and functionally different in MM patients compared to healthy donors (HDs) and to patients with pre-malignant monoclonal gammopathies. Alterations of both MSCs and OBs underly the osteolytic lesions that characterize myeloma-associated bone disease. In this review, we will discuss the different characteristics of MSCs and OBs in MM patients, analyzing the transcriptome, the deregulated molecular pathways and the role performed by miRNAs and exosome in the pathophysiology of MM.
Collapse
|
18
|
Connors JP, Stelzer JW, Garvin PM, Wellington IJ, Solovyova O. The Role of the Innate Immune System in Wear Debris-Induced Inflammatory Peri-Implant Osteolysis in Total Joint Arthroplasty. Bioengineering (Basel) 2022; 9:764. [PMID: 36550970 PMCID: PMC9774505 DOI: 10.3390/bioengineering9120764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Periprosthetic osteolysis remains a leading complication of total hip and knee arthroplasty, often resulting in aseptic loosening of the implant and necessitating revision surgery. Wear-induced particulate debris is the main cause initiating this destructive process. The purpose of this article is to review recent advances in understanding of how wear debris causes osteolysis, and emergent strategies for the avoidance and treatment of this disease. A strong activator of the peri-implant innate immune this debris-induced inflammatory cascade is dictated by macrophage secretion of TNF-α, IL-1, IL-6, and IL-8, and PGE2, leading to peri-implant bone resorption through activation of osteoclasts and inhibition of osteoblasts through several mechanisms, including the RANK/RANKL/OPG pathway. Therapeutic agents against proinflammatory mediators, such as those targeting tumor necrosis factor (TNF), osteoclasts, and sclerostin, have shown promise in reducing peri-implant osteolysis in vitro and in vivo; however, radiographic changes and clinical diagnosis often lag considerably behind the initiation of osteolysis, making timely treatment difficult. Considerable efforts are underway to develop such diagnostic tools, therapies, and identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- John Patrick Connors
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - John W Stelzer
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Patrick M Garvin
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Ian J Wellington
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Olga Solovyova
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| |
Collapse
|
19
|
Liu Q, Ran R, Song M, Li X, Wu Z, Dai G, Xia R. LncRNA HCP5 acts as a miR-128-3p sponge to promote the progression of multiple myeloma through activating Wnt/β-catenin/cyclin D1 signaling via PLAGL2. Cell Biol Toxicol 2022; 38:979-993. [PMID: 34331612 DOI: 10.1007/s10565-021-09628-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although long non-coding RNA (lncRNA) HCP plays essential roles in human cancers, its function and mechanism in multiple myeloma (MM) have not crystallized. METHODS HCP5 level in MM was assessed through qRT-PCR. A series of functional investigations were conducted to evaluate the influences of HCP5 on proliferation and apoptosis. Bioinformatics analysis and RIP/RNA pull-down assays were carried out to determine the relationships among HCP5, miR-128-3p, and PLAGL2. Relative protein level was determined through Western blot. A xenograft tumor model was applied for validating the roles of HCP5/miR-128-3p/PLAGL2 axis in vivo. RESULTS HCP5 was significantly increased in MM. HCP5 knockdown effectively thwarted the proliferative rate and cell cycle of MM cell lines and suppressed tumor growth. HCP5 regulated PLAGL2 expression by sponging miR-128-3p. PLAGL2 overexpression effectively rescued cells from influences by sh-HCP5 on cell proliferative and apoptotic rates. Additionally, HCP5 knockdown significantly inhibited Wnt/β-catenin/cyclin D1 signaling, and these effects were eliminated by PLAGL2 overexpression. CONCLUSION Our study revealed that HCP5/miR-128-3p/PLAGL2 is closely correlated to MM development by modulating Wnt/β-catenin/cyclin D1 signaling. HCP5 promoted cell proliferation and tumor formation of MM cells by activating the Wnt/β-catenin/CCND1 signaling pathway by sponging miR-128-3p to increase PLAGL2 expression.
Collapse
Affiliation(s)
- Qinhua Liu
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ruonan Ran
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingyue Song
- Department of Hematology, the Chaohu Hospital Affiliated to Anhui Medical University, Chaohu, Anhui, China
| | - Xiaodan Li
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Zhengsheng Wu
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guanrong Dai
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ruixiang Xia
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
| |
Collapse
|
20
|
Li R, Ke M, Qi M, Han Z, Cao Y, Deng Z, Qian J, Yang Y, Gu C. G6PD promotes cell proliferation and dexamethasone resistance in multiple myeloma via increasing anti-oxidant production and activating Wnt/β-catenin pathway. Exp Hematol Oncol 2022; 11:77. [PMID: 36271440 PMCID: PMC9587560 DOI: 10.1186/s40164-022-00326-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/30/2022] [Indexed: 11/12/2022] Open
Abstract
Background Glucose-6-phosphate dehydrogenase (G6PD) as the rate-limiting enzyme in the pentose phosphate pathway (PPP) is well-established as an aberrantly expressed protein in numerous clinical diseases; however, its role in cancer, specifically in multiple myeloma (MM) remains elusive. Methods In this study, serum metabolites in 70 normal people and 70 newly diagnosed MM patients were analyzed using untargeted metabolomics and the results were verified using ELISA. The survival analysis of multiple clinical datasets was performed to identify a potential target gene in MM. The oncogenic role of G6PD was investigated using lentivirus-based overexpression or knockdown of G6PD using RNAi or an inhibitor in vitro, and in a xenograft mouse model in vivo. The mechanisms of induced Dexamethasone (Dexa)-resistance of G6PD were further explored using the above established MM cell lines in vitro. Results Based on the screening of potential genes, PPP was shown to be involved in the occurrence of MM, which was evidenced by the differential expression of serum metabolites of G6P and Dehydroepiandrosterone sulfate (DHEAS, the more stable sulfate ester form of an endogenously uncompetitive G6PD inhibitor known as DHEA). Elevated G6PD promoted MM cell proliferation. Mechanistically, high G6PD expression enhanced enzymatic generation of the antioxidant NADPH via the PPP and decreased the production of reactive oxygen species (ROS), thus inducing the proliferation and Dexa resistance in MM cells. Furthermore, canonical Wnt/β-catenin signaling also participated in regulating G6PD-induced drug resistance and cellular redox levels of ROS. Intriguingly, DHEA treatment could enhance the sensitivity of MM cells to Dexa primarily through augmenting cellular oxidative stress. Conclusions Our data demonstrate that G6PD enhances the generation of the enzymatic anti-oxidant NADPH and decreases ROS generation, thereby promoting resistance to Dexa-induced apoptosis via the enzymatic PPP and non-enzymatic Wnt/β-catenin signaling pathway in MM. Targeting G6PD to harness cellular redox may serve as a promising novel strategy for the management of MM. Supplementary Information The online version contains supplementary material available at 10.1186/s40164-022-00326-6.
