1
|
da Silva Fernandes T, Gillard BM, Dai T, Martin JC, Chaudhry KA, Dugas SM, Fisher AA, Sharma P, Wu R, Attwood KM, Dasgupta S, Takabe K, Rosario SR, Bianchi-Smiraglia A. Inosine monophosphate dehydrogenase 2 (IMPDH2) modulates response to therapy and chemo-resistance in triple negative breast cancer. Sci Rep 2025; 15:1061. [PMID: 39774345 PMCID: PMC11707137 DOI: 10.1038/s41598-024-85094-5] [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: 10/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Triple negative breast cancer (TNBC) is one of the deadliest subtypes of breast cancer, whose high frequency of relapse is often due to resistance to chemotherapy. Here, we identify inosine monophosphate dehydrogenase 2 (IMPDH2) as a contributor to doxorubicin resistance, in multiple TNBC models. Analysis of publicly available datasets reveals elevated IMPDH2 expression to associate with worse overall TNBC prognosis in the clinic, including lower recurrence-free survival post adjuvant/neoadjuvant therapy. Importantly, both genetic depletion and pharmacological inhibition of IMPDH2 leads to reduction of pro-tumorigenic phenotypes in multiple doxorubicin-resistant TNBC models, both in vitro and in vivo. Overall, we propose IMPDH2 as a novel vulnerability that could be leveraged therapeutically to suppress and/or prevent the growth of chemo-resistant lesions.
Collapse
Affiliation(s)
- Tatiane da Silva Fernandes
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Bryan M Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Jeffrey C Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Kanita A Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Scott M Dugas
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Alyssa A Fisher
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Pia Sharma
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - RongRong Wu
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher M Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, RSC R-410, Buffalo, NY, 14263, USA
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA
| | - Kazuaki Takabe
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer R Rosario
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, RSC R-410, Buffalo, NY, 14263, USA.
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, CGP L3-317, Buffalo, NY, 14263, USA.
| |
Collapse
|
2
|
Wang X, Li Z, Shen J, Liu L. Targeting protein tyrosine phosphatase non-receptor type 6 (PTPN6) as a therapeutic strategy in acute myeloid leukemia. Cell Biol Toxicol 2024; 41:11. [PMID: 39707066 PMCID: PMC11662038 DOI: 10.1007/s10565-024-09965-3] [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: 07/06/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by the clonal expansion of myeloid progenitor cells. Despite advancements in treatment, the prognosis for AML patients remains poor, highlighting the need for novel therapeutic targets. Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6), also known as SHP-1, is a critical regulator of hematopoietic cell signaling and has been implicated in various leukemias. This study investigates the therapeutic potential of targeting PTPN6 in AML. We employed both in vitro and in vivo models to evaluate the effects of PTPN6 inhibition on AML cell proliferation, apoptosis, and differentiation. Our results demonstrate that PTPN6 inhibition leads to a significant reduction in AML cell viability, induces apoptosis, and promotes differentiation of leukemic cells into mature myeloid cells. Mechanistic studies revealed that PTPN6 inhibition disrupts key signaling pathways involved in AML pathogenesis, including the JAK/STAT and PI3K/AKT pathways. Furthermore, the combination of PTPN6 inhibitors with standard chemotherapeutic agents exhibited a synergistic effect, enhancing the overall therapeutic efficacy. These findings suggest that PTPN6 is a promising therapeutic target in AML and warrants further investigation into the development of PTPN6 inhibitors for clinical application in AML treatment.
Collapse
Affiliation(s)
- Xiaoou Wang
- Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China
| | - Zhenggang Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Jing Shen
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, 110001, People's Republic of China.
| | - Lin Liu
- Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, People's Republic of China.
