1
|
Xiong S, Zhang Y, Zhou X, Pant V, Mirani A, Gencel-Augusto J, Chau G, You MJ, Lozano G. Dependence on Mdm2 for Mdm4 inhibition of p53 activity. Cancer Lett 2025; 621:217622. [PMID: 40081463 DOI: 10.1016/j.canlet.2025.217622] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Both Mdm2 and Mdm4 inhibit p53 activity by masking of its transcriptional activation domain. In addition, Mdm2 functions as an E3 ubiquitin ligase, targeting p53 for degradation. The amino terminus of Mdm4 binds wild type and mutant p53 while its RING domain, which lacks E3 ligase activity, is required for heterodimerization with Mdm2. To determine how these domains of Mdm4 regulate p53, we generated mouse models with either a deletion of the Mdm4 RING domain (Mdm4ΔR) or all of Mdm4 (Mdm4─) on a hypomorphic (p53neo) background. Mdm4ΔR mice exhibited elevated p53 levels and activity, albeit to a lesser extent than mice with complete Mdm4 loss, indicating that the amino terminus of Mdm4 contributes to p53 inhibition. Moreover, in the absence of Mdm2, neither the deletion of the Mdm4 RING domain nor the complete loss of Mdm4 further increased p53 protein levels on a mutant p53 background, indicating that Mdm4 modulates Mdm2 in its regulation of p53 stability. Collectively, our findings suggest that Mdm4 contributes to p53 inhibition by modulating Mdm2 activity via both its amino terminus and RING domains.
Collapse
Affiliation(s)
- Shunbin Xiong
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yun Zhang
- Department of Pharmaceutical Sciences, Joan M. Lafleur College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, 77004, USA
| | - Xin Zhou
- Department of Pediatrics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Vinod Pant
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Akshita Mirani
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Gilda Chau
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - M James You
- Department of Hematopathology, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Guillermina Lozano
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Zhu IY, Lloyd A, Critchley WR, Saikia Q, Jade D, Divan A, Zeqiraj E, Harrison MA, Brown CJ, Ponnambalam S. Structure and function of MDM2 and MDM4 in health and disease. Biochem J 2025; 482:BCJ20240757. [PMID: 39960347 PMCID: PMC12096895 DOI: 10.1042/bcj20240757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 05/09/2025]
Abstract
Both mouse double-minute 2 (MDM2), an E3 ubiquitin ligase, and its closely related paralog, MDM4, which lacks E3 activity, play central roles in cellular homeostasis. MDM-linked dysfunction is associated with an increased risk of oncogenesis, primarily through targeting the tumor suppressor protein p53 for ubiquitination and degradation. Recent studies have revealed multifaceted roles of MDM proteins that are p53 independent with implications for their oncogenic properties. This review aims to provide an overview of MDM2 and MDM4, by assessing gene and protein structure and implications for protein-protein interactions and functions in cell and animal physiology. We also explore MDM2 and MDM4 role(s) in angiogenesis, a critical feature of solid tumor growth and progression. Finally, we discuss the current landscape in the development of MDM2 and MDM4 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Ivy Yiyi Zhu
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - Alec Lloyd
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - William R. Critchley
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - Queen Saikia
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - Dhananjay Jade
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Aysha Divan
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - Elton Zeqiraj
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - Michael A. Harrison
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | - Christopher J. Brown
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
3
|
Khanna V, Eslami G, Reyes R, Diep R, Fernandez-Pol S, Stehr H, Suarez CJ, Pinto H, Ford JM, Zhang TY, Chen CT. MDM2 inhibition is associated with the emergence of TP53-altered clonal hematopoiesis. NPJ Precis Oncol 2025; 9:34. [PMID: 39900989 PMCID: PMC11790943 DOI: 10.1038/s41698-025-00823-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: 07/15/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
Murine double minute 2 (MDM2) inhibitors have shown promising activity in TP53-wild type tumors and are under active investigation across a spectrum of malignancies. Herein, we report a 51-year-old female with MDM2-amplified, TP53-wild type adenoid cystic carcinoma who was treated with a MDM2 inhibitor and developed persistent pancytopenia despite drug discontinuation. Her pancytopenia was associated with 20 distinct pathogenic TP53 mutations in peripheral blood and bone marrow not present in drug-resistant tumor tissue. Plasma TP53 mutations were similarly detected among 4 other patients treated at our institution, with the number of mutations correlating strongly with duration of treatment. This case suggests that MDM2 inhibitors are associated with TP53 clonal hematopoiesis, which may confer a risk of subsequent myeloid malignancy. As multiple MDM2 inhibitor trials are ongoing, our findings underscore the need for further investigation into the potential long-term deleterious effects of these inhibitors in the hematopoietic stem and progenitor compartment.
Collapse
Affiliation(s)
- Vishesh Khanna
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford, CA, 94305, USA
| | - Gohar Eslami
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford, CA, 94305, USA
| | - Rochelle Reyes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford, CA, 94305, USA
| | - Robert Diep
- Stanford Cancer Institute, Stanford, CA, 94305, USA
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Henning Stehr
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Carlos Jose Suarez
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Harlan Pinto
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford, CA, 94305, USA
| | - James M Ford
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford, CA, 94305, USA
| | - Tian Yi Zhang
- Stanford Cancer Institute, Stanford, CA, 94305, USA
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Christopher T Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Stanford Cancer Institute, Stanford, CA, 94305, USA.
| |
Collapse
|
4
|
Wu KKL, Xu X, Wu M, Li X, Hoque M, Li GHY, Lian Q, Long K, Zhou T, Piao H, Xu A, Hui HX, Cheng KKY. MDM2 induces pro-inflammatory and glycolytic responses in M1 macrophages by integrating iNOS-nitric oxide and HIF-1α pathways in mice. Nat Commun 2024; 15:8624. [PMID: 39366973 PMCID: PMC11452520 DOI: 10.1038/s41467-024-53006-w] [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: 10/20/2022] [Accepted: 09/28/2024] [Indexed: 10/06/2024] Open
Abstract
M1 macrophages induce protective immunity against infection, but also contribute to metabolic and inflammatory diseases. Here we show that the E3 ubiquitin ligase, MDM2, promotes the glycolytic and inflammatory activities of M1 macrophage by increasing the production of IL-1β, MCP-1 and nitric oxide (NO). Mechanistically, MDM2 triggers the ubiquitination and degradation of E3 ligase, SPSB2, to stabilize iNOS and increases production of NO, which s-nitrosylates and activates HIF-1α for triggering the glycolytic and pro-inflammatory programs in M1 macrophages. Myeloid-specific haplodeletion of MDM2 in mice not only blunts LPS-induced endotoxemia and NO production, but also alleviates obesity-induced adipose tissue-resident macrophage inflammation. By contrast, MDM2 haplodeletion induces higher mortality, tissue damage and bacterial burden, and also suppresses M1 macrophage response, in the cecal ligation and puncture-induced sepsis mouse model. Our findings thus identify MDM2 as an activator of glycolytic and inflammatory responses in M1 macrophages by connecting the iNOS-NO and HIF-1α pathways.
Collapse
Affiliation(s)
- Kelvin Ka-Lok Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaofan Xu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Manyin Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital Fudan University, Shanghai, China
| | - Moinul Hoque
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Gloria Hoi Yee Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Qizhou Lian
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Prenatal Diagnostic Center and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Kekao Long
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Tongxi Zhou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hailong Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Beijing, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Hannah Xiaoyan Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
5
|
Meader E, Walcheck MT, Leder MR, Jing R, Wrighton PJ, Sugden WW, Najia MA, Oderberg IM, Taylor VM, LeBlanc ZC, Quenzer ED, Lim SE, Daley GQ, Goessling W, North TE. Bnip3lb-driven mitophagy sustains expansion of the embryonic hematopoietic stem cell pool. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614531. [PMID: 39386657 PMCID: PMC11463499 DOI: 10.1101/2024.09.23.614531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Embryonic hematopoietic stem and progenitor cells (HSPCs) have the unique ability to undergo rapid proliferation while maintaining multipotency, a clinically-valuable quality which currently cannot be replicated in vitro. Here, we show that embryonic HSPCs achieve this state by precise spatio-temporal regulation of reactive oxygen species (ROS) via Bnip3lb-associated developmentally-programmed mitophagy, a distinct autophagic regulatory mechanism from that of adult HSPCs. While ROS drives HSPC specification in the dorsal aorta, scRNAseq and live-imaging of Tg(ubi:mitoQC) zebrafish indicate that mitophagy initiates as HSPCs undergo endothelial-to-hematopoietic transition and colonize the caudal hematopoietic tissue (CHT). Knockdown of bnip3lb reduced mitophagy and HSPC numbers in the CHT by promoting myeloid-biased differentiation and apoptosis, which was rescued by anti-oxidant exposure. Conversely, induction of mitophagy enhanced both embryonic HSPC and lymphoid progenitor numbers. Significantly, mitophagy activation improved ex vivo functional capacity of hematopoietic progenitors derived from human-induced pluripotent stem cells (hiPSCs), enhancing serial-replating hematopoietic colony forming potential. HIGHLIGHTS ROS promotes HSPC formation in the dorsal aorta but negatively affects maintenance thereafter.HSPCs colonizing secondary niches control ROS levels via Bnip3lb-directed mitophagy.Mitophagy protects nascent HSPCs from ROS-associated apoptosis and maintains multipotency.Induction of mitophagy enhances long-term hematopoietic potential of iPSC-derived HSPCs.
Collapse
|
6
|
Shen E, Piao M, Li Y, Wu Y, Li S, Lee SH, Jin L, Lee KY. CMTM3 Suppresses Proliferation and Osteogenic Transdifferentiation of C2C12 Myoblasts through p53 Upregulation. Cells 2024; 13:1352. [PMID: 39195242 PMCID: PMC11352514 DOI: 10.3390/cells13161352] [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: 07/18/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
CKLF-like MARVEL transmembrane domain-containing 3 (CMTM3), a member of the CMTM family that is closely related to tumor occurrence and progression, plays crucial roles in the immune system, cardiovascular system, and male reproductive system. Recently, CMTM3 has emerged as a potential target for treating diseases related to bone formation. However, additional studies are needed to understand the mechanisms by which CMTM3 regulates the process of osteogenic differentiation. In this study, we observed a significant downregulation of Cmtm3 expression during the transdifferentiation of C2C12 myoblasts into osteoblasts induced by BMP4. Cmtm3 overexpression suppressed proliferation and osteogenic differentiation in BMP4-induced C2C12 cells, whereas its knockdown conversely facilitated the process. Mechanistically, Cmtm3 overexpression upregulated both the protein and mRNA levels of p53 and p21. Conversely, Cmtm3 knockdown exerted the opposite effects. Additionally, we found that Cmtm3 interacts with p53 and increases protein stability by inhibiting proteasome-mediated ubiquitination and degradation. Notably, Trp53 downregulation abrogated the inhibitory effect of Cmtm3 on BMP4-induced proliferation and osteogenic differentiation of C2C12 myoblasts. Collectively, our findings provide key insights into the role of CMTM3 in regulating myoblast proliferation and transdifferentiation into osteoblasts, highlighting its significance in osteogenesis research.