Collapse
Affiliation(s)
- Rui Li
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China
| | - Mengying Ke
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China
| | - Mingming Qi
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenru Han
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China
| | - Yuhao Cao
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China
| | - Zhendong Deng
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China
| | - Jinjun Qian
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China.
| | - Ye Yang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China. .,School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China.
| |
Collapse
|
21
|
Chen C, Li Y, Miao P, Xu Y, Xie Y, Chen Z, Qian S. Tumor immune cell infiltration score based model predicts prognosis in multiple myeloma. Sci Rep 2022; 12:17082. [PMID: 36224246 PMCID: PMC9556830 DOI: 10.1038/s41598-022-21763-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/30/2022] [Indexed: 01/10/2023] Open
Abstract
The tumor microenvironment plays an important role in various processes, including tumorigenesis, cancer progression, and metastasis. Immune signatures have been identified and verified for use in diagnosis and prognosis prediction. We used single-sample Gene Set Enrichment Analysis to evaluate tumor immune cell infiltration score (TIICs) and verify their prognostic significance in both training and validation cohorts and using this information to build a prognostic model. A total of 1281 samples were obtained for further evaluation of the immune enrichment scores of 28 immune cells, showing that Th17 cell contributed most significantly to survival. Using the median TIICs as a cutoff to divide the samples into two groups, we found that the high-TIICs group was associated with favorable outcomes in both the training and validation sets. We then constructed a prognostic model to predict the 6, 8, and 10-year survival outcomes. Further analysis showed that immune score and tumor purity were higher in the high-TIICs group, while the matrix score was lower in this group. Forty-two differentially expressed genes were identified between the two groups. This new prognostic model based on immune cell infiltration indicates the potential for TIICs in predicting prognosis and as targets for treatment.
Collapse
Affiliation(s)
- Can Chen
- grid.13402.340000 0004 1759 700XDepartment of Hematology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Yiwei Li
- grid.13402.340000 0004 1759 700XDepartment of Critical Care Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Peiwen Miao
- grid.268505.c0000 0000 8744 8924Department of Hematology, Affiliated Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Ying Xu
- grid.13402.340000 0004 1759 700XDepartment of Hematology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Yaping Xie
- grid.13402.340000 0004 1759 700XDepartment of Hematology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Zhenzhen Chen
- grid.13402.340000 0004 1759 700XDepartment of Hematology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Shenxian Qian
- grid.13402.340000 0004 1759 700XDepartment of Hematology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, 216 Huansha Road, Hangzhou, 310006 Zhejiang China
| |
Collapse
|
22
|
Solimando AG, Da Vià MC, Bolli N, Steinbrunn T. The Route of the Malignant Plasma Cell in Its Survival Niche: Exploring “Multiple Myelomas”. Cancers (Basel) 2022; 14:cancers14133271. [PMID: 35805041 PMCID: PMC9265748 DOI: 10.3390/cancers14133271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Growing evidence points to multiple myeloma (MM) and its stromal microenvironment using several mechanisms to subvert effective immune and anti-tumor responses. Recent advances have uncovered the tumor-stromal cell influence in regulating the immune-microenvironment and have envisioned targeting these suppressive pathways to improve therapeutic outcomes. Nevertheless, some subgroups of patients include those with particularly unfavorable prognoses. Biological stratification can be used to categorize patient-, disease- or therapy-related factors, or alternatively, these biological determinants can be included in a dynamic model that customizes a given treatment to a specific patient. Genetic heterogeneity and current knowledge enforce a systematic and comprehensive bench-to-bedside approach. Given the increasing role of cancer stem cells (CSCs) in better characterizing the pathogenesis of solid and hematological malignancies, disease relapse, and drug resistance, identifying and describing CSCs is of paramount importance in the management of MM. Even though the function of CSCs is well-known in other cancer types, their role in MM remains elusive. With this review, we aim to provide an update on MM homing and resilience in the bone marrow micro milieu. These data are particularly interesting for clinicians facing unmet medical needs while designing novel treatment approaches for MM.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| | - Matteo Claudio Da Vià
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Torsten Steinbrunn
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| |
Collapse
|
23
|
Kim SJ, Kim S, Choi YJ, Kim UJ, Kang KW. CKD-581 Downregulates Wnt/β-Catenin Pathway by DACT3 Induction in Hematologic Malignancy. Biomol Ther (Seoul) 2022; 30:435-446. [PMID: 35794797 PMCID: PMC9424334 DOI: 10.4062/biomolther.2022.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 11/09/2022] Open
Abstract
The present study evaluated the anti-cancer activity of histone deacetylase (HDAC)-inhibiting CKD-581 in multiple myeloma (MM) and its pharmacological mechanisms. CKD-581 potently inhibited a broad spectrum of HDAC isozymes. It concentration-dependently inhibited proliferation of hematologic cancer cells including MM (MM.1S and RPMI8226) and T cell lymphoma (HH and MJ). It increased the expression of the dishevelled binding antagonist of β-catenin 3 (DACT3) in T cell lymphoma and MM cells, and decreased the expression of c-Myc and β-catenin in MM cells. Additionally, it enhanced phosphorylated p53, p21, cleaved caspase-3 and the subG1 population, and reversely, downregulated cyclin D1, CDK4 and the anti-apoptotic BCL-2 family. Finally, administration of CKD-581 exerted a significant anti-cancer activity in MM.1S-implanted xenografts. Overall, CKD-581 shows anti-cancer activity via inhibition of the Wnt/β-catenin signaling pathway in hematologic malignancies. This finding is evidence of the therapeutic potential and rationale of CKD-581 for treatment of MM.
Collapse
Affiliation(s)
- Soo Jin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, Yongin 16995, Republic of Korea
| | - Suntae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong June Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - U Ji Kim
- CKD Research Institution, Chong Kun Dang Pharmaceutical Corporation, Yongin 16995, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
24
|
Jo S, Hee Im S, Seo D, Ryu H, Hoon Kim S, Baek D, Baek A, Cho SR. Low-frequency repetitive magnetic stimulation suppresses neuroblastoma progression by downregulating the Wnt/β-catenin signaling pathway. Bioelectrochemistry 2022; 147:108205. [DOI: 10.1016/j.bioelechem.2022.108205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/31/2022] [Accepted: 07/05/2022] [Indexed: 11/02/2022]
|
25
|
Wu C. β-catenin inhibitors ICG-001 and pyrvinium sensitize bortezomib-resistant multiple myeloma cells to bortezomib. Oncol Lett 2022; 24:205. [PMID: 35720475 PMCID: PMC9178704 DOI: 10.3892/ol.2022.13326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 08/23/2021] [Indexed: 11/06/2022] Open
Abstract
Although bortezomib (BTZ) displays efficacy in treating multiple myeloma (MM), BTZ resistance in MM patients has been reported. Meanwhile, treating BTZ resistant MM cells with β-catenin inhibitors have demonstrated the ability to reserve BTZ resistance. Thus, the present study aimed to investigate the synergistic effect of the β-catenin inhibitors, ICG-001 and pyrvinium (PP), with BTZ in the treatment of BTZ-resistant MM cells. Different concentrations of ICG-001 (0–32 µM) or PP (0–32 nM) were used to treat the BTZ-resistant RPMI-8226 (RPMI-8226BR) and BTZ-resistant KMS-11 (KMS-11BR) cell lines, followed by a BTZ combination treatment. Subsequently, cell viability and apoptosis in these two cell lines were determined by CCK-8 assay and flow cytometry, respectively. The proteins involved in the Wnt/β-catenin signaling pathway were detected using western blotting. The Wnt/β-catenin signaling pathway was activated in the RPMI-8226BR and the KMS-11BR cells. In addition, the cell viability of RPMI-8226BR and KMS-11BR cells were decreased following β-catenin inhibitor (ICG-001 and PP) treatment alone. Furthermore, the β-catenin inhibitors, ICG-001 and PP, plus BTZ combination treatment revealed a notable decrease in cell viability and a marked increase in cell apoptosis rate, compared with that in cells treated with ICG-001, PP or BTZ alone in the RPMI-8226BR and KMS-11BR cell lines. In conclusion, the β-catenin inhibitors, ICG-001 and PP not only increased apoptosis, but also sensitized BTZ-resistant MM cells to BTZ, indicating their potential therapeutic application in MM.