| |
Collapse
|
3
|
Vinod Kumar K, Kumar A, Kundal K, Sengupta A, R K, Singh S, Korra BT, Sharma S, Suresh V, Nishana M, Kumar R. AMLdb: a comprehensive multi-omics platform to identify biomarkers and drug targets for acute myeloid leukemia. Brief Funct Genomics 2024; 23:798-805. [PMID: 38864430 DOI: 10.1093/bfgp/elae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Acute myeloid leukemia (AML) is one of the leading leukemic malignancies in adults. The heterogeneity of the disease makes the diagnosis and treatment extremely difficult. With the advent of next-generation sequencing (NGS) technologies, exploration at the molecular level for the identification of biomarkers and drug targets has been the focus for the researchers to come up with novel therapies for better prognosis and survival outcomes of AML patients. However, the huge amount of data from NGS platforms requires a comprehensive AML platform to streamline literature mining efforts and save time. To facilitate this, we developed AMLdb, an interactive multi-omics platform that allows users to query, visualize, retrieve, and analyse AML related multi-omics data. AMLdb contains 86 datasets for gene expression profiles, 15 datasets for methylation profiles, CRISPR-Cas9 knockout screens of 26 AML cell lines, sensitivity of 26 AML cell lines to 288 drugs, mutations in 41 unique genes in 23 AML cell lines, and information on 41 experimentally validated biomarkers. In this study, we have reported five genes, i.e. CBFB, ENO1, IMPDH2, SEPHS2, and MYH9 identified via our analysis using AMLdb. ENO1 is uniquely identified gene which requires further investigation as a novel potential target while other reported genes have been previously confirmed as targets through experimental studies. Top of form we believe that these findings utilizing AMLdb can make it an invaluable resource to accelerate the development of effective therapies for AML and assisting the research community in advancing their understanding of AML pathogenesis. AMLdb is freely available at https://project.iith.ac.in/cgntlab/amldb.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Molecular Targeted Therapy
- Genomics/methods
- Databases, Genetic
- Multiomics
Collapse
Affiliation(s)
- Keerthana Vinod Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Ambuj Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Kavita Kundal
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Avik Sengupta
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Kunjulakshmi R
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Berhampur, Odisha 760010, India
| | - Subashani Singh
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Bhanu Teja Korra
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Simran Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Vandana Suresh
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Mayilaadumveettil Nishana
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Maruthamala P. O., Vithura, Kerala 695551, India
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| |
Collapse
|
4
|
Yabushita T, Goyama S. Nucleic acid metabolism: the key therapeutic target for myeloid tumors. Exp Hematol 2024; 142:104693. [PMID: 39647658 DOI: 10.1016/j.exphem.2024.104693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Nucleic acid analogs, including cytarabine, decitabine, and azacitidine, have significantly advanced therapeutic approaches for myeloid tumors over the past five decades. Nucleic acid metabolism is a crucial pathway driving myeloid tumorigenesis, with emerging evidence indicating that myeloid tumors are particularly dependent on the de novo nucleotide synthesis pathway, underscoring its potential as a therapeutic target. This review provides a comprehensive overview of nucleic acid metabolism, focusing on de novo nucleotide synthesis. We then described the range of clinically utilized agents targeting nucleic acid metabolism and discussed our recent findings on the nonepigenetic actions of decitabine, as well as the therapeutic effects of inosine monophosphate dehydrogenase (IMPDH) inhibitors in the treatment of myeloid tumors.
Collapse
Affiliation(s)
- Tomohiro Yabushita
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Japan.
| |
Collapse
|
5
|
Kieliszek AM, Mobilio D, Bassey-Archibong BI, Johnson JW, Piotrowski ML, de Araujo ED, Sedighi A, Aghaei N, Escudero L, Ang P, Gwynne WD, Zhang C, Quaile A, McKenna D, Subapanditha M, Tokar T, Vaseem Shaikh M, Zhai K, Chafe SC, Gunning PT, Montenegro-Burke JR, Venugopal C, Magolan J, Singh SK. De novo GTP synthesis is a metabolic vulnerability for the interception of brain metastases. Cell Rep Med 2024; 5:101755. [PMID: 39366383 PMCID: PMC11513854 DOI: 10.1016/j.xcrm.2024.101755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/21/2024] [Accepted: 09/06/2024] [Indexed: 10/06/2024]
Abstract
Patients with brain metastases (BM) face a 90% mortality rate within one year of diagnosis and the current standard of care is palliative. Targeting BM-initiating cells (BMICs) is a feasible strategy to treat BM, but druggable targets are limited. Here, we apply Connectivity Map analysis to lung-, breast-, and melanoma-pre-metastatic BMIC gene expression signatures and identify inosine monophosphate dehydrogenase (IMPDH), the rate-limiting enzyme in the de novo GTP synthesis pathway, as a target for BM. We show that pharmacological and genetic perturbation of IMPDH attenuates BMIC proliferation in vitro and the formation of BM in vivo. Metabolomic analyses and CRISPR knockout studies confirm that de novo GTP synthesis is a potent metabolic vulnerability in BM. Overall, our work employs a phenotype-guided therapeutic strategy to uncover IMPDH as a relevant target for attenuating BM outgrowth, which may provide an alternative treatment strategy for patients who are otherwise limited to palliation.