Collapse
Affiliation(s)
- Enzhao Shen
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Meiyu Piao
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| | - Yuankuan Li
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| | - Yuecheng Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Sihang Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Sung Ho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (S.L.)
| | - Kwang Youl Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (E.S.); (M.P.); (Y.L.)
| |
Collapse
|
7
|
Kanbar K, El Darzi R, Jaalouk DE. Precision oncology revolution: CRISPR-Cas9 and PROTAC technologies unleashed. Front Genet 2024; 15:1434002. [PMID: 39144725 PMCID: PMC11321987 DOI: 10.3389/fgene.2024.1434002] [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: 05/16/2024] [Accepted: 07/02/2024] [Indexed: 08/16/2024] Open
Abstract
Cancer continues to present a substantial global health challenge, with its incidence and mortality rates persistently reflecting its significant impact. The emergence of precision oncology has provided a breakthrough in targeting oncogenic drivers previously deemed "undruggable" by conventional therapeutics and by limiting off-target cytotoxicity. Two groundbreaking technologies that have revolutionized the field of precision oncology are primarily CRISPR-Cas9 gene editing and more recently PROTAC (PROteolysis TArgeting Chimeras) targeted protein degradation technology. CRISPR-Cas9, in particular, has gained widespread recognition and acclaim due to its remarkable ability to modify DNA sequences precisely. Rather than editing the genetic code, PROTACs harness the ubiquitin proteasome degradation machinery to degrade proteins of interest selectively. Even though CRISPR-Cas9 and PROTAC technologies operate on different principles, they share a common goal of advancing precision oncology whereby both approaches have demonstrated remarkable potential in preclinical and promising data in clinical trials. CRISPR-Cas9 has demonstrated its clinical potential in this field due to its ability to modify genes directly and indirectly in a precise, efficient, reversible, adaptable, and tissue-specific manner, and its potential as a diagnostic tool. On the other hand, the ability to administer in low doses orally, broad targeting, tissue specificity, and controllability have reinforced the clinical potential of PROTAC. Thus, in the field of precision oncology, gene editing using CRISPR technology has revolutionized targeted interventions, while the emergence of PROTACs has further expanded the therapeutic landscape by enabling selective protein degradation. Rather than viewing them as mutually exclusive or competing methods in the field of precision oncology, their use is context-dependent (i.e., based on the molecular mechanisms of the disease) and they potentially could be used synergistically complementing the strengths of CRISPR and vice versa. Herein, we review the current status of CRISPR and PROTAC designs and their implications in the field of precision oncology in terms of clinical potential, clinical trial data, limitations, and compare their implications in precision clinical oncology.
Collapse
Affiliation(s)
- Karim Kanbar
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Roy El Darzi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Diana E. Jaalouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
8
|
Wang W, Albadari N, Du Y, Fowler JF, Sang HT, Xian W, McKeon F, Li W, Zhou J, Zhang R. MDM2 Inhibitors for Cancer Therapy: The Past, Present, and Future. Pharmacol Rev 2024; 76:414-453. [PMID: 38697854 PMCID: PMC11068841 DOI: 10.1124/pharmrev.123.001026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 05/05/2024] Open
Abstract
Since its discovery over 35 years ago, MDM2 has emerged as an attractive target for the development of cancer therapy. MDM2's activities extend from carcinogenesis to immunity to the response to various cancer therapies. Since the report of the first MDM2 inhibitor more than 30 years ago, various approaches to inhibit MDM2 have been attempted, with hundreds of small-molecule inhibitors evaluated in preclinical studies and numerous molecules tested in clinical trials. Although many MDM2 inhibitors and degraders have been evaluated in clinical trials, there is currently no Food and Drug Administration (FDA)-approved MDM2 inhibitor on the market. Nevertheless, there are several current clinical trials of promising agents that may overcome the past failures, including agents granted FDA orphan drug or fast-track status. We herein summarize the research efforts to discover and develop MDM2 inhibitors, focusing on those that induce MDM2 degradation and exert anticancer activity, regardless of the p53 status of the cancer. We also describe how preclinical and clinical investigations have moved toward combining MDM2 inhibitors with other agents, including immune checkpoint inhibitors. Finally, we discuss the current challenges and future directions to accelerate the clinical application of MDM2 inhibitors. In conclusion, targeting MDM2 remains a promising treatment approach, and targeting MDM2 for protein degradation represents a novel strategy to downregulate MDM2 without the side effects of the existing agents blocking p53-MDM2 binding. Additional preclinical and clinical investigations are needed to finally realize the full potential of MDM2 inhibition in treating cancer and other chronic diseases where MDM2 has been implicated. SIGNIFICANCE STATEMENT: Overexpression/amplification of the MDM2 oncogene has been detected in various human cancers and is associated with disease progression, treatment resistance, and poor patient outcomes. This article reviews the previous, current, and emerging MDM2-targeted therapies and summarizes the preclinical and clinical studies combining MDM2 inhibitors with chemotherapy and immunotherapy regimens. The findings of these contemporary studies may lead to safer and more effective treatments for patients with cancers overexpressing MDM2.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Najah Albadari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Yi Du
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Josef F Fowler
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Hannah T Sang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Wa Xian
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Frank McKeon
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Wei Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Jia Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy (W.W., Y.D., J.F.F., H.T.S., R.Z.), Drug Discovery Institute (W.W., R.Z.), Stem Cell Center, Department of Biology and Biochemistry (W.X., F.M.), University of Houston, Houston, Texas; College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee (N.A., W.L.); and Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas (J.Z.)
| |
Collapse
|
9
|
Shome R, Sen P, Sarkar S, Ghosh SS. Single-cell transcriptomics reveals the intra-tumoral heterogeneity and SQSTM1/P62 and Wnt/β-catenin mediated epithelial to mesenchymal transition and stemness of triple-negative breast cancer. Exp Cell Res 2024; 438:114032. [PMID: 38583856 DOI: 10.1016/j.yexcr.2024.114032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Triple-negative breast cancer (TNBC) is characterized by the complex tumor microenvironment (TME) consisting of an abundance of mesenchymal stem cells (MSCs), which is known to facilitate epithelial-to-mesenchymal transition (EMT). The development of single-cell genomics is a powerful method for defining the intricate genetic landscapes of malignancies. In this study, we have employed single-cell RNA sequencing (scRNA-seq) to dissect the intra-tumoral heterogeneity and analyze the single-cell transcriptomic landscape to detect rare consequential cell subpopulations of significance. The scRNA-seq analysis of TNBC and Normal patient derived samples revealed that EMT markers and transcription factors were most upregulated in MSC population. Further, exploration of gene expression analysis among TNBC and Normal patient-derived MSCs ascertained the role of SQSTM1/P62 and Wnt/β-catenin in TNBC progression. Wnt/β-catenin and Wnt/PCP signaling pathways are prominent contributors of EMT, stemness, and cancer stem cell (CSC) properties of TNBC. SQSTM1/P62 cooperates with the components of the Wnt/PCP signaling pathway and is critically involved at the interface of autophagy and EMT. Moreover, siRNA targeting SQSTM1/P62 and inhibitor of Wnt/β-catenin (FH535) in conjunction was used to explore molecular modification of EMT and stemness markers. Although SQSTM1/P62 is not crucial for cell survival, cytotoxicity assay revealed synergistic interaction between the siRNA/inhibitor. Modulation of these important pathways helped in reduction of expression of genes and proteins contributing to CSC properties. Gene and protein expression analysis revealed the induction of EMT to MET. Moreover, co-treatment resulted in inactivation of non-canonical Wnt VANGL2-JNK signaling axis. The synergistic impact of inhibition of SQSTM1/P62 and Wnt/β-catenin signaling facilitates the development of a potential therapeutic regimen for TNBC.
Collapse
Affiliation(s)
- Rajib Shome
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 39, Assam, India
| | - Plaboni Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 39, Assam, India
| | - Shilpi Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 39, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 39, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 39, Assam, India.
| |
Collapse
|
10
|
Ishida T, Heck AM, Varnum-Finney B, Dozono S, Nourigat-McKay C, Kraskouskas K, Wellington R, Waltner O, Root, Jackson DL, Delaney C, Rafii S, Bernstein ID, Trapnell, Hadland B. Differentiation latency and dormancy signatures define fetal liver HSCs at single cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543314. [PMID: 37333272 PMCID: PMC10274697 DOI: 10.1101/2023.06.01.543314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Decoding the gene regulatory mechanisms mediating self-renewal of hematopoietic stem cells (HSCs) during their amplification in the fetal liver (FL) is relevant for advancing therapeutic applications aiming to expand transplantable HSCs, a long-standing challenge. Here, to explore intrinsic and extrinsic regulation of self-renewal in FL-HSCs at the single cell level, we engineered a culture platform designed to recapitulate the FL endothelial niche, which supports the amplification of serially engraftable HSCs ex vivo. Leveraging this platform in combination with single cell index flow cytometry, serial transplantation assays, and single cell RNA-sequencing, we elucidated previously unrecognized heterogeneity in immunophenotypically defined FL-HSCs and demonstrated that differentiation latency and transcriptional signatures of biosynthetic dormancy are distinguishing properties of self-renewing FL-HSCs with capacity for serial, long-term multilineage hematopoietic reconstitution. Altogether, our findings provide key insights into HSC expansion and generate a novel resource for future exploration of the intrinsic and niche-derived signaling pathways that support FL-HSC self-renewal.
Collapse
Affiliation(s)
- Takashi Ishida
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adam M. Heck
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Barbara Varnum-Finney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stacey Dozono
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cynthia Nourigat-McKay
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Katie Kraskouskas
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rachel Wellington
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA
| | - Olivia Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Root
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Colleen Delaney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Deverra Therapeutics, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Irwin D. Bernstein
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brandon Hadland
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
11
|
Xu X, Li G, Zhang D, Zhu H, Liu G, Zhang Z. Gut Microbiota is Associated with Aging-Related Processes of a Small Mammal Species under High-Density Crowding Stress. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205346. [PMID: 36965140 PMCID: PMC10190659 DOI: 10.1002/advs.202205346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/15/2023] [Indexed: 05/18/2023]
Abstract
Humans and animals frequently encounter high-density crowding stress, which may accelerate their aging processes; however, the roles of gut microbiota in the regulation of aging-related processes under high-density crowding stress remain unclear. In the present study, it is found that high housing density remarkably increases the stress hormone (corticosterone), accelerates aging-related processes as indicated by telomere length (in brain and liver cells) and DNA damage or inflammation (as revealed by tumor necrosis factor-α and interleukin-10 levels), and reduces the lifespan of Brandt's vole (Lasiopodomys brandtii). Fecal microbiota transplantation from donor voles of habitats with different housing densities induces similar changes in aging-related processes in recipient voles. The elimination of high housing density or butyric acid administration delays the appearance of aging-related markers in the brain and liver cells of voles housed at high-density. This study suggests that gut microorganisms may play a significant role in regulating the density-dependent aging-related processes and subsequent population dynamics of animals, and can be used as potential targets for alleviating stress-related aging in humans exposed to high-density crowding stress.