Collapse
Affiliation(s)
- Cuicui Wu
- Department of Hematology, Yueyang Second People's Hospital, Yueyang, Hunan 414000, P.R. China
| |
Collapse
|
26
|
Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ 2022; 29:2190-2202. [PMID: 35534546 PMCID: PMC9613693 DOI: 10.1038/s41418-022-01008-w] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
The development of chemotherapy resistance is the most vital obstacle to clinical efficacy in gastric cancer (GC). The dysregulation of the Wnt/beta-catenin signaling pathway is critically associated with GC development and chemotherapy resistance. Ferroptosis is a form of regulated cell death, induced by an iron-dependent accumulation of lipid peroxides during chemotherapy. However, whether the Wnt/beta-catenin signaling directly controls resistance to cell death, remains unclear. Here, we show that the activation of the Wnt/beta-catenin signaling attenuates cellular lipid ROS production and subsequently inhibits ferroptosis in GC cells. The beta-catenin/TCF4 transcription complex directly binds to the promoter region of GPX4 and induces its expression, resulting in the suppression of ferroptotic cell death. Concordantly, TCF4 deficiency promotes cisplatin-induced ferroptosis in vitro and in vivo. Thus, we demonstrate that the aberrant activation of the Wnt/beta-catenin signaling confers ferroptosis resistance and suggests a potential therapeutic strategy to enhance chemo-sensitivity for advanced GC patients.
Collapse
|
27
|
Zhang X, Yang J, Chen S, Liu C, Wang Z, Ren H, Zhou L. Pre-existing hypertension is associated with poor progression-free survival in newly diagnosed multiple myeloma patients. J Thromb Thrombolysis 2022; 54:542-549. [PMID: 35445377 DOI: 10.1007/s11239-022-02653-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2022] [Indexed: 11/29/2022]
Abstract
Approximately 31% of patients diagnosed with multiple myeloma (MM) have pre-existing hypertension, but its effects on patient survival have not been investigated. We collected data from 228 newly diagnosed patients with MM and found that 71 (31.1%) had pre-existing hypertension. The impact of pre-existing hypertension on MM patients was determined by evaluating progression-free survival (PFS). Kaplan-Meier analyses revealed a significantly lower PFS in the pre-existing hypertension group than their non-hypertensive counterparts (median 22.6 vs 34.8 months, respectively). The multivariable Cox proportional hazards model showed that pre-existing hypertension was an independent risk factor for PFS reduction in MM patients. Moreover, the risk of disease progression in MM patients with pre-existing hypertension was higher than in non-hypertension comparator patients (hazard ratio 1.735, 95% confidence interval 1.261-2.387). In MM patients with pre-existing hypertension, Kaplan-Meier analyses found that those with a higher risk of hypertension had a significantly shorter PFS than those with lower risk (median 19.3 vs 25.4 months, respectively). However, multivariate Cox regression analysis showed that the risk stratification of hypertension was not an independent risk factor for poor PFS in MM patients with pre-existing hypertension. Our study demonstrates that pre-existing hypertension was significantly associated with a lower PFS in newly diagnosed MM patients.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Jieli Yang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Sai Chen
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Chang Liu
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Zhenhua Wang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Hefei Ren
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Lin Zhou
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
28
|
Targeting c-Myc Unbalances UPR towards Cell Death and Impairs DDR in Lymphoma and Multiple Myeloma Cells. Biomedicines 2022; 10:biomedicines10040731. [PMID: 35453482 PMCID: PMC9033049 DOI: 10.3390/biomedicines10040731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 01/11/2023] Open
Abstract
Multiple myeloma (MM) and primary effusion lymphoma (PEL) are aggressive hematological cancers, for which the search for new and more effective therapies is needed. Both cancers overexpress c-Myc and are highly dependent on this proto-oncogene for their survival. Although c-Myc inhibition has been shown to reduce PEL and MM survival, the underlying mechanisms leading to such an effect are not completely clarified. In this study, by pharmacologic inhibition and silencing, we show that c-Myc stands at the cross-road between UPR and DDR. Indeed, it plays a key role in maintaining the pro-survival function of UPR, through the IRE1α/XBP1 axis, and sustains the expression level of DDR molecules such as RAD51 and BRCA1 in MM and PEL cells. Moreover, we found that c-Myc establishes an interplay with the IRE1α/XBP1 axis whose inhibition downregulated c-Myc, skewed UPR towards cell death and enhanced DNA damage. In conclusion, this study unveils new insights into the molecular mechanisms leading to the cytotoxic effects of c-Myc inhibition and reinforces the idea that its targeting may be a promising therapeutic approach against MM and PEL that, although different cancers, share some similarities, including c-Myc overexpression, constitutive ER stress and poor response to current chemotherapies.
Collapse
|
29
|
Gau YC, Yeh TJ, Hsu CM, Hsiao SY, Hsiao HH. Pathogenesis and Treatment of Myeloma-Related Bone Disease. Int J Mol Sci 2022; 23:ijms23063112. [PMID: 35328533 PMCID: PMC8951013 DOI: 10.3390/ijms23063112] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma is a hematologic malignancy of plasma cells that causes bone-destructive lesions and associated skeletal-related events (SREs). The pathogenesis of myeloma-related bone disease (MBD) is the imbalance of the bone-remodeling process, which results from osteoclast activation, osteoblast suppression, and the immunosuppressed bone marrow microenvironment. Many important signaling cascades, including the RANKL/RANK/OPG axis, Notch signaling, the Wnt/β-Catenin signaling pathways, and signaling molecules, such as DKK-1, sclerostin, osteopontin, activin A, chemokines, and interleukins are involved and play critical roles in MBD. Currently, bisphosphonate and denosumab are the gold standard for MBD prevention and treatment. As the molecular mechanisms of MBD become increasingly well understood, novel agents are being thoroughly explored in both preclinical and clinical settings. Herein, we will provide an updated overview of the pathogenesis of MBD, summarize the clinical management and guidelines, and discuss novel bone-modifying therapies for further management of MBD.
Collapse
Affiliation(s)
- Yuh-Ching Gau
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (Y.-C.G.); (T.-J.Y.); (C.-M.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tsung-Jang Yeh
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (Y.-C.G.); (T.-J.Y.); (C.-M.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chin-Mu Hsu
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (Y.-C.G.); (T.-J.Y.); (C.-M.H.)
| | - Samuel Yien Hsiao
- Department of Biology, University of Rutgers-Camden, Camden, NJ 08102, USA;
| | - Hui-Hua Hsiao
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (Y.-C.G.); (T.-J.Y.); (C.-M.H.)
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +816-7-3162429
| |
Collapse
|
30
|
Combination of Histone Deacetylase Inhibitor Panobinostat (LBH589) with β-Catenin Inhibitor Tegavivint (BC2059) Exerts Significant Anti-Myeloma Activity Both In Vitro and In Vivo. Cancers (Basel) 2022; 14:cancers14030840. [PMID: 35159107 PMCID: PMC8834319 DOI: 10.3390/cancers14030840] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023] Open
Abstract
Over the last three decades changes in the treatment paradigm for newly diagnosed multiple myeloma (MM) have led to a significant increase in overall survival. Despite this, the majority of patients relapse after one or more lines of treatment while acquiring resistance to available therapies. Panobinostat, a pan-histone deacetylase inhibitor, was approved by the FDA in 2015 for patients with relapsed MM but how to incorporate panobinostat most effectively into everyday practice remains unclear. Dysregulation of the Wnt canonical pathway, and its key mediator β-catenin, has been shown to be important for the evolution of MM and the acquisition of drug resistance, making it a potentially attractive therapeutic target. Despite concerns regarding the safety of Wnt pathway inhibitors, we have recently shown that the β-catenin inhibitor Tegavivint is deliverable and effective in in vivo models of MM. In this study we show that the combination of low concentrations of panobinostat and Tegavivint have significant in vitro and in vivo anti-MM effects including in the context of proteasome inhibitor resistance, by targeting both aerobic glycolysis and mitochondrial respiration and the down-regulation of down-stream β-catenin targets including myc, cyclinD1, and cyclinD2. The significant anti-MM effect of this novel combination warrants further evaluation for the treatment of MM patients with relapsed and/or refractory MM.