Collapse
Affiliation(s)
- Agata M Kieliszek
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Daniel Mobilio
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Blessing I Bassey-Archibong
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Jarrod W Johnson
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mathew L Piotrowski
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Elvin D de Araujo
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Abootaleb Sedighi
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Nikoo Aghaei
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Laura Escudero
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Patrick Ang
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - William D Gwynne
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Cunjie Zhang
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Andrew Quaile
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Dillon McKenna
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | | | - Tomas Tokar
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Muhammad Vaseem Shaikh
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Kui Zhai
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Shawn C Chafe
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Patrick T Gunning
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON, Canada
| | - J Rafael Montenegro-Burke
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Chitra Venugopal
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sheila K Singh
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Department of Surgery, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
6
|
Li J, Jin M, Yang J. Enhanced prognostic accuracy in severe TBI: a comprehensive nomogram analysis. Neurochirurgie 2024; 71:101597. [PMID: 39393588 DOI: 10.1016/j.neuchi.2024.101597] [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: 06/10/2024] [Revised: 08/20/2024] [Accepted: 09/15/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE This study aims to enhance prognostic accuracy in severe traumatic brain injury (STBI) by developing a novel nomogram that integrates clinical and paraclinical data. METHODS Data from 263 STBI patients were analyzed, focusing on critical variables such as age, Glasgow Coma Scale scores, pupil responsiveness, CT findings, and blood markers. A rigorous regression analysis was conducted to identify significant predictors. The nomogram underwent internal and external validation, and its predictive performance was compared with existing models through a meta-analysis. RESULTS The novel nomogram demonstrated superior predictive accuracy for STBI outcomes compared to traditional models. Key predictors, including age, Glasgow Coma Scale scores, pupil responsiveness, CT findings, and specific blood markers, were harmonized to provide a more precise prognostic tool. Validation processes confirmed the robustness and reliability of the nomogram. CONCLUSION The developed nomogram represents a significant advancement in STBI prognosis, offering clinicians a powerful tool to improve patient care strategies. By integrating CT imaging and blood parameters, the nomogram enhances the precision of outcome predictions, facilitating better-informed clinical decisions.
Collapse
Affiliation(s)
- Jie Li
- Department of Emergency Medicine, Guangyuan Central Hospital, Guangyuan 628000, China.
| | - Ming Jin
- Department of Emergency Medicine, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Jing Yang
- Department of Emergency Medicine, Guangyuan Central Hospital, Guangyuan 628000, China
| |
Collapse
|
7
|
Xie D, An B, Yang M, Wang L, Guo M, Luo H, Huang S, Sun F. Application and research progress of single cell sequencing technology in leukemia. Front Oncol 2024; 14:1389468. [PMID: 39267837 PMCID: PMC11390353 DOI: 10.3389/fonc.2024.1389468] [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: 02/21/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Leukemia is a malignant tumor with high heterogeneity and a complex evolutionary process. It is difficult to resolve the heterogeneity and clonal evolution of leukemia cells by applying traditional bulk sequencing techniques, thus preventing a deep understanding of the mechanisms of leukemia development and the identification of potential therapeutic targets. However, with the development and application of single-cell sequencing technology, it is now possible to investigate the gene expression profile, mutations, and epigenetic features of leukemia at the single-cell level, thus providing a new perspective for leukemia research. In this article, we review the recent applications and advances of single-cell sequencing technology in leukemia research, discuss its potential for enhancing our understanding of the mechanisms of leukemia development, discovering therapeutic targets and personalized treatment, and provide reference guidelines for the significance of this technology in clinical research.