Collapse
Affiliation(s)
- Xiaoming Xu
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Biotic InteractionsUniversity of Chinese Academy of SciencesBeijing100049China
| | - Da Zhang
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hanyi Zhu
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Guang‐hui Liu
- Institute for Stem cell and RegenerationCASBeijing100049China
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Biotic InteractionsUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
12
|
Zhan Q, Wang J, Zhang H, Zhang L. E3 ubiquitin ligase on the biological properties of hematopoietic stem cell. J Mol Med (Berl) 2023; 101:543-556. [PMID: 37081103 PMCID: PMC10163092 DOI: 10.1007/s00109-023-02315-6] [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: 11/08/2022] [Revised: 02/25/2023] [Accepted: 03/30/2023] [Indexed: 04/22/2023]
Abstract
Hematopoietic stem cells are a group of heterogeneity cells with the potential to differentiate into various types of mature blood cells. Their basic biological properties include quiescence, self-renewal, multilineage differentiation, and homing ability, with the homing of exogenous hematopoietic stem cells after transplantation becoming a new focus, while the first three properties share some similarity in mechanism due to connectivity. In various complex mechanisms, the role of E3 ubiquitin ligases in hematopoietic homeostasis and malignant transformation is receiving increasing attention. As a unique part, E3 ubiquitin ligases play an important role in physiological regulation mechanism of posttranslational modification. In this review, we focus on the recent progress of the crucial role of E3 ubiquitin ligases that target specific proteins for ubiquitination to regulate biological properties of hematopoietic stem cells. Additionally, this paper deals with E3 ubiquitin ligases that affect the biological properties through aging and summarizes the relevant applications of targeting E3 ligases in hematopoietic malignancies. We present some ideas on the clinical application of E3 ubiquitin ligase to regulate hematopoietic stem cells and also believe that it is meaningful to study the upstream signal of these E3 ubiquitin ligases because hematopoietic stem cell dysfunction is caused by deficiency of some E3 ligases.
Collapse
Affiliation(s)
- Qianru Zhan
- Department of Hematology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang, Liaoning, People's Republic of China
| | - Jing Wang
- Department of Hematology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang, Liaoning, People's Republic of China
| | - Heyang Zhang
- Department of Hematology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang, Liaoning, People's Republic of China.
| | - Lijun Zhang
- Department of Hematology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
13
|
Grey W, Atkinson S, Rix B, Casado P, Ariza-McNaughton L, Hawley C, Sopoena ML, Bridge KS, Kent D, Cutillas PR, Bonnet D. The CKS1/CKS2 Proteostasis Axis Is Crucial to Maintain Hematopoietic Stem Cell Function. Hemasphere 2023; 7:e853. [PMID: 36874381 PMCID: PMC9977483 DOI: 10.1097/hs9.0000000000000853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/30/2023] [Indexed: 03/04/2023] Open
Abstract
Long-term hematopoietic stem cells are rare, highly quiescent stem cells of the hematopoietic system with life-long self-renewal potential and the ability to transplant and reconstitute the entire hematopoietic system of conditioned recipients. Most of our understanding of these rare cells has relied on cell surface identification, epigenetic, and transcriptomic analyses. Our knowledge of protein synthesis, folding, modification, and degradation-broadly termed protein homeostasis or "proteostasis"-in these cells is still in its infancy, with very little known about how the functional state of the proteome is maintained in hematopoietic stem cells. We investigated the requirement of the small phospho-binding adaptor proteins, the cyclin-dependent kinase subunits (CKS1 and CKS2), for maintaining ordered hematopoiesis and long-term hematopoietic stem cell reconstitution. CKS1 and CKS2 are best known for their roles in p27 degradation and cell cycle regulation, and by studying the transcriptome and proteome of Cks1 -/- and Cks2 -/- mice, we demonstrate regulation of key signaling pathways that govern hematopoietic stem cell biology including AKT, FOXO1, and NFκB, together balancing protein homeostasis and restraining reactive oxygen species to ensure healthy hematopoietic stem cell function.
Collapse
Affiliation(s)
- William Grey
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Samantha Atkinson
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Beatrice Rix
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro Casado
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, United Kingdom
| | | | - Cathy Hawley
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Miriam L. Sopoena
- Bioinformatics Core, The Francis Crick Institute, London, United Kingdom
| | - Katherine S. Bridge
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - David Kent
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro R. Cutillas
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, United Kingdom
| | - Dominique Bonnet
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
14
|
Zhou S, Zhu J, Zhou PK, Gu Y. Alveolar type 2 epithelial cell senescence and radiation-induced pulmonary fibrosis. Front Cell Dev Biol 2022; 10:999600. [PMID: 36407111 PMCID: PMC9666897 DOI: 10.3389/fcell.2022.999600] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a chronic and progressive respiratory tract disease characterized by collagen deposition. The pathogenesis of RIPF is still unclear. Type 2 alveolar epithelial cells (AT2), the essential cells that maintain the structure and function of lung tissue, are crucial for developing pulmonary fibrosis. Recent studies indicate the critical role of AT2 cell senescence during the onset and progression of RIPF. In addition, clearance of senescent AT2 cells and treatment with senolytic drugs efficiently improve lung function and radiation-induced pulmonary fibrosis symptoms. These findings indicate that AT2 cell senescence has the potential to contribute significantly to the innovative treatment of fibrotic lung disorders. This review summarizes the current knowledge from basic and clinical research about the mechanism and functions of AT2 cell senescence in RIPF and points to the prospects for clinical treatment by targeting senescent AT2 cells.
Collapse
Affiliation(s)
- Shenghui Zhou
- Hengyang Medical College, University of South China, Hengyang, China,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China
| | - Jiaojiao Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China
| | - Ping-Kun Zhou
- Hengyang Medical College, University of South China, Hengyang, China,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China,*Correspondence: Yongqing Gu, ; Ping-Kun Zhou,
| | - Yongqing Gu
- Hengyang Medical College, University of South China, Hengyang, China,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China,*Correspondence: Yongqing Gu, ; Ping-Kun Zhou,
| |
Collapse
|
15
|
Fujino T, Asada S, Goyama S, Kitamura T. Mechanisms involved in hematopoietic stem cell aging. Cell Mol Life Sci 2022; 79:473. [PMID: 35941268 PMCID: PMC11072869 DOI: 10.1007/s00018-022-04356-5] [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: 01/28/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
Abstract
Hematopoietic stem cells (HSCs) undergo progressive functional decline over time due to both internal and external stressors, leading to aging of the hematopoietic system. A comprehensive understanding of the molecular mechanisms underlying HSC aging will be valuable in developing novel therapies for HSC rejuvenation and to prevent the onset of several age-associated diseases and hematological malignancies. This review considers the general causes of HSC aging that range from cell-intrinsic factors to cell-extrinsic factors. In particular, epigenetics and inflammation have been implicated in the linkage of HSC aging, clonality, and oncogenesis. The challenges in clarifying mechanisms of HSC aging have accelerated the development of therapeutic interventions to rejuvenate HSCs, the major goal of aging research; these details are also discussed in this review.
Collapse
Affiliation(s)
- Takeshi Fujino
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Shuhei Asada
- The Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, 1628666, Japan
| | - Susumu Goyama
- Division of Molecular Oncology Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 1088639, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
16
|
Rg1 Protects Hematopoietic Stem Cells from LiCl-Induced Oxidative Stress via Wnt Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2875583. [PMID: 35388306 PMCID: PMC8977299 DOI: 10.1155/2022/2875583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/09/2022] [Accepted: 02/11/2022] [Indexed: 11/22/2022]
Abstract
Background Ginsenoside Rg1 is a major component of ginseng with antioxidative and antiaging effects, which is a traditional Chinese medicine. In this study, we investigated the potential spillover and mechanism of action of Rg1 on LiCl-driven hematopoietic stem cell aging. Results Collect the purified Sca-1+ hematopoietic cells for differentiation ability detection and biochemical and molecular labeling. The experiment found that Rg1 plays an antiaging role in reversing the SA-β-gal staining associated with LiCl-induced hematopoietic stem cell senescence, the increase in p53 and p21 proteins, and sustained DNA damage. At the same time, Rg1 protects hematopoietic cells from the reduced differentiation ability caused by LiCl. In addition, Rg1 increased the excessive inhibition of intracellular GSK-3β protein, resulting in the maintenance of β-catenin protein levels in hematopoietic cells after LiCl treatment. Then, the target gene level of β-catenin can be maintained. Conclusions Rg1 exerts the pharmacological effect of maintaining the activity of GSK-3β in Sca-1+ hematopoietic cells, enhances the antioxidant potential of cells, improves the redox homeostasis, and thus protects cells from the decline in differentiation ability caused by aging. This study provides a potential therapeutic strategy to reduce stem cell pool failure caused by chronic oxidative damage to hematopoietic stem cells.
Collapse
|
17
|
Klimovich B, Meyer L, Merle N, Neumann M, König AM, Ananikidis N, Keber CU, Elmshäuser S, Timofeev O, Stiewe T. Partial p53 reactivation is sufficient to induce cancer regression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:80. [PMID: 35232479 PMCID: PMC8889716 DOI: 10.1186/s13046-022-02269-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/20/2022] [Indexed: 01/21/2023]
Abstract
Background Impaired p53 function is one of the central molecular features of a tumor cell and even a partial reduction in p53 activity can increase the cancer risk in mice and men. From a therapeutic perspective it is noteworthy that tumor cells often become addicted to the absence of p53 providing a rationale for developing p53 reactivating compounds to treat cancer patients. Unfortunately, many of the compounds that are currently undergoing preclinical and clinical testing fail to fully reactivate mutant p53 proteins, raising the crucial question: how much p53 activity is needed to elicit a therapeutic effect? Methods We have genetically modelled partial p53 reactivation using knock-in mice with inducible expression of the p53 variant E177R. This variant has a reduced ability to bind and transactivate target genes and consequently causes moderate cancer susceptibility. We have generated different syngeneically transplanted and autochthonous mouse models of p53-deficient acute myeloid leukemia and B or T cell lymphoma. After cancer manifestation we have activated E177R expression and analyzed the in vivo therapy response by bioluminescence or magnetic resonance imaging. The molecular response was further characterized in vitro by assays for gene expression, proliferation, senescence, differentiation, apoptosis and clonogenic growth. Results We report the conceptually intriguing observation that the p53 variant E177R, which promotes de novo leukemia and lymphoma formation, inhibits proliferation and viability, induces immune cell infiltration and triggers cancer regression in vivo when introduced into p53-deficient leukemia and lymphomas. p53-deficient cancer cells proved to be so addicted to the absence of p53 that even the low-level activity of E177R is detrimental to cancer growth. Conclusions The observation that a partial loss-of-function p53 variant promotes tumorigenesis in one setting and induces regression in another, underlines the highly context-specific effects of individual p53 mutants. It further highlights the exquisite sensitivity of cancer cells to even small changes in p53 activity and reveals that changes in activity level are more important than the absolute level. As such, the study encourages ongoing research efforts into mutant p53 reactivating drugs by providing genetic proof-of-principle evidence that incomplete p53 reactivation may suffice to elicit a therapeutic response. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02269-6.