Collapse
|
31
|
Zhang Y, Yu X, Sun R, Min J, Tang X, Lin Z, Xie S, Li X, Lu S, Tian Z, Gu C, Teng L, Yang Y. Splicing factor arginine/serine-rich 8 promotes multiple myeloma malignancy and bone lesion through alternative splicing of CACYBP and exosome-based cellular communication. Clin Transl Med 2022; 12:e684. [PMID: 35184390 PMCID: PMC8858635 DOI: 10.1002/ctm2.684] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a distinctive malignancy of plasma cell within the bone marrow (BM), of which alternative splicing factors play vital roles in the progression. Splicing factor arginine/serine-rich 8 (SFRS8) is the exclusive factor associated with MM prognosis, however its role in MM remains undefined. METHODS The analyses of 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide (MTT) assay, immunohistochemistry, flow cytometry and xenograft model were performed to examine cell proliferation, cell cycle and apoptosis in SFRS8 overexpression or knockdown MM cells in vitro and in vivo. The SFRS8-regulated alternative splicing events were identified by RNA immunoprecipitation sequencing (RIP-seq) and validated by RIP-qPCR and Co-IP methods. Exosomes were extracted from the supernatant of myeloma cells by ultracentrifugation. Bone lesion was evaluated by TRAP staining in vitro and SCID/NOD-TIBIA mouse model. A neon electroporation system was utilised to deliver siRNA through exosomes. The effect of siRNA-loaded exosomes in vivo was evaluated by using a patient-derived tumor xenograft (PDX) model and SCID/NOD-TIBIA mouse model. RESULTS SFRS8 was significantly upregulated in MM samples and positively associated with poor overall survival (OS) in MM patients. SFRS8 promoted MM cell proliferation in vitro and in vivo. Furthermore, calcyclin binding protein (CACYBP) was identified as the downstream target of SFRS8. Particularly, SFRS8 could reduce CACYBP isoform1 (NM_014412.3) and increase CACYBP isoform2 (NM_001007214.1) by mediating the alternative splicing of CACYBP, thereby altering the ubiquitination degradation of β-catenin to promote MM progression. In addition, SFRS8 promoted osteoclast differentiation through exosomes in vitro and in vivo. More importantly, exosomal siRNA targeting CACYBP isoform2 inhibited tumour growth in PDX and SCID/NOD-TIBIA mouse models. CONCLUSION Our findings demonstrate that targeting the SFRS8/CACYBP/β-catenin axis may be a promising strategy for MM diagnosis and treatment.
Collapse
Affiliation(s)
- Yuanjiao Zhang
- Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Xichao Yu
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Rongze Sun
- School of Life ScienceJilin UniversityChangchunChina
| | - Jie Min
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Xiaozhu Tang
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Zigen Lin
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Siyuan Xie
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Xinying Li
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Shengfeng Lu
- Key Laboratory of A cupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese MedicineNanjingChina
| | - Zhidan Tian
- Department of Pathology, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
| | - Lesheng Teng
- School of Life ScienceJilin UniversityChangchunChina
| | - Ye Yang
- School of Medicine & Holistic Integrative MedicineNanjing University of Chinese MedicineNanjingChina
- Key Laboratory of A cupuncture and Medicine Research of Ministry of EducationNanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
32
|
Yassin NYS, AbouZid SF, El-Kalaawy AM, Ali TM, Almehmadi MM, Ahmed OM. Silybum marianum total extract, silymarin and silibinin abate hepatocarcinogenesis and hepatocellular carcinoma growth via modulation of the HGF/c-Met, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. Biomed Pharmacother 2022; 145:112409. [PMID: 34781148 DOI: 10.1016/j.biopha.2021.112409] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has been identified as one of the most deadly malignancies with limited therapeutic efficacy worldwide. However, understanding the molecular mechanisms of crosstalk between signaling pathways in HCC and predicting cancer cell responses to targeted therapeutic interventions remain to be challenge. Thus, in this study, we aimed to evaluate the anticancerous efficacy of Silybum marianum total extract (STE), silymarin (Sm), and silibinin (Sb) against experimentally-induced HCC in rats. In vitro investigations were also performed and the anticancer effects against HCC cell lines (HepG2 and Huh7) were confirmed. Wistar rats were given diethylnitrosamine (DEN)/2-acetylaminofluorene (AAF)/carbon tetrachloride (CCl4) and were orally treated with STE (200 mg/kg body weight (bw)), Sm (150 mg/kg bw), and Sb (5 mg/kg bw) every other day from the 1st or 16th week to the 25th week of DEN/AAF/CCl4 injection. Treatment with STE, Sm, and Sb inhibited the growth of cancerous lesions in DEN/AAF/CCl4-treated rats. This inhibition was associated with inhibition of Ki-67 expression and repression of HGF/cMet, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. STE, Sm, and Sb improved liver function biomarkers and tumor markers (AFP, CEA, and CA19.9) and increased total protein and albumin levels in serum. STE, Sm, and Sb treatment was also noted to reduce the hepatic production of lipid peroxides, increase hepatic glutathione content, and induce the activities of hepatic antioxidant enzymes in DEN/AAF/CCl4-treated rats. These results indicate that STE, Sm, and Sb exert anti-HCC effects through multiple pathways, including suppression of Ki-67 expression and HGF/cMet, Wnt/β-catenin, and PI3K/Akt/mTOR pathways and enhancement of antioxidant defense mechanisms.
Collapse
Affiliation(s)
- Nour Y S Yassin
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sameh F AbouZid
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt
| | - Asmaa M El-Kalaawy
- Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Tarek M Ali
- Department of Physiology, College of Medicine, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia
| | - Mazen M Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia
| | - Osama M Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt.
| |
Collapse
|
33
|
Wang HN, Yang J, Xie DH, Liang Z, Wang Y, Fu RY, Liu X, Xia ZJ, Jia G, Wang L. Single-cell RNA sequencing infers the role of malignant cells in drug-resistant multiple myeloma. Clin Transl Med 2021; 11:e653. [PMID: 34918874 PMCID: PMC8678945 DOI: 10.1002/ctm2.653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- He-Nan Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jing Yang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - De-Huan Xie
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhigang Liang
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Yang Wang
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otorinolaringology, Beijing, China
| | - Rui-Ying Fu
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xindi Liu
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhong-Jun Xia
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guangshuai Jia
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
34
|
Song GR, Choi YJ, Park SJ, Shin S, Lee G, Choi HJ, Lee DY, Song GY, Oh S. Root Bark of Morus alba L. and Its Bioactive Ingredient, Ursolic Acid, Suppress the Proliferation of Multiple Myeloma Cells by Inhibiting Wnt/β-Catenin Pathway. J Microbiol Biotechnol 2021; 31:1559-1567. [PMID: 34584036 PMCID: PMC9706038 DOI: 10.4014/jmb.2109.09002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022]
Abstract
The root bark of Morus alba L. has cytotoxic activity against several types of cancer cells. However, little is known about its chemopreventive mechanisms and bioactive metabolites. In this study, we showed that M. alba L. root bark extracts (MRBE) suppressed β-catenin response transcription (CRT), which is aberrantly activated in various cancers, by promoting the degradation of β-catenin. In addition, MRBE repressed the expression of the β-catenin/T-cell factor (TCF)-dependent genes, cmyc and cyclin D1, thus inhibiting the proliferation of RPMI-8226 multiple myeloma (MM) cells. MRBE induced apoptosis in MM cells, as evidenced by the increase in the population of annexin VFITC- positive cells and caspase-3/7 activity. We identified ursolic acid in MRBE through LC/mass spectrum (MS) and observed that it also decreased intracellular β-catenin, c-myc, and cyclin D1 levels. Furthermore, it suppressed the proliferation of RPMI-8226 cells by stimulating cell cycle arrest and apoptosis. These findings suggest that MRBE and its active ingredient, ursolic acid, exert antiproliferative activity by promoting the degradation of β-catenin and may have significant chemopreventive potential against MM.