Collapse
Affiliation(s)
- Dan Xie
- Medical College, Guizhou University, Guiyang, China
| | - Bangquan An
- Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Mingyue Yang
- Medical College, Guizhou University, Guiyang, China
| | - Lei Wang
- Medical College, Guizhou University, Guiyang, China
| | - Min Guo
- Medical College, Guizhou University, Guiyang, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, Guizhou, China
| | - Shengwen Huang
- Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Fa Sun
- Medical College, Guizhou University, Guiyang, China
| |
Collapse
|
8
|
Kaito S, Aoyama K, Oshima M, Tsuchiya A, Miyota M, Yamashita M, Koide S, Nakajima-Takagi Y, Kozuka-Hata H, Oyama M, Yogo T, Yabushita T, Ito R, Ueno M, Hirao A, Tohyama K, Li C, Kawabata KC, Yamaguchi K, Furukawa Y, Kosako H, Yoshimi A, Goyama S, Nannya Y, Ogawa S, Agger K, Helin K, Yamazaki S, Koseki H, Doki N, Harada Y, Harada H, Nishiyama A, Nakanishi M, Iwama A. Inhibition of TOPORS ubiquitin ligase augments the efficacy of DNA hypomethylating agents through DNMT1 stabilization. Nat Commun 2024; 15:7359. [PMID: 39198387 PMCID: PMC11358161 DOI: 10.1038/s41467-024-50498-4] [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: 07/27/2023] [Accepted: 07/02/2024] [Indexed: 09/01/2024] Open
Abstract
DNA hypomethylating agents (HMAs) are used for the treatment of myeloid malignancies, although their therapeutic effects have been unsatisfactory. Here we show that CRISPR-Cas9 screening reveals that knockout of topoisomerase 1-binding arginine/serine-rich protein (TOPORS), which encodes a ubiquitin/SUMO E3 ligase, augments the efficacy of HMAs on myeloid leukemic cells with little effect on normal hematopoiesis, suggesting that TOPORS is involved in resistance to HMAs. HMAs are incorporated into the DNA and trap DNA methyltransferase-1 (DNMT1) to form DNA-DNMT1 crosslinks, which undergo SUMOylation, followed by proteasomal degradation. Persistent crosslinking is cytotoxic. The TOPORS RING finger domain, which mediates ubiquitination, is responsible for HMA resistance. In TOPORS knockout cells, DNMT1 is stabilized by HMA treatment due to inefficient ubiquitination, resulting in the accumulation of unresolved SUMOylated DNMT1. This indicates that TOPORS ubiquitinates SUMOylated DNMT1, thereby promoting the resolution of DNA-DNMT1 crosslinks. Consistently, the ubiquitination inhibitor, TAK-243, and the SUMOylation inhibitor, TAK-981, show synergistic effects with HMAs through DNMT1 stabilization. Our study provides a novel HMA-based therapeutic strategy that interferes with the resolution of DNA-DNMT1 crosslinks.