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Laura Meyer
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Michelle Neumann
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Alexander M König
- Clinic of Diagnostic and Interventional Radiology, Core Facility 7T-small animal MRI, Philipps-University, Marburg, Germany
| | - Nikolaos Ananikidis
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Corinna U Keber
- Institute for Pathology, University Hospital Marburg, Philipps-University, Marburg, Germany
| | - Sabrina Elmshäuser
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany. .,German Center for Lung Research (DZL), Philipps-University, Marburg, Germany.
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany. .,German Center for Lung Research (DZL), Philipps-University, Marburg, Germany. .,Genomics Core Facility, Philipps-University, Marburg, Germany.
| |
Collapse
|
18
|
Zheng H, Fu J, Chen Z, Yang G, Yuan G. Dlx3 Ubiquitination by Nuclear Mdm2 Is Essential for Dentinogenesis in Mice. J Dent Res 2022; 101:1064-1074. [PMID: 35220830 DOI: 10.1177/00220345221077202] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dentin is a major mineralized component of teeth. Odontoblasts are responsible for synthesis and secretion of dentin matrix. Previously, it has been demonstrated in a cell culture system that the E3 ubiquitin ligase, murine double minute 2 (Mdm2), promotes odontoblast-like differentiation of mouse dental papilla cells (mDPCs) by ubiquitinating p53 and the odontoblast-specific substrate Dlx3. However, whether Mdm2 plays an essential role in vivo in odontoblast differentiation and dentin formation remains unknown. In this study, we investigated the in vivo functions of Mdm2 using Dmp1-Cre;Mdm2 flox/flox mice combined with multiple histological and molecular biological methods. The results showed that Mdm2 deletion in the odontoblast layer led to defects in odontoblast differentiation and dentin formation. Unexpectedly, specific inhibition of the Mdm2-p53 axis in wild-type mice by injection of a small-molecule inhibitor Nutlin-3a indicated that the role of Mdm2 in dentinogenesis was p53 independent, which was inconsistent with the previous in vitro study. In situ proximity ligation assay (PLA) showed that Mdm2 interacted with and ubiquitinated Dlx3 in the odontoblast nucleus of mouse molars. Dlx3 promoted the translocation of Mdm2 to the nucleus, and in turn, the nuclear Mdm2 mediated ubiquitination of Dlx3 and promoted the odontoblast-like differentiation of mDPCs. Dlx3 interacted with Mdm2 through its C-terminal domain. Deletion of the C-terminal domain of Dlx3 reversed the enhanced odontoblast-like differentiation and the activation of Dspp promoter mediated by overexpression of wild-type or nuclear Mdm2. Our findings suggest that nuclear Mdm2 mediates ubiquitination of the transcription factor Dlx3, which is essential for Dlx3 transcriptional activity on Dspp as well as subsequent odontoblast differentiation and dentin formation.
Collapse
Affiliation(s)
- H. Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - J. Fu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Z. Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - G. Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - G. Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Klimovich B, Merle N, Neumann M, Elmshäuser S, Nist A, Mernberger M, Kazdal D, Stenzinger A, Timofeev O, Stiewe T. p53 partial loss-of-function mutations sensitize to chemotherapy. Oncogene 2022; 41:1011-1023. [PMID: 34907344 PMCID: PMC8837531 DOI: 10.1038/s41388-021-02141-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/23/2022]
Abstract
The tumor suppressive transcription factor p53 is frequently inactivated in cancer cells by missense mutations that cluster in the DNA binding domain. 30% hit mutational hotspot residues, resulting in a complete loss of transcriptional activity and mutant p53-driven chemotherapy resistance. Of the remaining 70% of non-hotspot mutants, many are partial loss-of-function (partial-LOF) mutants with residual transcriptional activity. The therapeutic consequences of a partial-LOF have remained largely elusive. Using a p53 mutation engineered to reduce DNA binding, we demonstrate that partial-LOF is sufficient to enhance oncogene-driven tumorigenesis in mouse models of lung and pancreatic ductal adenocarcinoma and acute myeloid leukemia. Interestingly, mouse and human tumors with partial-LOF mutations showed mutant p53 protein accumulation similar as known for hotspot mutants. Different from the chemotherapy resistance caused by p53-loss, the partial-LOF mutant sensitized to an apoptotic chemotherapy response and led to a survival benefit. Mechanistically, the pro-apoptotic transcriptional activity of mouse and human partial-LOF mutants was rescued at high mutant protein levels, suggesting that accumulation of partial-LOF mutants enables the observed apoptotic chemotherapy response. p53 non-hotspot mutants with partial-LOF, therefore, represent tumorigenic p53 mutations that need to be distinguished from other mutations because of their beneficial impact on survival in a therapy context.
Collapse
MESH Headings
- Animals
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Humans
- Mice
- Loss of Function Mutation
- Drug Resistance, Neoplasm/genetics
- Apoptosis/genetics
- Apoptosis/drug effects
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Michelle Neumann
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Sabrina Elmshäuser
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps-University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany.
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany.
- Genomics Core Facility, Philipps-University, Marburg, Germany.
| |
Collapse
|
20
|
Li G, Zhu Q, Wang B, Luo R, Xiao X, Zhang Y, Ma L, Feng X, Huang J, Sun X, Wen Z, Pan Y, Yang C. Rejuvenation of Senescent Bone Marrow Mesenchymal Stromal Cells by Pulsed Triboelectric Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100964. [PMID: 34258884 PMCID: PMC8456218 DOI: 10.1002/advs.202100964] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 05/11/2023]
Abstract
Stem cell senescence contributes to stem cell exhaustion and drives various aging-associated disorders. However, strategies to rejuvenate senescent stem cells are limited. The present study proposes an approach based on triboelectric stimulation to rejuvenate senescent bone marrow mesenchymal stromal cells (BMSCs) by fabricating a pulsed triboelectric nanogenerator (P-TENG) that can produce stable pulsed current output unaffected by the triggered frequency. The senescence phenotypes of aged BMSCs are reversed by triboelectric stimulation at 30 µA at 1.5 Hz. Triboelectric stimulation enhances the proliferation of aged BMSCs and increases their pluripotency and differentiation capacity. Additionally, mechanistic investigations reveal that pulsed triboelectric stimulation by P-TENG rejuvenates senescent BMSCs by enhancing MDM2-dependent p53 degradation, which is demonstrated by loss-of-function studies of MDM2 and p53. Overall, this study identifies a new approach for the rejuvenation of senescent BMSCs and describes a promising therapeutic intervention for many diseases associated with aged BMSCs.
Collapse
Affiliation(s)
- Gaocai Li
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Qianqian Zhu
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhou215123China
| | - Bingjin Wang
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Rongjin Luo
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xiaohui Xiao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineDepartment of CardiologyMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
| | - Yi Zhang
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhou215123China
| | - Liang Ma
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xiaobo Feng
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jingang Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineDepartment of CardiologyMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
| | - Xuhui Sun
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhou215123China
| | - Zhen Wen
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhou215123China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineDepartment of CardiologyMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
| | - Cao Yang
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
21
|
Kruta M, Sunshine MJ, Chua BA, Fu Y, Chawla A, Dillingham CH, Hidalgo San Jose L, De Jong B, Zhou FJ, Signer RAJ. Hsf1 promotes hematopoietic stem cell fitness and proteostasis in response to ex vivo culture stress and aging. Cell Stem Cell 2021; 28:1950-1965.e6. [PMID: 34388375 DOI: 10.1016/j.stem.2021.07.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 05/18/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022]
Abstract
Maintaining proteostasis is key to resisting stress and promoting healthy aging. Proteostasis is necessary to preserve stem cell function, but little is known about the mechanisms that regulate proteostasis during stress in stem cells, and whether disruptions of proteostasis contribute to stem cell aging is largely unexplored. We determined that ex-vivo-cultured mouse and human hematopoietic stem cells (HSCs) rapidly increase protein synthesis. This challenge to HSC proteostasis was associated with nuclear accumulation of Hsf1, and deletion of Hsf1 impaired HSC maintenance ex vivo. Strikingly, supplementing cultures with small molecules that enhance Hsf1 activation partially suppressed protein synthesis, rebalanced proteostasis, and supported retention of HSC serial reconstituting activity. Although Hsf1 was dispensable for young adult HSCs in vivo, Hsf1 deficiency increased protein synthesis and impaired the reconstituting activity of middle-aged HSCs. Hsf1 thus promotes proteostasis and the regenerative activity of HSCs in response to culture stress and aging.
Collapse
Affiliation(s)
- Miriama Kruta
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mary Jean Sunshine
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bernadette A Chua
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yunpeng Fu
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ashu Chawla
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Christopher H Dillingham
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA; La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Lorena Hidalgo San Jose
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bijou De Jong
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fanny J Zhou
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert A J Signer
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
22
|
Bowen ME, Mulligan AS, Sorayya A, Attardi LD. Puma- and Caspase9-mediated apoptosis is dispensable for p53-driven neural crest-based developmental defects. Cell Death Differ 2021; 28:2083-2094. [PMID: 33574585 PMCID: PMC8257737 DOI: 10.1038/s41418-021-00738-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/20/2023] Open
Abstract
Inappropriate activation of the p53 transcription factor is thought to contribute to the developmental phenotypes in a range of genetic syndromes. Whether p53 activation drives these developmental phenotypes by triggering apoptosis, cell cycle arrest, or other p53 cellular responses, however, has remained elusive. As p53 hyperactivation in embryonic neural crest cells (NCCs) drives a number of phenotypes, including abnormal craniofacial and neuronal development, we investigate the basis for p53 action in this context. We show that p53-driven developmental defects are associated with the induction of a robust pro-apoptotic transcriptional signature. Intriguingly, however, deleting Puma or Caspase9, which encode key components of the intrinsic apoptotic pathway, does not rescue craniofacial, neuronal or pigmentation defects triggered by p53 hyperactivation in NCCs. Immunostaining analyses for two key apoptosis markers confirm that deleting Puma or Caspase9 does indeed impair p53-hyperactivation-induced apoptosis in NCCs. Furthermore, we demonstrate that p53 hyperactivation does not trigger a compensatory dampening of cell cycle progression in NCCs upon inactivation of apoptotic pathways. Together, our results indicate that p53-driven craniofacial, neuronal and pigmentation defects can arise in the absence of apoptosis and cell cycle arrest, suggesting that p53 hyperactivation can act via alternative pathways to trigger developmental phenotypes.