Collapse
Affiliation(s)
- Geu Rim Song
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
- Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea
| | - Yoon Jung Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Soo Jin Park
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Subeen Shin
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
- Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea
| | - Giseong Lee
- College of General Education, Kookmin University, Seoul 02707, Republic of Korea
| | - Hui Ji Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Gyu-Yong Song
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sangtaek Oh
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
- Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
35
|
What are the roles of global DNA and APC 2 gene promotor hypermethylation in multiple myeloma? Mol Biol Rep 2021; 48:7875-7882. [PMID: 34637096 PMCID: PMC8505470 DOI: 10.1007/s11033-021-06813-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022]
Abstract
Background In today's practice, gene-based approaches come to the fore in the determination of prognosis and treatment preferences of multiple myeloma (MM). DNA methylation is one of the new approach parameters. DNA methylation occurs by the addition of a methyl group to cytosines in CpG dinucleotides. In this study, besides comparing the global DNA and APC 2 gene promotor hypermethylation between our patients with MM and healthy control group, we aimed to demonstrate the effect of hypermethylation on MM treatment responses and survival. Methods and results 38 patients diagnosed with MM between January 2016 and January 2020 and 50 healthy controls were included in the study. The initial hypermethylation of the patients and the healthy control group were statistically analyzed. In addition, the increase in hypermethylation in the MM group before and after the first series of treatments were analyzed within themselves. There is a significant difference between the patients with MM diagnosis and the healthy control group in terms of the initial global hypermethylation (P = 0.001). In patients with MM, hypermethylation was significantly higher. Global hypermethylation in the post-treatment measurements was significantly increased in comparison to the pre-treatment state (P = 0.012). In terms of APC 2 promotor gene-specific hypermethylation, no significant differences were detected between pre- and post-treatment values (P = 0.368). Conclusions This study represents valuable data with the initial global DNA hypermethylation results in the MM patient group and the increase in hypermethylation post-treatment. it will shed light on future studies.
Collapse
|
36
|
Visconti RJ, Kolaja K, Cottrell JA. A functional three-dimensional microphysiological human model of myeloma bone disease. J Bone Miner Res 2021; 36:1914-1930. [PMID: 34173283 DOI: 10.1002/jbmr.4404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022]
Abstract
Human myeloma bone disease (MBD) occurs when malignant plasma cells migrate to the bone marrow and commence inimical interactions with stromal cells, disrupting the skeletal remodeling process. The myeloma cells simultaneously suppress osteoblastic bone formation while promoting excessive osteoclastic resorption. This bone metabolism imbalance produces osteolytic lesions that cause chronic bone pain and reduce trabecular and cortical bone structural integrity, and often culminate in pathological fractures. Few bone models exist that enable scientists to study MBD and the effect therapies have on restoring the bone metabolism imbalance. The purpose of this research was to develop a well characterized three-dimensional (3D) bone organoid that could be used to study MBD and current or potential treatment options. First, bone marrow stromal cell-derived osteoblasts (OBs) mineralized an endosteal-like extracellular matrix (ECM) over 21 days. Multiple analyses confirmed the generation of hydroxyapatite (HA)-rich bone-like tissue fragments that were abundant in alkaline phosphatase, calcium, and markers of osteoblastic gene expression. On day 22, bone marrow macrophage (BMM)-derived osteoclasts (OCs) were introduced to enhance the resorptive capability of the model and recapitulate the balanced homeostatic nature of skeletal remodeling. Tartrate-resistant acid phosphatase 5b (TRAcP-5b), type I collagen C-telopeptide (CTX-1), and gene expression analysis confirmed OC activity in the normal 3D organoid (3D in vitro model of normal bonelike fragments [3D-NBF]). On day 30, a human multiple myeloma (MM)-derived plasmacytoma cell line was introduced to the 3D-NBF to generate the 3D-myeloma bone disease organoid (3D-MBD). After 12 days, the 3D-MBD had significantly reduced total HA, increased TRAcP-5b levels, increases levels of CTX-1, and decreased expression of osteoblastic genes. Therapeutic intervention with pharmaceutical agents including an immunomodulatory drug, a bisphosphonate, and monoclonal restored HA content and reduced free CTX-1 in a dose-dependent manner. This osteogenically functional model of MBD provides a novel tool to study biological mechanisms guiding the disease and to screen potential therapeutics. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Richard J Visconti
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, USA.,Investigative Toxicology, Nonclinical Research and Development, Bristol Myers Squibb, Summit, New Jersey, USA
| | - Kyle Kolaja
- Investigative Toxicology, Nonclinical Research and Development, Bristol Myers Squibb, Summit, New Jersey, USA
| | - Jessica A Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, New Jersey, USA
| |
Collapse
|
37
|
Neiheisel A, Kaur M, Ma N, Havard P, Shenoy AK. Wnt pathway modulators in cancer therapeutics: An update on completed and ongoing clinical trials. Int J Cancer 2021; 150:727-740. [PMID: 34536299 DOI: 10.1002/ijc.33811] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/31/2021] [Accepted: 09/06/2021] [Indexed: 01/17/2023]
Abstract
Wnt signaling plays an essential role in the initiation and progression of various types of cancer. Besides, the Wnt pathway components have been established as reliable biomarkers and potential targets for cancer therapy. Wnt signaling is categorized into canonical and noncanonical pathways. The canonical pathway is involved in cell survival, proliferation, differentiation and migration, while the noncanonical pathway regulates cell polarity and migration. Apart from its biological role in development and homeostasis, the Wnt pathway has been implicated in several pathological disorders, including cancer. As a result, inhibiting this pathway has been a focus of cancer research with multiple targetable candidates in development. In this review, our focus will be to summarize information about ongoing and completed clinical trials targeting various Wnt pathway components, along with describing current and emerging Wnt targeted therapies. In addition, we will discuss potential opportunities and associated challenges of inhibiting Wnt signaling for cancer therapy.