Collapse
Affiliation(s)
- Satoshi Kaito
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Kazumasa Aoyama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akiho Tsuchiya
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Makiko Miyota
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masayuki Yamashita
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shuhei Koide
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yaeko Nakajima-Takagi
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroko Kozuka-Hata
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaaki Oyama
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takao Yogo
- Division of Cell Regulation, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomohiro Yabushita
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryoji Ito
- Central Institute for Experimental Animals, Yokohama, Kanagawa, Japan
| | - Masaya Ueno
- Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Atsushi Hirao
- Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kaoru Tohyama
- Department of Laboratory Medicine, Kawasaki Medical School, Okayama, Japan
| | - Chao Li
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Kimihito Cojin Kawabata
- Division of Clinical Precision Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Akihide Yoshimi
- Division of Cancer RNA Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuhito Nannya
- Division of Hematopoietic Disease Control, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Karl Agger
- BRIC University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- BRIC University of Copenhagen, Copenhagen, Denmark
- The Institute of Cancer Research (ICR), London, UK
| | - Satoshi Yamazaki
- Division of Cell Regulation, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Cell Engineering, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Molecular and Cellular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Yuka Harada
- Clinical Research Support Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hironori Harada
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Atsuya Nishiyama
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Laboratory of Cellular and Molecular Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
9
|
Shen B, Luo F, Yuan N, Yin J, Chai Y, Sun L, Zhang L, Luo C. Outcomes and complications of hemodialysis in patients with renal cancer following bilateral nephrectomy. Open Med (Wars) 2024; 19:20241009. [PMID: 39221033 PMCID: PMC11365463 DOI: 10.1515/med-2024-1009] [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: 11/15/2023] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Objectives The management of patients undergoing bilateral nephrectomy for renal cancer presents significant challenges, particularly in addressing hypotension, anemia, and tumor recurrence during hemodialysis. Case presentation A patient diagnosed with renal clear cell carcinoma in 2009 was followed until his demise in June 2022, with detailed documentation of symptoms, signs, laboratory results, diagnosis, and treatment. In the presented case, post-nephrectomy, the patient experienced frequent hypotension and anemia during dialysis, improving with erythropoietin-stimulating agents and subsequently with rosuvastatin. Later, multiple metastases were detected, correlating with normalized blood pressure and hemoglobin. Literature review A literature search up to September 2023 was also conducted, gathering data on hypotension, anemia, and tumor recurrence post-nephrectomy. Literature analysis of six cases revealed a 100% tumor recurrence rate in elderly patients (>50 years). Conclusion Treatment of anemia in bilateral nephrectomy patients warrants consideration of medication-induced tumor recurrence, highlighting early kidney transplantation to avoid adverse reactions like hypotension.
Collapse
Affiliation(s)
- Bing Shen
- Qingdao University, Qingdao266071, P.R. China
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Feng Luo
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Nan Yuan
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Jiaming Yin
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Yalin Chai
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Lijie Sun
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Lin Zhang
- The Affiliated Hospital of Qingdao University, Qingdao266000, Shandong, P.R. China
| | - Congjuan Luo
- The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao266000, Shandong, P.R. China
| |
Collapse
|
10
|
Guo J, Yang Y, Xiang Y, Zhang S, Guo X. Application of smart hydrogel materials in cartilage injury repair: A systematic review and meta-analysis. J Biomater Appl 2024; 39:96-116. [PMID: 38708775 DOI: 10.1177/08853282241248779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
OBJECTIVE Cartilage injury is a common clinical condition, and treatment approaches have evolved over time from traditional conservative and surgical methods to regenerative repair. In this context, hydrogels, as widely used biomaterials in the field of cartilage repair, have garnered significant attention. Particularly, responsive hydrogels (also known as "smart hydrogels") have shown immense potential due to their ability to respond to various physicochemical properties and environmental changes. This paper aims to review the latest research developments of hydrogels in cartilage repair, utilizing a more systematic and comprehensive meta-analysis approach to evaluate the research status and application value of responsive hydrogels. The goal is to determine whether these materials demonstrate favorable therapeutic effects for subsequent clinical applications, thereby offering improved treatment methods for patients with cartilage injuries. METHOD This study employed a systematic literature search method to summarize the research progress of responsive hydrogels by retrieving literature on the subject and review studies. The search terms included "hydrogel" and "cartilage," covering data from database inception up to October 2023. The quality of the literature was independently evaluated using Review Manager v5.4 software. Quantifiable data was statistically analyzed using the R language. RESULTS A total of 7 articles were retrieved for further meta-analysis. In the quality assessment, the studies demonstrated reliability and accuracy. The results of the meta-analysis indicated that responsive hydrogels exhibit unique advantages and effective therapeutic outcomes in the field of cartilage repair. Subgroup analysis revealed potential influences of factors such as different types of hydrogels and animal models on treatment effects. CONCLUSION Responsive hydrogels show significant therapeutic effects and substantial application potential in the field of cartilage repair. This study provides strong scientific evidence for their further clinical applications and research, with the hope of promoting advancements in the treatment of cartilage injuries.