Collapse
Affiliation(s)
- Margot E Bowen
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Abigail S Mulligan
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Aryo Sorayya
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
23
|
Radiation-induced bystander effects impair transplanted human hematopoietic stem cells via oxidative DNA damage. Blood 2021; 137:3339-3350. [PMID: 33881475 DOI: 10.1182/blood.2020007362] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Total body irradiation (TBI) is commonly used in host conditioning regimens for human hematopoietic stem cell (HSC) transplantation to treat various hematological disorders. Exposure to TBI not only induces acute myelosuppression and immunosuppression, but also injures the various components of the HSC niche in recipients. Our previous study demonstrated that radiation-induced bystander effects (RIBE) of irradiated recipients decreased the long-term repopulating ability of transplanted mouse HSCs. However, RIBE on transplanted human HSCs have not been studied. Here, we report that RIBE impaired the long-term hematopoietic reconstitution of human HSCs as well as the colony-forming ability of human hematopoietic progenitor cells (HPCs). Our further analyses revealed that the RIBE-affected human hematopoietic cells showed enhanced DNA damage responses, cell-cycle arrest, and p53-dependent apoptosis, mainly because of oxidative stress. Moreover, multiple antioxidants could mitigate these bystander effects, though at different efficacies in vitro and in vivo. Taken together, these findings suggest that RIBE impair human HSCs and HPCs by oxidative DNA damage. This study provides definitive evidence for RIBE on transplanted human HSCs and further justifies the necessity of conducting clinical trials to evaluate different antioxidants to improve the efficacy of HSC transplantation for the patients with hematological or nonhematological disorders.
Collapse
|
24
|
Fujino T, Goyama S, Sugiura Y, Inoue D, Asada S, Yamasaki S, Matsumoto A, Yamaguchi K, Isobe Y, Tsuchiya A, Shikata S, Sato N, Morinaga H, Fukuyama T, Tanaka Y, Fukushima T, Takeda R, Yamamoto K, Honda H, Nishimura EK, Furukawa Y, Shibata T, Abdel-Wahab O, Suematsu M, Kitamura T. Mutant ASXL1 induces age-related expansion of phenotypic hematopoietic stem cells through activation of Akt/mTOR pathway. Nat Commun 2021; 12:1826. [PMID: 33758188 PMCID: PMC7988019 DOI: 10.1038/s41467-021-22053-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/23/2021] [Indexed: 01/31/2023] Open
Abstract
Somatic mutations of ASXL1 are frequently detected in age-related clonal hematopoiesis (CH). However, how ASXL1 mutations drive CH remains elusive. Using knockin (KI) mice expressing a C-terminally truncated form of ASXL1-mutant (ASXL1-MT), we examined the influence of ASXL1-MT on physiological aging in hematopoietic stem cells (HSCs). HSCs expressing ASXL1-MT display competitive disadvantage after transplantation. Nevertheless, in genetic mosaic mouse model, they acquire clonal advantage during aging, recapitulating CH in humans. Mechanistically, ASXL1-MT cooperates with BAP1 to deubiquitinate and activate AKT. Overactive Akt/mTOR signaling induced by ASXL1-MT results in aberrant proliferation and dysfunction of HSCs associated with age-related accumulation of DNA damage. Treatment with an mTOR inhibitor rapamycin ameliorates aberrant expansion of the HSC compartment as well as dysregulated hematopoiesis in aged ASXL1-MT KI mice. Our findings suggest that ASXL1-MT provokes dysfunction of HSCs, whereas it confers clonal advantage on HSCs over time, leading to the development of CH.
Collapse
Affiliation(s)
- Takeshi Fujino
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Susumu Goyama
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, and Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Suematsu Gas Biology Project, Shinjuku-ku, Tokyo Japan
| | - Daichi Inoue
- grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan−Kettering Cancer Center and Weill Cornell Medical College, New York, USA ,grid.417982.10000 0004 0623 246XDepartment of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe City, Hyogo Japan
| | - Shuhei Asada
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan ,grid.410818.40000 0001 0720 6587Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo Japan
| | - Satoshi Yamasaki
- grid.26999.3d0000 0001 2151 536XLaboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Akiko Matsumoto
- grid.26999.3d0000 0001 2151 536XLaboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Kiyoshi Yamaguchi
- grid.26999.3d0000 0001 2151 536XDivision of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Yumiko Isobe
- grid.26999.3d0000 0001 2151 536XDivision of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Akiho Tsuchiya
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Shiori Shikata
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Naru Sato
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Hironobu Morinaga
- grid.265073.50000 0001 1014 9130Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo Japan
| | - Tomofusa Fukuyama
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Yosuke Tanaka
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Tsuyoshi Fukushima
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Reina Takeda
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Keita Yamamoto
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Hiroaki Honda
- grid.410818.40000 0001 0720 6587Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo Japan
| | - Emi K. Nishimura
- grid.265073.50000 0001 1014 9130Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo Japan
| | - Yoichi Furukawa
- grid.26999.3d0000 0001 2151 536XDivision of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Tatsuhiro Shibata
- grid.26999.3d0000 0001 2151 536XLaboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| | - Omar Abdel-Wahab
- grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan−Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, and Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Suematsu Gas Biology Project, Shinjuku-ku, Tokyo Japan
| | - Toshio Kitamura
- grid.26999.3d0000 0001 2151 536XDivision of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo Japan
| |
Collapse
|
25
|
Chen Y, Luo X, Zou Z, Liang Y. The Role of Reactive Oxygen Species in Tumor Treatment and its Impact on Bone Marrow Hematopoiesis. Curr Drug Targets 2021; 21:477-498. [PMID: 31736443 DOI: 10.2174/1389450120666191021110208] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/21/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS), an important molecule inducing oxidative stress in organisms, play a key role in tumorigenesis, tumor progression and recurrence. Recent findings on ROS have shown that ROS can be used to treat cancer as they accelerate the death of tumor cells. At present, pro-oxidant drugs that are intended to increase ROS levels of the tumor cells have been widely used in the clinic. However, ROS are a double-edged sword in the treatment of tumors. High levels of ROS induce not only the death of tumor cells but also oxidative damage to normal cells, especially bone marrow hemopoietic cells, which leads to bone marrow suppression and (or) other side effects, weak efficacy of tumor treatment and even threatening patients' life. How to enhance the killing effect of ROS on tumor cells while avoiding oxidative damage to the normal cells has become an urgent issue. This study is a review of the latest progress in the role of ROS-mediated programmed death in tumor treatment and prevention and treatment of oxidative damage in bone marrow induced by ROS.
Collapse
Affiliation(s)
- Yongfeng Chen
- Taizhou University Hosipital, Taizhou University, Taizhou, 318000, Zhejiang, China.,Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Xingjing Luo
- Taizhou University Hosipital, Taizhou University, Taizhou, 318000, Zhejiang, China.,Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Zhenyou Zou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Yong Liang
- Taizhou University Hosipital, Taizhou University, Taizhou, 318000, Zhejiang, China.,Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, 318000, Zhejiang, China
| |
Collapse
|
26
|
Wang T, Xia C, Weng Q, Wang K, Dong Y, Hao S, Dong F, Liu X, Liu L, Geng Y, Guan Y, Du J, Cheng T, Cheng H, Wang J. Loss of <i>Nupr1</i> promotes engraftment by tuning the quiescence threshold of hematopoietic stem cell repository via regulating p53-checkpoint pathway. Haematologica 2020; 107:154-166. [PMID: 33299232 PMCID: PMC8719103 DOI: 10.3324/haematol.2019.239186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cells (HSC) are dominantly quiescent under homeostasis, which is a key mechanism of maintaining the HSC pool for life-long hematopoiesis. Dormant HSC are poised to be immediately activated in certain conditions and can return to quiescence after homeostasis has been regained. At present, the molecular networks of regulating the threshold of HSC dormancy, if existing, remain largely unknown. Here, we show that deletion of Nupr1, a gene preferentially expressed in HSC, activated quiescent HSC under homeostasis, which conferred a competitive engraftment advantage for these HSC without compromising their stemness or multi-lineage differentiation capacity in serial transplantation settings. Following an expansion protocol, the Nupr1-/- HSC proliferated more robustly than their wild-type counterparts in vitro. Nupr1 inhibits the expression of p53 and rescue of this inhibition offsets the engraftment advantage. Our data reveal a new role for Nupr1 as a regulator of HSC quiescence, which provides insights for accelerating the engraftment efficacy of HSC transplantation by targeting the HSC quiescence-controlling network.
Collapse
Affiliation(s)
- Tongjie Wang
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China; Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Chengxiang Xia
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Qitong Weng
- State Key Laboratory of Experimental Hematology and National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Kaitao Wang
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Yong Dong
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Sha Hao
- State Key Laboratory of Experimental Hematology and National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Fang Dong
- State Key Laboratory of Experimental Hematology and National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Xiaofei Liu
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Lijuan Liu
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Yang Geng
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Yuxian Guan
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Juan Du
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology and National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology and National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin.
| | - Jinyong Wang
- State Key Laboratory of Experimental Hematology, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China; Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou.
| |
Collapse
|
27
|
Timofeev O, Koch L, Niederau C, Tscherne A, Schneikert J, Klimovich M, Elmshäuser S, Zeitlinger M, Mernberger M, Nist A, Osterburg C, Dötsch V, Hrabé de Angelis M, Stiewe T. Phosphorylation Control of p53 DNA-Binding Cooperativity Balances Tumorigenesis and Aging. Cancer Res 2020; 80:5231-5244. [PMID: 32873634 DOI: 10.1158/0008-5472.can-20-2002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 11/16/2022]
Abstract
Posttranslational modifications are essential for regulating the transcription factor p53, which binds DNA in a highly cooperative manner to control expression of a plethora of tumor-suppressive programs. Here we show at the biochemical, cellular, and organismal level that the cooperative nature of DNA binding is reduced by phosphorylation of highly conserved serine residues (human S183/S185, mouse S180) in the DNA-binding domain. To explore the role of this inhibitory phosphorylation in vivo, new phosphorylation-deficient p53-S180A knock-in mice were generated. Chromatin immunoprecipitation sequencing and RNA sequencing studies of S180A knock-in cells demonstrated enhanced DNA binding and increased target gene expression. In vivo, this translated into a tissue-specific vulnerability of the bone marrow that caused depletion of hematopoietic stem cells and impaired proper regeneration of hematopoiesis after DNA damage. Median lifespan was significantly reduced by 20% from 709 days in wild type to only 568 days in S180A littermates. Importantly, lifespan was reduced by a loss of general fitness and increased susceptibility to age-related diseases, not by increased cancer incidence as often seen in other p53-mutant mouse models. For example, S180A knock-in mice showed markedly reduced spontaneous tumorigenesis and increased resistance to Myc-driven lymphoma and Eml4-Alk-driven lung cancer. Preventing phosphorylation of S183/S185 in human cells boosted p53 activity and allowed tumor cells to be killed more efficiently. Together, our data identify p53 DNA-binding domain phosphorylation as a druggable mechanism that balances tumorigenesis and aging. SIGNIFICANCE: These findings demonstrate that p53 tumor suppressor activity is reduced by DNA-binding domain phosphorylation to prevent aging and identify this phosphorylation as a potential target for cancer therapy.See related commentary by Horikawa, p. 5164.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany.