Collapse
Affiliation(s)
- Ann Neiheisel
- College of Pharmacy, California Health Sciences University, Clovis, California, USA
| | - Manpreet Kaur
- College of Pharmacy, California Health Sciences University, Clovis, California, USA
| | - Nancy Ma
- College of Pharmacy, California Health Sciences University, Clovis, California, USA
| | - Patty Havard
- Kaweah Health Foundation, Kaweah Health, Visalia, California, USA
| | - Anitha K Shenoy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Health Sciences University, Clovis, California, USA
| |
Collapse
|
38
|
Andrews RE, Brown JE, Lawson MA, Chantry AD. Myeloma Bone Disease: The Osteoblast in the Spotlight. J Clin Med 2021; 10:jcm10173973. [PMID: 34501423 PMCID: PMC8432062 DOI: 10.3390/jcm10173973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/17/2022] Open
Abstract
Lytic bone disease remains a life-altering complication of multiple myeloma, with up to 90% of sufferers experiencing skeletal events at some point in their cancer journey. This tumour-induced bone disease is driven by an upregulation of bone resorption (via increased osteoclast (OC) activity) and a downregulation of bone formation (via reduced osteoblast (OB) activity), leading to phenotypic osteolysis. Treatments are limited, and currently exclusively target OCs. Despite existing bone targeting therapies, patients successfully achieving remission from their cancer can still be left with chronic pain, poor mobility, and reduced quality of life as a result of bone disease. As such, the field is desperately in need of new and improved bone-modulating therapeutic agents. One such option is the use of bone anabolics, drugs that are gaining traction in the osteoporosis field following successful clinical trials. The prospect of using these therapies in relation to myeloma is an attractive option, as they aim to stimulate OBs, as opposed to existing therapeutics that do little to orchestrate new bone formation. The preclinical application of bone anabolics in myeloma mouse models has demonstrated positive outcomes for bone repair and fracture resistance. Here, we review the role of the OB in the pathophysiology of myeloma-induced bone disease and explore whether novel OB targeted therapies could improve outcomes for patients.
Collapse
Affiliation(s)
- Rebecca E. Andrews
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
- Correspondence:
| | - Janet E. Brown
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| | - Michelle A. Lawson
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
| | - Andrew D. Chantry
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| |
Collapse
|
39
|
Wnt signaling: A prospective therapeutic target for chronic pain. Pharmacol Ther 2021; 231:107984. [PMID: 34480969 DOI: 10.1016/j.pharmthera.2021.107984] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 02/08/2023]
Abstract
Despite the rapid advance over the past decades to design effective therapeutic pharmacological interventions, chronic pain remains to be an unresolved healthcare concern. Long term use of opioids, the first line analgesics, often causes detrimental side effects. Therefore, a profound understanding of the mechanisms underlying the development and maintenance of chronic pain states is urgently needed for the management of chronic pain. Substantial evidence indicates aberrant activation of Wnt signaling pathways in sciatic nerve, dorsal root ganglia and spinal cord dorsal horn in rodent models of chronic pain. Moreover, growing evidence shows that pharmacological blockage of aberrant activation of Wnt signaling pathways attenuates pain behaviors in animal models of chronic pain. Importantly, both intrathecal injection of Wnt agonists and Wnt ligands to naïve rats lead to the development of mechanical allodynia, which was inhibited by Wnt inhibitors. In this review, we summarized and discussed the therapeutic potential of pharmacological inhibitors of Wnt signaling in chronic pain in preclinical studies. These evidence showed that aberrant activation of Wnt signaling pathways contributed to chronic pain via enhancing neuroinflammation, regulating synaptic plasticity and reducing intraepidermal nerve fiber density. However, these findings raise further questions. Overall, despite the future challenges, these pioneering studies suggest that Wnt signaling is a promising therapeutic target for chronic pain.
Collapse
|
40
|
Song S, Fan G, Li Q, Su Q, Zhang X, Xue X, Wang Z, Qian C, Jin Z, Li B, Zhuang W. IDH2 contributes to tumorigenesis and poor prognosis by regulating m6A RNA methylation in multiple myeloma. Oncogene 2021; 40:5393-5402. [PMID: 34274946 DOI: 10.1038/s41388-021-01939-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 06/12/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
Epigenetic alterations have been previously shown to contribute to multiple myeloma (MM) pathogenesis via DNA methylations and histone modifications. RNA methylation, a novel epigenetic modification, is required for cancer cell survival, and targeting this pathway has been proposed as a new therapeutic strategy. The extent to the N6-methyladenosine (m6A)-regulatory pathway functions in MM remains unknown. Here, we show that an imbalance of RNA methylation may underlies the tumorigenesis of MM. Mechanistically, isocitrate dehydrogenase 2 (IDH2) is highly expressed in CD138+ cells from MM and its levels appear a progressive increase in the progression of plasma cell dyscrasias. Downregulation of IDH2 increases global m6A RNA levels and reduces myeloma cell growth in vitro, decreases the burden of disease and prolongs overall survival in vivo. IDH2 regulates RNA methylation by activating the RNA demethylase FTO, which is an α-KG-dependent dioxygenase. Furthermore, IDH2-mediated FTO activation decreases the m6A level on WNT7B transcripts, then increases WNT7B expression and thus activated Wnt signaling pathway. Moreover, survival analysis indicates that the elevated expression of IDH2 predicts a poor prognosis. Higher expression of FTO is related to higher International Staging System (ISS) stage and higher Revised-ISS (R-ISS) stage of MM. Collectively, our studies reveal that IDH2 regulates global m6A RNA modification in MM via targeting RNA demethylases FTO. The imbalance of m6A methylation activates the Wnt signaling pathway by enhancing the WNT7B expression, and thus promoting tumorigenesis and progression of MM. IDH2 might be used as a therapeutic target and a possible prognostic factor for MM.
Collapse
Affiliation(s)
- Sha Song
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Gao Fan
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qi Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Su
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xinyun Zhang
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaofeng Xue
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiming Wang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Chen'ao Qian
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhou Jin
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Bingzong Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wenzhuo Zhuang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
41
|
Zhou F, Wang D, Zhou N, Chen H, Shi H, Peng R, Wei W, Wu L. Circular RNA Protein Tyrosine Kinase 2 Promotes Cell Proliferation, Migration and Suppresses Apoptosis via Activating MicroRNA-638 Mediated MEK/ERK, WNT/β-Catenin Signaling Pathways in Multiple Myeloma. Front Oncol 2021; 11:648189. [PMID: 34395238 PMCID: PMC8355695 DOI: 10.3389/fonc.2021.648189] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
Our previous study observed that circular RNA protein tyrosine kinase 2 (circ-PTK2) was upregulated and correlated with worse clinical features and unfavorable prognosis in multiple myeloma (MM) patients. Thus, this study aimed to further characterize the regulatory function of circ-PTK2 on cell malignant activities and its target microRNA-638 (miR-638) as well as downstream MEK/ERK, WNT/β-catenin signaling pathways in MM. The effect of circ-PTK2 on MM cell proliferation, apoptosis, migration, invasion and its potential target miRNAs was assessed by transfecting circ-PTK2 overexpression plasmids into U226 cells and circ-PTK2 knock-down plasmids into LP-1 cells. Furthermore, the interaction between circ-PTK2 and miR-638 mediated MEK/ERK and WNT/β-catenin signaling pathways was validated by rescue experiments. Circ-PTK2 was overexpressed in most MM cell lines compared to normal plasma cells. Overexpressing circ-PTK2 promoted proliferation and migration, inhibited apoptosis in U266 cells, but did not affect cell invasion; knocking down circ-PTK2 achieved opposite effect in LP-1 cells. Besides, circ-PTK2 reversely regulated miR-638 expression but not miR-4690, miR-6724, miR-6749 or miR-6775. The following luciferase reporter assay illustrated the direct bind of circ-PTK2 towards miR-638. In rescue experiments, overexpressing miR-638 suppressed proliferation, migration, while promoted apoptosis in both wild U266 cells and circ-PTK2-overexpressed U266 cells; meanwhile, overexpressing miR-638 also suppressed MEK/ERK and WNT/β-catenin pathways in both wild U266 cells and circ-PTK2-overexpressed U266 cells. Knocking down miR-638 achieved opposite effect in both wild LP-1 cells and circ-PTK2-knocked-down LP-1 cells. In conclusion, circ-PTK2 promotes cell proliferation, migration, suppresses cell apoptosis via miR-638 mediated MEK&ERK and WNT&β-catenin signaling pathways in MM.