Collapse
Affiliation(s)
- Juncheng Guo
- Central Laboratory of Haikou People's Hospital, Haikou Affiliated Hospital of Xiangya Medical College, Central South University, Haikou, P. R. China
| | - Yijun Yang
- Haikou People's Hospital, Xiangya School of Medicine, Haikou Affiliated Hospital of Central South University, Haikou, P. R. China
| | - Yang Xiang
- Haikou People's Hospital, Xiangya School of Medicine, Haikou Affiliated Hospital of Central South University, Haikou, P. R. China
| | - Shufang Zhang
- Central Laboratory, Haikou People's Hospital, Xiangya School of Medicine, Haikou Affiliated Hospital of Central South University, Haikou, P. R. China
| | - Xueyi Guo
- Central South University, Changsha, P. R. China
| |
Collapse
|
11
|
Tasca CI, Zuccarini M, Di Iorio P, Ciruela F. Lessons from the physiological role of guanosine in neurodegeneration and cancer: Toward a multimodal mechanism of action? Purinergic Signal 2024:10.1007/s11302-024-10033-y. [PMID: 39004650 DOI: 10.1007/s11302-024-10033-y] [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: 03/07/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases and brain tumours represent important health challenges due to their severe nature and debilitating consequences that require substantial medical care. Interestingly, these conditions share common physiological characteristics, namely increased glutamate, and adenosine transmission, which are often associated with cellular dysregulation and damage. Guanosine, an endogenous nucleoside, is safe and exerts neuroprotective effects in preclinical models of excitotoxicity, along with cytotoxic effects on tumour cells. However, the lack of well-defined mechanisms of action for guanosine hinders a comprehensive understanding of its physiological effects. In fact, the absence of specific receptors for guanosine impedes the development of structure-activity research programs to develop guanosine derivatives for therapeutic purposes. Alternatively, given its apparent interaction with the adenosinergic system, it is plausible that guanosine exerts its neuroprotective and anti-tumorigenic effects by modulating adenosine transmission through undisclosed mechanisms involving adenosine receptors, transporters, and purinergic metabolism. Here, several potential molecular mechanisms behind the protective actions of guanosine will be discussed. First, we explore its potential interaction with adenosine receptors (A1R and A2AR), including the A1R-A2AR heteromer. In addition, we consider the impact of guanosine on extracellular adenosine levels and the role of guanine-based purine-converting enzymes. Collectively, the diverse cellular functions of guanosine as neuroprotective and antiproliferative agent suggest a multimodal and complementary mechanism of action.
Collapse
Affiliation(s)
- Carla Inês Tasca
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
- Laboratory of Neurochemistry-4, Neuroscience Program/Biochemistry Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, 08907L'Hospitalet de Llobregat, Bellvitge, Spain
| |
Collapse
|
12
|
Zhang Y, Jin X, Lian L. Rare co-occurrence of tonsillar follicular dendritic cell sarcoma and schizophrenia: A comprehensive study. Clin Case Rep 2024; 12:e8700. [PMID: 38585586 PMCID: PMC10995268 DOI: 10.1002/ccr3.8700] [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: 12/08/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
This study investigated the infrequent occurrence of tonsillar follicular dendritic cell sarcoma (FDCS) co-existing with schizophrenia, presenting a comprehensive examination of clinical, pathological, and literature aspects. A systematic literature review was conducted, focusing on articles related to "schizophrenia" and "sarcoma," with in-depth analysis of included case reports. Clinical data, pathological findings, and patient follow-up information were collected and synthesized. The study detailed a rare case of FDCS in the tonsil concurrent with schizophrenia, providing insights into diagnosis, treatment, and follow-up. A literature review of combined FDCS in the tonsil and schizophrenia cases highlighted their clinical and pathological characteristics. Eight case reports encompassing 11 patients diagnosed with sarcoma and schizophrenia were included. Surgical resection was the preferred primary treatment, while chemotherapy was suggested for recurrences. Instances of co-occurring FDCS and schizophrenia were exceptionally limited, with tonsillar FDCS being particularly uncommon. The coexistence of tonsillar FDCS and schizophrenia was an exceptionally rare condition, posing diagnostic and therapeutic challenges. This study contributed valuable insights into clinical and pathological practice through a systematic review, underscoring the significance of early diagnosis and comprehensive management.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathology, Hangzhou Hospital of Traditional Chinese MedicineZhejiang Chinese Medicine UniversityHangzhouP. R. China