| | - Lukas Koch
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Constantin Niederau
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Alina Tscherne
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Jean Schneikert
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Maria Klimovich
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Sabrina Elmshäuser
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Marie Zeitlinger
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps-University Marburg, Marburg, Germany
| | | | | | - Martin Hrabé de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany. .,Genomics Core Facility, Philipps-University Marburg, Marburg, Germany
| | | |
Collapse
|
28
|
Ikonomi N, Kühlwein SD, Schwab JD, Kestler HA. Awakening the HSC: Dynamic Modeling of HSC Maintenance Unravels Regulation of the TP53 Pathway and Quiescence. Front Physiol 2020; 11:848. [PMID: 32848827 PMCID: PMC7411231 DOI: 10.3389/fphys.2020.00848] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cells (HSCs) provide all types of blood cells during the entire life of the organism. HSCs are mainly quiescent and can eventually enter the cell cycle to differentiate. HSCs are maintained and tightly regulated in a particular environment. The stem cell niche regulates dormancy and awakening. Deregulations of this interplay can lead to hematopoietic failure and diseases. In this paper, we present a Boolean network model that recapitulates HSC regulation in virtue of external signals coming from the niche. This Boolean network integrates and summarizes the current knowledge of HSC regulation and is based on extensive literature research. Furthermore, dynamic simulations suggest a novel systemic regulation of TP53 in homeostasis. Thereby, our model indicates that TP53 activity is balanced depending on external stimulations, engaging a regulatory mechanism involving ROS regulators and RAS activated transcription factors. Finally, we investigated different mouse models and compared them to in silico knockout simulations. Here, the model could recapitulate in vivo observed behaviors and thus sustains our results.
Collapse
Affiliation(s)
- Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Silke D Kühlwein
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Julian D Schwab
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| |
Collapse
|
29
|
SOX2 and p53 Expression Control Converges in PI3K/AKT Signaling with Versatile Implications for Stemness and Cancer. Int J Mol Sci 2020; 21:ijms21144902. [PMID: 32664542 PMCID: PMC7402325 DOI: 10.3390/ijms21144902] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Stemness and reprogramming involve transcriptional master regulators that suppress cell differentiation while promoting self-renewal. A distinguished example thereof is SOX2, a high mobility group (HMG)-box transcription factor (TF), whose subcellular localization and turnover regulation in embryonic, induced-pluripotent, and cancer stem cells (ESCs, iPSCs, and CSCs, respectively) is mediated by the PI3K/AKT/SOX2 axis, a stem cell-specific branch of the PI3K/AKT signaling pathway. Further effector functions associated with PI3K/AKT induction include cell cycle progression, cellular (mass) growth, and the suppression of apoptosis. Apoptosis, however, is a central element of DNA damage response (DDR), where it provides a default mechanism for cell clearance when DNA integrity cannot be maintained. A key player in DDR is tumor suppressor p53, which accumulates upon DNA-damage and is counter-balanced by PI3K/AKT enforced turnover. Accordingly, stemness sustaining SOX2 expression and p53-dependent DDR mechanisms show molecular–functional overlap in PI3K/AKT signaling. This constellation proves challenging for stem cells whose genomic integrity is a functional imperative for normative ontogenesis. Unresolved mutations in stem and early progenitor cells may in fact provoke transformation and cancer development. Such mechanisms are also particularly relevant for iPSCs, where genetic changes imposed through somatic cell reprogramming may promote DNA damage. The current review aims to summarize the latest advances in the understanding of PI3K/AKT/SOX2-driven stemness and its intertwined relations to p53-signaling in DDR under conditions of pluripotency, reprogramming, and transformation.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Protein homeostasis (proteostasis) is maintained by an integrated network of physiological mechanisms and stress response pathways that regulate the content and quality of the proteome. Maintenance of cellular proteostasis is key to ensuring normal development, resistance to environmental stress, coping with infection, and promoting healthy aging and lifespan. Recent studies have revealed that several proteostasis mechanisms can function in a cell-type-specific manner within hematopoietic stem cells (HSCs). Here, we review recent studies demonstrating that the proteostasis network functions uniquely in HSCs to promote their maintenance and regenerative function. RECENT FINDINGS The proteostasis network is regulated differently in HSCs as compared with restricted hematopoietic progenitors. Disruptions in proteostasis are particularly detrimental to HSC maintenance and function. These findings suggest that multiple aspects of cellular physiology are uniquely regulated in HSCs to maintain proteostasis, and that precise control of proteostasis is particularly important to support life-long HSC maintenance and regenerative function. SUMMARY The proteostasis network is uniquely configured within HSCs to promote their longevity and hematopoietic function. Future work uncovering cell-type-specific differences in proteostasis network configuration, integration, and function will be essential for understanding how HSCs function during homeostasis, in response to stress, and in disease.
Collapse
Affiliation(s)
- Bernadette A Chua
- Department of Medicine, Division of Regenerative Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | | |
Collapse
|
31
|
Hidalgo San Jose L, Sunshine MJ, Dillingham CH, Chua BA, Kruta M, Hong Y, Hatters DM, Signer RAJ. Modest Declines in Proteome Quality Impair Hematopoietic Stem Cell Self-Renewal. Cell Rep 2020; 30:69-80.e6. [PMID: 31914399 PMCID: PMC7004491 DOI: 10.1016/j.celrep.2019.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/02/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023] Open
Abstract
Low protein synthesis is a feature of somatic stem cells that promotes regeneration in multiple tissues. Modest increases in protein synthesis impair stem cell function, but the mechanisms by which this occurs are largely unknown. We determine that low protein synthesis within hematopoietic stem cells (HSCs) is associated with elevated proteome quality in vivo. HSCs contain less misfolded and unfolded proteins than myeloid progenitors. Increases in protein synthesis cause HSCs to accumulate misfolded and unfolded proteins. To test how proteome quality affects HSCs, we examine Aarssti/sti mice that harbor a tRNA editing defect that increases amino acid misincorporation. Aarssti/sti mice exhibit reduced HSC numbers, increased proliferation, and diminished serial reconstituting activity. Misfolded proteins overwhelm the proteasome within Aarssti/sti HSCs, which is associated with increased c-Myc abundance. Deletion of one Myc allele partially rescues serial reconstitution defects in Aarssti/sti HSCs. Thus, HSCs are dependent on low protein synthesis to maintain proteostasis, which promotes their self-renewal.
Collapse
Affiliation(s)
- Lorena Hidalgo San Jose
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Mary Jean Sunshine
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher H Dillingham
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Bernadette A Chua
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Miriama Kruta
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Yuning Hong
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia
| | - Danny M Hatters
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robert A J Signer
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
32
|
Palmitic Acid Methyl Ester Induces G 2/M Arrest in Human Bone Marrow-Derived Mesenchymal Stem Cells via the p53/p21 Pathway. Stem Cells Int 2019; 2019:7606238. [PMID: 31885624 PMCID: PMC6915012 DOI: 10.1155/2019/7606238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/14/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022] Open
Abstract
Bone marrow-derived mesenchymal cells (BM-MSCs) are able to differentiate into adipocytes, which can secrete adipokines to affect BM-MSC proliferation and differentiation. Recent evidences indicated that adipocytes can secrete fatty acid metabolites, such as palmitic acid methyl ester (PAME), which is able to cause vasorelaxation and exerts anti-inflammatory effects. However, effects of PAME on BM-MSC proliferation remain unclear. The aim of this study was to investigate the effect of PAME on human BM-MSC (hBM-MSC) proliferation and its underlying molecular mechanisms. hBM-MSCs were treated with PAME for 48 h and then subjected to various analyses. The results from the present study show that PAME significantly reduced the levels of G2/M phase regulatory proteins, cyclin-dependent kinase 1 (Cdk1), and cyclin B1 and inhibited proliferation in hBM-MSCs. Moreover, the level of Mdm2 protein decreased, while the levels of p21 and p53 protein increased in the PAME-treated hBM-MSCs. However, PAME treatment did not significantly affect apoptosis/necrosis, ROS generation, and the level of Cdc25C protein. PAME also induced intracellular acidosis and increased intracellular Ca2+ levels. Cotreatment with PAME and Na+/H+ exchanger inhibitors together further reduced the intracellular pH but did not affect the PAME-induced decreases of cell proliferation and increases of the cell population at the G2/M phase. Cotreatment with PAME and a calcium chelator together inhibited the PAME-increased intracellular Ca2+ levels but did not affect the PAME-induced cell proliferation inhibition and G2/M cell cycle arrest. Moreover, the half-life of p53 protein was prolonged in the PAME-treated hBM-MSCs. Taken together, these results suggest that PAME induced p53 stabilization, which in turn increased the levels of p53/p21 proteins and decreased the levels of Cdk1/cyclin B1 proteins, thereby preventing the activation of Cdk1, and eventually caused cell cycle arrest at the G2/M phase. The findings from the present study might help get insight into the physiological roles of PAME in regulating hBM-MSC proliferation.
Collapse
|
33
|
Klimovich B, Stiewe T, Timofeev O. Inactivation of Mdm2 restores apoptosis proficiency of cooperativity mutant p53 in vivo. Cell Cycle 2019; 19:109-123. [PMID: 31749402 DOI: 10.1080/15384101.2019.1693748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
TP53 mutations are found in 50% of all cancers and mutated TP53 status is considered poor for treatment. However, some TP53 mutations exhibit only partial loss-of-function (LOF), meaning they retain residual transcriptional and non-transcriptional activities that are potentially beneficial for therapy. Earlier we have characterized a knock-in mouse model for the partial LOF mutant Trp53E177R (p53RR). Reduced DNA binding cooperativity of this mutant led to the loss of p53-dependent apoptosis, while p53 functions in cell cycle control, senescence, metabolism, and antioxidant defense remained intact. Concomitantly, tumor suppression was evident but strongly compromised compared to wild-type mice. Here we used the Trp53E177R mouse as a model to investigate whether residual functions of mutant p53 can be engaged to induce cell death, which is considered the most desirable outcome of tumor therapy. We made use of Mdm2 knock-out in developing embryos as a sensitive tool for detecting remaining p53 activities. Genetic ablation of Mdm2 led to embryonic lethality in Trp53E177R/E177R homozygotes at days 9.5-11.5. This effect was not rescued by concomitant p21-knockout, indicating its independence of p21-mediated cell cycle arrest. Instead, immunohistochemical analysis showed widespread apoptosis in tissues of defective embryos accompanied by persistent accumulation of p53RR protein. This led to partial restoration of the mutant's proficiency in transcriptional induction of the pro-apoptotic genes Bbc3 (Puma) and Bax. These data indicate that increased quantity can compensate for qualitative defects of p53 mutants and suggest that Mdm2-targeting (potentially in combination with other drugs) might be effective against cells bearing p53 partial LOF mutants.