Collapse
Affiliation(s)
- Fan Zhou
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Dongjiao Wang
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Nian Zhou
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Haimin Chen
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Haotian Shi
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Rong Peng
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Wei Wei
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| | - Lixia Wu
- Department of Hematology and Oncology, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, China
| |
Collapse
|
42
|
Guo W, Wang H, Chen P, Shen X, Zhang B, Liu J, Peng H, Xiao X. Identification and Characterization of Multiple Myeloma Stem Cell-Like Cells. Cancers (Basel) 2021; 13:cancers13143523. [PMID: 34298738 PMCID: PMC8306148 DOI: 10.3390/cancers13143523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/30/2021] [Accepted: 07/04/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell tumor of the blood system with high incidence and poor prognosis. With a further understanding of the pathogenesis of MM and the bone marrow microenvironment, a variety of adjuvant cell therapies and new drugs have been developed. However, the drug resistance and high relapse rate of MM have not been fundamentally resolved. Studies have shown that, in patients with MM, there is a type of poorly differentiated progenitor cell (MM stem cell-like cells, MMSCs). Although there is no recognized standard for identification and classification, it is confirmed that they are closely related to the drug resistance and relapse of MM. This article therefore systematically summarizes the latest developments in MMSCs with possible markers of MMSCs, introduces the mechanism of how MMSCs work in MM resistance and recurrence, and discusses the active pathways that related to stemness of MM.
Collapse
Affiliation(s)
- Wancheng Guo
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (H.W.); (J.L.)
- Xiangya Medical School, Central South University, Changsha 410013, China; (P.C.); (X.S.); (B.Z.)
| | - Haiqin Wang
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (H.W.); (J.L.)
| | - Peng Chen
- Xiangya Medical School, Central South University, Changsha 410013, China; (P.C.); (X.S.); (B.Z.)
| | - Xiaokai Shen
- Xiangya Medical School, Central South University, Changsha 410013, China; (P.C.); (X.S.); (B.Z.)
| | - Boxin Zhang
- Xiangya Medical School, Central South University, Changsha 410013, China; (P.C.); (X.S.); (B.Z.)
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (H.W.); (J.L.)
| | - Hongling Peng
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (H.W.); (J.L.)
- Correspondence: (H.P.); (X.X.); Tel.: +86-731-85295296 (H.P.); +86-731-84805449 (X.X.)
| | - Xiaojuan Xiao
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (W.G.); (H.W.); (J.L.)
- Correspondence: (H.P.); (X.X.); Tel.: +86-731-85295296 (H.P.); +86-731-84805449 (X.X.)
| |
Collapse
|
43
|
Lu H, Pundole X, Lee HC. The role of bone-modifying agents in myeloma bone disease. JBMR Plus 2021; 5:e10518. [PMID: 34368608 PMCID: PMC8328802 DOI: 10.1002/jbm4.10518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 01/23/2023] Open
Abstract
Bone disease is common in patients with multiple myeloma (MM), which manifests as bone pain and skeletal-related events (SREs) such as pathological fractures and spinal cord compression. Myeloma bone disease (MBD) can adversely affect the quality of life of patients and have negative effects on morbidity and mortality. The pathogenesis of MBD is complex, and several factors are involved in the dysregulation of bone metabolism and uncoupling of bone remodeling, which result in net bone loss and devastating SREs. Broadly speaking, elevated osteoclast activity, suppressed osteoblast activity, and an aberrant marrow microenvironment play a role in MBD. Interaction of MM cells with the main bone cell osteocytes also promote further bone destruction. This review focuses on the role of bone-modifying agents in the prevention and treatment of MBD. The mainstay of MBD prevention are antiresorptive agents, bisphosphonates and denosumab. However, these agents do not play a direct role in bone formation and repair of existing MBD. Newer agents with anabolic effects such as anti-sclerostin antibodies, parathyroid hormone, anti-Dickkopf-1 antibodies, and others have shown potential in repair of MBD lesions. With the development of several new agents, the treatment landscape of MBD is likely to evolve in the coming years. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Huifang Lu
- Department of General Internal Medicine Section of Rheumatology and Clinical Immunology Houston Texas USA
| | - Xerxes Pundole
- Department of Health Services Research The University of Texas MD Anderson Cancer Center Houston Texas USA.,Present address: Amgen Inc. Thousand Oaks CA USA
| | - Hans C Lee
- Department of Lymphoma/Myeloma The University of Texas MD Anderson Cancer Center Houston Texas USA
| |
Collapse
|
44
|
Cotoraci C, Ciceu A, Sasu A, Miutescu E, Hermenean A. Bioactive Compounds from Herbal Medicine Targeting Multiple Myeloma. APPLIED SCIENCES 2021; 11:4451. [DOI: 10.3390/app11104451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Multiple myeloma (MM) is one of the most widespread hematological cancers. It is characterized by a clonal proliferation of malignant plasma cells in the bone marrow and by the overproduction of monoclonal proteins. In recent years, the survival rate of patients with multiple myeloma has increased significantly due to the use of transplanted stem cells and of the new therapeutic agents that have significantly increased the survival rate, but it still cannot be completely cured and therefore the development of new therapeutic products is needed. Moreover, many patients have various side effects and face the development of drug resistance to current therapies. The purpose of this review is to highlight the bioactive active compounds (flavonoids) and herbal extracts which target dysregulated signaling pathway in MM, assessed by in vitro and in vivo experiments or clinical studies, in order to explore their healing potential targeting multiple myeloma. Mechanistically, they demonstrated the ability to promote cell cycle blockage and apoptosis or autophagy in cancer cells, as well as inhibition of proliferation/migration/tumor progression, inhibition of angiogenesis in the tumor vascular network. Current research provides valuable new information about the ability of flavonoids to enhance the apoptotic effects of antineoplastic drugs, thus providing viable therapeutic options based on combining conventional and non-conventional therapies in MM therapeutic protocols.
Collapse
Affiliation(s)
- Coralia Cotoraci
- Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Alina Ciceu
- “Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Alciona Sasu
- Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Eftimie Miutescu
- Department of Gastroenterology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
- Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
| |
Collapse
|
45
|
Kumar V, Vashishta M, Kong L, Wu X, Lu JJ, Guha C, Dwarakanath BS. The Role of Notch, Hedgehog, and Wnt Signaling Pathways in the Resistance of Tumors to Anticancer Therapies. Front Cell Dev Biol 2021; 9:650772. [PMID: 33968932 PMCID: PMC8100510 DOI: 10.3389/fcell.2021.650772] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
Resistance to therapy is the major hurdle in the current cancer management. Cancer cells often rewire their cellular process to alternate mechanisms to resist the deleterious effect mounted by different therapeutic approaches. The major signaling pathways involved in the developmental process, such as Notch, Hedgehog, and Wnt, play a vital role in development, tumorigenesis, and also in the resistance to the various anticancer therapies. Understanding how cancer utilizes these developmental pathways in acquiring the resistance to the multi-therapeutic approach cancer can give rise to a new insight of the anti-therapy resistance mechanisms, which can be explored for the development of a novel therapeutic approach. We present a brief overview of Notch, Hedgehog, and Wnt signaling pathways in cancer and its role in providing resistance to various cancer treatment modalities such as chemotherapy, radiotherapy, molecular targeted therapy, and immunotherapy. Understanding the importance of these molecular networks will provide a rational basis for novel and safer combined anticancer therapeutic approaches for the improvement of cancer treatment by overcoming drug resistance.