| | - Xiaoxi Jin
- Department of PathologyWenzhou People's HospitalWenzhouP. R. China
| | - Liyan Lian
- Department of Pathologythe First Affiliated Hospital of Zhejiang UniversityHangzhouP. R. China
| |
Collapse
|
13
|
Long R, Abulimiti N, Wang X. Proteomics-based clustering of lung adenocarcinoma identifies three subtypes with significantly different clinical and molecular features. Clin Transl Oncol 2024; 26:538-548. [PMID: 37603150 DOI: 10.1007/s12094-023-03275-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a predominant subtype of lung cancer. Although molecular classification of LUAD has been widely explored, proteomics-based subtyping of LUAD remains scarce. METHODS We proposed a subtyping method for LUAD based on the expression profiles of 500 proteins with the largest expression variability across LUAD. Furthermore, we comprehensively compared molecular and clinical features among the LUAD subtypes. RESULTS Consensus clustering identified three subtypes of LUAD, namely MtE, DrE, and StE. We demonstrated this subtyping method to be reproducible by analyzing two independent LUAD cohorts. MtE was characterized by high enrichment of metabolic pathways, high EGFR mutation rate, low stemness, proliferation, invasion, metastasis and inflammation signatures, favorable prognosis; DrE was characterized by high enrichment of DNA repair pathways, high TP53 mutation rate, and high levels of genomic instability, stemness, proliferation, and intratumor heterogeneity (ITH); and StE was characterized by high enrichment of stroma-related pathways, high KRAS mutation rate, and low levels of genomic instability. CONCLUSIONS The proteomics-based clustering analysis identified three LUAD subtypes with significantly different molecular and clinical properties. The novel subtyping method offers new perspectives on the cancer biology and holds promise in improving the clinical management of LUAD.
Collapse
Affiliation(s)
- Rongzhuo Long
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Nayila Abulimiti
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
14
|
Sezaki M, Huang G. Repurposing immunosuppressants for antileukemia therapy. EMBO Mol Med 2022; 15:e17042. [PMID: 36453114 PMCID: PMC9832814 DOI: 10.15252/emmm.202217042] [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: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
Drug repurposing, the strategy to identify new therapeutic use for clinically approved drugs has attracted much attention in recent years. This strategy offers various advantages over traditional approaches to develop new drugs, including shorter development timelines, low cost, and reduced risk of failure. In this issue of EMBO Molecular Medicine, Liu et al show that inosine monophosphate dehydrogenase (IMPDH) inhibitors, the well-known immunosuppressants have a potent therapeutic effect on the aggressive blood cancer, acute myeloid leukemia with MLL rearrangements. Intriguingly, the antileukemia effect of IMPDH inhibitors is mediated, at least in part through the overactivation of TLR signaling and Vcam1 upregulation. The robust antileukemia effect of IMPDH inhibitors, both in vitro and in vivo, together with their mechanistic findings provides a rational basis for repurposing IMPDH inhibitors for antileukemia therapy.
Collapse
Affiliation(s)
- Maiko Sezaki
- Department of Cell Systems and AnatomyUT Health San AntonioJoe R. & Teresa Lozano Long School of MedicineSan AntonioTXUSA
| | - Gang Huang
- Department of Cell Systems and AnatomyUT Health San AntonioJoe R. & Teresa Lozano Long School of MedicineSan AntonioTXUSA,Department of Pathology and Laboratory MedicineUT Health San AntonioJoe R. & Teresa Lozano Long School of MedicineSan AntonioTXUSA,Mays Cancer Center at UT Health San AntonioSan AntonioTXUSA
| |
Collapse
|