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| |
Collapse
|
34
|
Singh S, Vaughan CA, Rabender C, Mikkelsen R, Deb S, Palit Deb S. DNA replication in progenitor cells and epithelial regeneration after lung injury requires the oncoprotein MDM2. JCI Insight 2019; 4:128194. [PMID: 31527309 PMCID: PMC6824310 DOI: 10.1172/jci.insight.128194] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022] Open
Abstract
Depletion of epithelial cells after lung injury prompts proliferation and epithelial mesenchymal transition (EMT) of progenitor cells, and this repopulates the lost epithelial layer. To investigate the cell proliferative function of human oncoprotein MDM2, we generated mouse models targeting human MDM2 expression in either lung Club or alveolar cells after doxycycline treatment. We report that MDM2 expression in lung Club or alveolar cells activates DNA replication specifically in lung progenitor cells only after chemical- or radiation-induced lung injury, irrespective of their p53 status. Activation of DNA replication by MDM2 triggered by injury leads to proliferation of lung progenitor cells and restoration of the lost epithelial layers. Mouse lung with no Mdm2 allele loses its ability to replicate DNA, whereas loss of 1 Mdm2 allele compromises this function, demonstrating the requirement of endogenous MDM2. We show that the p53-independent ability of MDM2 to activate Akt signaling is essential for initiating DNA replication in lung progenitor cells. Furthermore, MDM2 activates the Notch signaling pathway and expression of EMT markers, indicative of epithelial regeneration. This is the first report to our knowledge demonstrating a direct p53-independent participation of MDM2 in progenitor cell proliferation and epithelial repair after lung injury, distinct from a p53-degrading antiapoptotic effect preventing injury.
Collapse
Affiliation(s)
- Shilpa Singh
- Department of Biochemistry and Molecular Biology
- VCU Massey Cancer Center, and
| | | | - Christopher Rabender
- VCU Massey Cancer Center, and
- Department of Radiation Oncology, Virginia Commonwealth, University, Richmond, Virginia, USA
| | - Ross Mikkelsen
- VCU Massey Cancer Center, and
- Department of Radiation Oncology, Virginia Commonwealth, University, Richmond, Virginia, USA
| | - Sumitra Deb
- Department of Biochemistry and Molecular Biology
- VCU Massey Cancer Center, and
| | - Swati Palit Deb
- Department of Biochemistry and Molecular Biology
- VCU Massey Cancer Center, and
| |
Collapse
|
35
|
Mattes K, Vellenga E, Schepers H. Differential redox-regulation and mitochondrial dynamics in normal and leukemic hematopoietic stem cells: A potential window for leukemia therapy. Crit Rev Oncol Hematol 2019; 144:102814. [PMID: 31593878 DOI: 10.1016/j.critrevonc.2019.102814] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/12/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
The prognosis for many patients with acute myeloid leukemia (AML) is poor, mainly due to disease relapse driven by leukemia stem cells (LSCs). Recent studies have highlighted the unique metabolic properties of LSCs, which might represent opportunities for LSC-selective targeting. LSCs characteristically have low levels of reactive oxygen species (ROS), which apparently result from a combination of low mitochondrial activity and high activity of ROS-removing pathways such as autophagy. Due to this low activity, LSCs are highly dependent on mitochondrial regulatory mechanisms. These include the anti-apoptotic protein BCL-2, which also has crucial roles in regulating the mitochondrial membrane potential, and proteins involved in mitophagy. Here we review the different pathways that impact mitochondrial activity and redox-regulation, and highlight their relevance for the functionality of both HSCs and LSCs. Additionally, novel AML therapy strategies that are based on interference with those pathways, including the promising BCL-2 inhibitor Venetoclax, are summarized.
Collapse
Affiliation(s)
- Katharina Mattes
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Edo Vellenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hein Schepers
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
36
|
Carvajal LA, Neriah DB, Senecal A, Benard L, Thiruthuvanathan V, Yatsenko T, Narayanagari SR, Wheat JC, Todorova TI, Mitchell K, Kenworthy C, Guerlavais V, Annis DA, Bartholdy B, Will B, Anampa JD, Mantzaris I, Aivado M, Singer RH, Coleman RA, Verma A, Steidl U. Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia. Sci Transl Med 2019; 10:10/436/eaao3003. [PMID: 29643228 DOI: 10.1126/scitranslmed.aao3003] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/12/2018] [Accepted: 03/23/2018] [Indexed: 12/14/2022]
Abstract
The tumor suppressor p53 is often inactivated via its interaction with endogenous inhibitors mouse double minute 4 homolog (MDM4 or MDMX) or mouse double minute 2 homolog (MDM2), which are frequently overexpressed in patients with acute myeloid leukemia (AML) and other cancers. Pharmacological disruption of both of these interactions has long been sought after as an attractive strategy to fully restore p53-dependent tumor suppressor activity in cancers with wild-type p53. Selective targeting of this pathway has thus far been limited to MDM2-only small-molecule inhibitors, which lack affinity for MDMX. We demonstrate that dual MDMX/MDM2 inhibition with a stapled α-helical peptide (ALRN-6924), which has recently entered phase I clinical testing, produces marked antileukemic effects. ALRN-6924 robustly activates p53-dependent transcription at the single-cell and single-molecule levels and exhibits biochemical and molecular biological on-target activity in leukemia cells in vitro and in vivo. Dual MDMX/MDM2 inhibition by ALRN-6924 inhibits cellular proliferation by inducing cell cycle arrest and apoptosis in cell lines and primary AML patient cells, including leukemic stem cell-enriched populations, and disrupts functional clonogenic and serial replating capacity. Furthermore, ALRN-6924 markedly improves survival in AML xenograft models. Our study provides mechanistic insight to support further testing of ALRN-6924 as a therapeutic approach in AML and other cancers with wild-type p53.
Collapse
Affiliation(s)
- Luis A Carvajal
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Daniela Ben Neriah
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Adrien Senecal
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lumie Benard
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Tatyana Yatsenko
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Swathi-Rao Narayanagari
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Justin C Wheat
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tihomira I Todorova
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Charles Kenworthy
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Boris Bartholdy
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Britta Will
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Division of Hemato-Oncology, Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jesus D Anampa
- Division of Hemato-Oncology, Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ioannis Mantzaris
- Division of Hemato-Oncology, Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Robert H Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Robert A Coleman
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Amit Verma
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Division of Hemato-Oncology, Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA. .,Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Division of Hemato-Oncology, Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
37
|
Timofeev O, Klimovich B, Schneikert J, Wanzel M, Pavlakis E, Noll J, Mutlu S, Elmshäuser S, Nist A, Mernberger M, Lamp B, Wenig U, Brobeil A, Gattenlöhner S, Köhler K, Stiewe T. Residual apoptotic activity of a tumorigenic p53 mutant improves cancer therapy responses. EMBO J 2019; 38:e102096. [PMID: 31483066 PMCID: PMC6792016 DOI: 10.15252/embj.2019102096] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53−/− mice. Surprisingly, stabilization of p53R178E in Mdm2−/− mice nevertheless triggers extensive apoptosis, indicative of residual wild‐type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E;Mdm2−/− embryos, it proves insufficient for suppression of spontaneous and oncogene‐driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53−/− mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53−/− ones, resulting in enhanced eradication of p53‐mutated tumor cells. Together, this provides genetic proof‐of‐principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Boris Klimovich
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Jean Schneikert
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany
| | | | - Julia Noll
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Samet Mutlu
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Boris Lamp
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Ulrich Wenig
- Institute of Pathology, Justus Liebig University, Giessen, Germany
| | | | | | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University, Giessen, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,Genomics Core Facility, Philipps University, Marburg, Germany
| |
Collapse
|
38
|
Hirata Y, Kakiuchi M, Robson SC, Fujisaki J. CD150 high CD4 T cells and CD150 high regulatory T cells regulate hematopoietic stem cell quiescence via CD73. Haematologica 2019; 104:1136-1142. [PMID: 30545927 PMCID: PMC6545860 DOI: 10.3324/haematol.2018.198283] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/10/2018] [Indexed: 12/31/2022] Open
Abstract
Various extrinsic signals tightly control hematopoietic stem cell quiescence. Our recent study showed that hematopoietic stem cells are regulated by a special FoxP3+ regulatory T-cell population with high expression of a hematopoietic stem cell marker, CD150. Extracellular adenosine generated via a cell-surface ectoenzyme CD39 on CD150high regulatory T cells maintained hematopoietic stem cell quiescence. It remains unclear how conventional T cells and the other cell-surface ectoenzyme, CD73, contribute to regulation of hematopoietic stem cells. This work shows that CD150high regulatory T cells as well as unique CD150high CD4+ conventional T cells regulate hematopoietic stem cells via CD73. Global CD73 deletion increased the numbers of hematopoietic stem cells, cycling stem cell frequencies, and levels of reactive oxygen species in hematopoietic stem cells. In vivo antioxidant treatment inhibited the increase of hematopoietic stem cells in CD73 knockout mice, suggesting that CD73 maintains stem cell quiescence by preventing oxidative stress. High levels of CD73 expression were frequently found on CD150high regulatory T cells and CD150high FoxP3-CD4+ T cells within the bone marrow. Transfer of these CD150high regulatory T cells and CD150high CD4+ conventional T cells abolished the increase of hematopoietic stem cells in CD73 knockout mice. In addition, the increase of stem cells in CD73 knockout mice was also inhibited by pharmacological activation of adenosine receptor 2A which is highly expressed by hematopoietic stem cells. Taken together, these results suggest that CD73 of CD150high regulatory T cells and CD150high CD4+ conventional T cells protects hematopoietic stem cells from oxidative stress, maintaining stem cell quiescence via adenosine receptor 2A.
Collapse
Affiliation(s)
- Yuichi Hirata
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
- MSD K.K., Tokyo, Japan
| | - Miwako Kakiuchi
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Simon C Robson
- Department of Medicine, Liver Center and Transplantation Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joji Fujisaki
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Department of Pediatrics, Division of Hematology and Oncology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
39
|
Cho W, Jin X, Pang J, Wang Y, Mivechi NF, Moskophidis D. The Molecular Chaperone Heat Shock Protein 70 Controls Liver Cancer Initiation and Progression by Regulating Adaptive DNA Damage and Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Signaling Pathways. Mol Cell Biol 2019; 39:e00391-18. [PMID: 30745413 PMCID: PMC6469921 DOI: 10.1128/mcb.00391-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/21/2018] [Accepted: 02/04/2019] [Indexed: 02/06/2023] Open
Abstract
Delineating the mechanisms that drive hepatic injury and hepatocellular carcinoma (HCC) progression is critical for development of novel treatments for recurrent and advanced HCC but also for the development of diagnostic and preventive strategies. Heat shock protein 70 (HSP70) acts in concert with several cochaperones and nucleotide exchange factors and plays an essential role in protein quality control that increases survival by protecting cells against environmental stressors. Specifically, the HSP70-mediated response has been implicated in the pathogenesis of cancer, but the specific mechanisms by which HSP70 may support malignant cell transformation remains to be fully elucidated. Here, we show that genetic ablation of HSP70 markedly impairs HCC initiation and progression by distinct but overlapping pathways. This includes the potentiation of the carcinogen-induced DNA damage response, at the tumor initiation stage, to increase the p53-dependent surveillance response leading to the cell cycle exit or death of genomically damaged differentiated pericentral hepatocytes, and this may also prevent their conversion into more proliferating HCC progenitor cells. Subsequently, activation of a mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) negative feedback pathway diminishes oncogenic signals, thereby attenuating premalignant cell transformation and tumor progression. Modulation of HSP70 function may be a strategy for interfering with oncogenic signals driving liver cell transformation and tumor progression, thus providing an opportunity for human cancer control.