Collapse
Affiliation(s)
- Vivek Kumar
- R&D Dept, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Mohit Vashishta
- R&D Dept, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Lin Kong
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Xiaodong Wu
- R&D Dept, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Jiade J Lu
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Chandan Guha
- Albert Einstein College of Medicine, The Bronx, NY, United States
| | - B S Dwarakanath
- R&D Dept, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| |
Collapse
|
46
|
Multiple Myeloma Bone Disease: Implication of MicroRNAs in Its Molecular Background. Int J Mol Sci 2021; 22:ijms22052375. [PMID: 33673480 PMCID: PMC7956742 DOI: 10.3390/ijms22052375] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a common hematological malignancy arising from terminally differentiated plasma cells. In the majority of cases, symptomatic disease is characterized by the presence of bone disease. Multiple myeloma bone disease (MMBD) is a result of an imbalance in the bone-remodeling process that leads to increased osteoclast activity and decreased osteoblast activity. The molecular background of MMBD appears intriguingly complex, as several signaling pathways and cell-to-cell interactions are implicated in the pathophysiology of MMBD. MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate the expression of their target mRNAs. Numerous miRNAs have been witnessed to be involved in cancer and hematological malignancies and their role has been characterized either as oncogenic or oncosuppressive. Recently, scientific research turned towards miRNAs as regulators of MMBD. Scientific data support that miRNAs finely regulate the majority of the signaling pathways implicated in MMBD. In this review, we provide concise information regarding the molecular pathways with a significant role in MMBD and the miRNAs implicated in their regulation. Moreover, we discuss their utility as molecular biomarkers and highlight the putative usage of miRNAs as novel molecular targets for targeted therapy in MMBD.
Collapse
|
47
|
In Vitro Investigation of the Cytotoxic Activity of Emodin 35 Derivative on Multiple Myeloma Cell Lines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6682787. [PMID: 33564319 PMCID: PMC7850823 DOI: 10.1155/2021/6682787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 11/24/2022]
Abstract
Background Bortezomib is used for treating multiple myeloma (MM); however, it has considerable adverse effects. Emodin has been reported to exhibit inhibitory effects on MM cell lines. We investigated the efficacy of emodin 35 (E35), an emodin derivative, using U266 and MM1s cell lines in treating MM and the efficacy of combining bortezomib and E35. Methods MTT assays were used to observe the effects of E35 on MM cell growth. The effects on cellular apoptosis were then observed using Annexin V/propidium iodide (PI) staining assay. The expression of apoptosis-related genes, including the caspase family, was examined. The efficacy of combining bortezomib and E35 was investigated by examining the expression of the Akt/mTOR/4EBP1 signaling pathway-related proteins. Results We report that E35 inhibited the growth of U266 and MM1s cells by inducing cellular apoptosis. Moreover, E35 downregulated the expression of apoptosis-related genes and suppressed the phosphorylation of Akt/mTOR/4EBP1 signaling pathway-related genes, thus exhibiting synergistic effects with bortezomib. All observed effects were dose-dependent. Conclusion The results showed that E35 exhibited cytotoxic effects in MM cell lines in protein levels. Thus, E35, particularly in combination with bortezomib, may be considered as a promising treatment for MM; however, this requires further investigation in vivo.
Collapse
|
48
|
Ding T, Deng R, Huang T. Long non-coding RNA T cell factor 7 is associated with increased disease risk and poor prognosis, and promotes cell proliferation, attenuates cell apoptosis and miR-200c expression in multiple myeloma. Oncol Lett 2021; 21:129. [PMID: 33552250 PMCID: PMC7798040 DOI: 10.3892/ol.2020.12390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to investigate the association of long non-coding RNA T cell factor 7 (lncRNA TCF7) with disease risk, prognosis and its cellular function in multiple myeloma (MM). A total of 132 de novo symptomatic patients with MM and 50 controls were enrolled. Plasma cells from patients with MM and controls were separated from bone marrow samples to detect lncRNA TCF7 expression using reverse transcription-quantitative PCR. In addition, treatment responses, event-free survival (EFS) and overall survival (OS) were measured. The effects of lncRNA TCF7 on proliferation, apoptosis and microRNA-200c (miR-200c) expression were assessed by gain- and loss-of-function experiments in RPMI-8226 and U-266 cells. The results demonstrated that lncRNA TCF7 expression was upregulated in patients with MM compared with controls, and the receiver operating characteristic curve revealed that lncRNA TCF7 could distinguish patients with MM from controls with an area under the curve of 0.793 (95% CI, 0.725-0.861). In patients with MM, high lncRNA TCF7 expression was associated with higher β2-microglobulin, more advanced International Staging System stage and increased t (14; 16) mutations. Furthermore, it was demonstrated that lncRNA TCF7 was downregulated in patients with complete response (CR) compared with patients without CR. Furthermore, high lncRNA TCF7 expression predicted worse EFS and OS. lncRNA TCF7 also promoted cell proliferation, whereas it reduced cell apoptosis and miR-200c expression in RPMI-8226 and U-266 cells. In conclusion, the present results suggested that lncRNA TCF7 may be used as a potential biomarker and as a treatment target for MM.
Collapse
Affiliation(s)
- Tianling Ding
- Department of Hematology, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Ruoyu Deng
- Shanghai Qeejen Bio-tech Institution, Shanghai 200434, P.R. China
| | - Ting Huang
- Department of Oncology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
49
|
Signaling Pathway Mediating Myeloma Cell Growth and Survival. Cancers (Basel) 2021; 13:cancers13020216. [PMID: 33435632 PMCID: PMC7827005 DOI: 10.3390/cancers13020216] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The bone marrow (BM) microenvironment plays a crucial role in pathogenesis of multiple myeloma (MM), and delineation of the intracellular signaling pathways activated in the BM microenvironment in MM cells is essential to develop novel therapeutic strategies to improve patient outcome. Abstract The multiple myeloma (MM) bone marrow (BM) microenvironment consists of different types of accessory cells. Both soluble factors (i.e., cytokines) secreted from these cells and adhesion of MM cells to these cells play crucial roles in activation of intracellular signaling pathways mediating MM cell growth, survival, migration, and drug resistance. Importantly, there is crosstalk between the signaling pathways, increasing the complexity of signal transduction networks in MM cells in the BM microenvironment, highlighting the requirement for combination treatment strategies to blocking multiple signaling pathways.
Collapse
|
50
|
Zhang Y, Wang X. Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol 2020; 13:165. [PMID: 33276800 PMCID: PMC7716495 DOI: 10.1186/s13045-020-00990-3] [Citation(s) in RCA: 757] [Impact Index Per Article: 151.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
The aberrant Wnt/β-catenin signaling pathway facilitates cancer stem cell renewal, cell proliferation and differentiation, thus exerting crucial roles in tumorigenesis and therapy response. Accumulated investigations highlight the therapeutic potential of agents targeting Wnt/β-catenin signaling in cancer. Wnt ligand/ receptor interface, β-catenin destruction complex and TCF/β-catenin transcription complex are key components of the cascade and have been targeted with interventions in preclinical and clinical evaluations. This scoping review aims at outlining the latest progress on the current approaches and perspectives of Wnt/β-catenin signaling pathway targeted therapy in various cancer types. Better understanding of the updates on the inhibitors, antagonists and activators of Wnt/β-catenin pathway rationalizes innovative strategies for personalized cancer treatment. Further investigations are warranted to confirm precise and secure targeted agents and achieve optimal use with clinical benefits in malignant diseases.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.,School of medicine, Shandong University, Jinan, 250021, Shandong, China.,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,School of medicine, Shandong University, Jinan, 250021, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 250021, China.
| |
Collapse
|