Collapse
Affiliation(s)
- Wonkyoung Cho
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Xiongjie Jin
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Junfeng Pang
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Yan Wang
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Nahid F Mivechi
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
- Department of Radiology and Imaging, Augusta University, Augusta, Georgia, USA
| | - Demetrius Moskophidis
- Molecular Chaperone Biology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
- Department of Medicine, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
40
|
Olivos DJ, Perrien DS, Hooker A, Cheng YH, Fuchs RK, Hong JM, Bruzzaniti A, Chun K, Eischen CM, Kacena MA, Mayo LD. The proto-oncogene function of Mdm2 in bone. J Cell Biochem 2018; 119:8830-8840. [PMID: 30011084 DOI: 10.1002/jcb.27133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 05/07/2018] [Indexed: 12/19/2022]
Abstract
Mouse double minute 2 (Mdm2) is a multifaceted oncoprotein that is highly regulated with distinct domains capable of cellular transformation. Loss of Mdm2 is embryonically lethal, making it difficult to study in a mouse model without additional genetic alterations. Global overexpression through increased Mdm2 gene copy number (Mdm2Tg ) results in the development of hematopoietic neoplasms and sarcomas in adult animals. In these mice, we found an increase in osteoblastogenesis, differentiation, and a high bone mass phenotype. Since it was difficult to discern the cell lineage that generated this phenotype, we generated osteoblast-specific Mdm2 overexpressing (Mdm2TgOb ) mice in 2 different strains, C57BL/6 and DBA. These mice did not develop malignancies; however, these animals and the MG63 human osteosarcoma cell line with high levels of Mdm2 showed an increase in bone mineralization. Importantly, overexpression of Mdm2 corrected age-related bone loss in mice, providing a role for the proto-oncogenic activity of Mdm2 in bone health of adult animals.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Daniel S Perrien
- Departments of Medicine and Orthopaedic Surgery and Rehabilitation, Vanderbilt University Medical Center, and Tennessee Valley Healthcare System, Nashville, Tennessee.,Department of Veterans Affairs, Nashville, Tennessee
| | - Adam Hooker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ying-Hua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Robyn K Fuchs
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, Indiana
| | - Jung Min Hong
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Angela Bruzzaniti
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Kristin Chun
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christine M Eischen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lindsey D Mayo
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
41
|
Prieto-Bermejo R, Romo-González M, Pérez-Fernández A, Ijurko C, Hernández-Hernández Á. Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:125. [PMID: 29940987 PMCID: PMC6019308 DOI: 10.1186/s13046-018-0797-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/15/2018] [Indexed: 02/08/2023]
Abstract
Oxidative stress is related to ageing and degenerative diseases, including cancer. However, a moderate amount of reactive oxygen species (ROS) is required for the regulation of cellular signalling and gene expression. A low level of ROS is important for maintaining quiescence and the differentiation potential of haematopoietic stem cells (HSCs), whereas the level of ROS increases during haematopoietic differentiation; thus, suggesting the importance of redox signalling in haematopoiesis. Here, we will analyse the importance of ROS for haematopoiesis and include evidence showing that cells from leukaemia patients live under oxidative stress. The potential sources of ROS will be described. Finally, the level of oxidative stress in leukaemic cells can also be harnessed for therapeutic purposes. In this regard, the reliance of front-line anti-leukaemia chemotherapeutics on increased levels of ROS for their mechanism of action, as well as the active search for novel compounds that modulate the redox state of leukaemic cells, will be analysed.
Collapse
Affiliation(s)
- Rodrigo Prieto-Bermejo
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Marta Romo-González
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Alejandro Pérez-Fernández
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Carla Ijurko
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Ángel Hernández-Hernández
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain. .,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain.
| |
Collapse
|
42
|
Stiewe T, Haran TE. How mutations shape p53 interactions with the genome to promote tumorigenesis and drug resistance. Drug Resist Updat 2018; 38:27-43. [PMID: 29857816 DOI: 10.1016/j.drup.2018.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 12/31/2022]
Abstract
The tumor suppressive transcription factor p53 regulates a wide array of cellular processes that confer upon cells an essential protection against cancer development. Wild-type p53 regulates gene expression by directly binding to DNA in a sequence-specific manner. p53 missense mutations are the most common mutations in malignant cells and can be regarded as synonymous with anticancer drug resistance and poor prognosis. The current review provides an overview of how the extraordinary variety of more than 2000 different mutant p53 proteins, known as the p53 mutome, affect the interaction of p53 with DNA. We discuss how the classification of p53 mutations to loss of function (LOF), gain of function (GOF), and dominant-negative (DN) inhibition of a remaining wild-type allele, hides a complex p53 mutation spectrum that depends on the distinctive nature of each mutant protein, requiring different therapeutic strategies for each mutant p53 protein. We propose to regard the different mutant p53 categories as continuous variables, that may not be independent of each other. In particular, we suggest here to consider GOF mutations as a special subset of LOF mutations, especially when mutant p53 binds to DNA through cooperation with other transcription factors, and we present a model for GOF mechanism that consolidates many observations on the GOF phenomenon. We review how novel mutant p53 targeting approaches aim to restore a wild-type-like DNA interaction and to overcome resistance to cancer therapy.
Collapse
Affiliation(s)
- Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35037 Marburg, Germany.
| | - Tali E Haran
- Department of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 32000, Israel.
| |
Collapse
|
43
|
Reidy PT, Fry CS, Dickinson JM, Drummond MJ, Rasmussen BB. Postexercise essential amino acid supplementation amplifies skeletal muscle satellite cell proliferation in older men 24 hours postexercise. Physiol Rep 2018; 5:5/11/e13269. [PMID: 28596299 PMCID: PMC5471431 DOI: 10.14814/phy2.13269] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/29/2022] Open
Abstract
Aged skeletal muscle has an attenuated and delayed ability to proliferate satellite cells in response to resistance exercise. The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is a focal point for cell growth, however, the effect of postexercise mTORC1 activation on human skeletal muscle satellite cell (SC) proliferation is unknown. To test the proliferative capacity of skeletal muscle SC in aging muscle to a potent mTORC1 activator (i.e., EAA; essential amino acids) we recruited older (~72y) men to conduct leg resistance exercise (8setsx10reps) without (−EAA; n = 8) and with (+EAA: n = 11) ingestion of 10 g of EAA 1 h postexercise. Muscle biopsies were taken before exercise (Pre) and 24 h postexercise (Post) for assessment of expression and fiber type‐specific Pax7+SC, Ki67+Pax7+SC and MyoD+SC. −EAA did not show an increase in Pax7+ satellite cells at Post(P > 0.82). Although statistical significance for an increase in Pax7 + SC at 24 h post‐RE was not observed in +EAA versus −EAA, we observed trends for a treatment difference (P < 0.1). When examining the change from Pre to Post trends were demonstrated (#/myofiber: P = 0.076; and %/myonuclei: P = 0.065) for a greater increase in +EAA versus −EAA. Notably, we found an increase SC proliferation in +EAA, but not −EAA with increase in Ki67+SC and MyoD+ cells (P < 0.05). Ki67+SC also exhibited a significant group difference Post (P < 0.010). Pax7+SC in fast twitch myofibers did not change and were not different between groups (P > 0.10). CDK2, MEF2C, RB1 mRNA only increased in +EAA (P < 0.05). Acute muscle satellite cell proliferative capacity may be partially rescued with postexercise EAA ingestion in older men.
Collapse
Affiliation(s)
- Paul T Reidy
- Center for Recovery, Physical Activity and Nutrition, University of Texas Medical Branch, Galveston, Texas.,Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Jared M Dickinson
- Center for Recovery, Physical Activity and Nutrition, University of Texas Medical Branch, Galveston, Texas.,Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Micah J Drummond
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Blake B Rasmussen
- Center for Recovery, Physical Activity and Nutrition, University of Texas Medical Branch, Galveston, Texas .,Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
44
|
Chen Y, Liu K, Shi Y, Shao C. The tango of ROS and p53 in tissue stem cells. Cell Death Differ 2018; 25:639-641. [PMID: 29487352 PMCID: PMC5864234 DOI: 10.1038/s41418-018-0062-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 01/14/2023] Open
Affiliation(s)
- Youguo Chen
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Radioprotection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Keli Liu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Radioprotection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu, 215123, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Radioprotection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
45
|
Kon N, Wang D, Li T, Jiang L, Qiang L, Gu W. Inhibition of Mdmx (Mdm4) in vivo induces anti-obesity effects. Oncotarget 2018; 9:7282-7297. [PMID: 29484110 PMCID: PMC5800902 DOI: 10.18632/oncotarget.23837] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/08/2017] [Indexed: 01/22/2023] Open
Abstract
Although cell-cycle arrest, senescence and apoptosis remain as major canonical activities of p53 in tumor suppression, the emerging role of p53 in metabolism has been a topic of great interest. Nevertheless, it is not completely understood how p53-mediated metabolic activities are regulated in vivo and whether this part of the activities has an independent role beyond tumor suppression. Mdmx (also called Mdm4), like Mdm2, acts as a major suppressor of p53 but the embryonic lethality of mdmx-null mice creates difficulties to evaluate its physiological significance in metabolism. Here, we report that the embryonic lethality caused by the deficiency of mdmx, in contrast to the case for mdm2, is fully rescued in the background of p533KR/3KR, an acetylation-defective mutant unable to induce cell-cycle arrest, senescence and apoptosis. p533KR/3KR/mdmx-/- mice are healthy but skinny without obvious developmental defects. p533KR/3KR/mdmx-/- mice are resistant to fat accumulation in adipose tissues upon high fat diet. Notably, the levels of p53 protein are only slightly increased and can be further induced upon DNA damage in p533KR/3KR/mdmx-/- mice, suggesting that Mdmx is only partially required for p53 degradation in vivo. Further analyses indicate that the anti-obesity phenotypes in p533KR/3KR/mdmx-/- mice are caused by activation of lipid oxidation and thermogenic programs in adipose tissues. These results demonstrate the specific effects of the p53/Mdmx axis in lipid metabolism and adipose tissue remodeling and reveal a surprising role of Mdmx inhibition in anti-obesity effects beyond, commonly expected, tumor suppression. Thus, our study has significant implications regarding Mdmx inhibitors in the treatment of obesity related diseases.
Collapse
Affiliation(s)
- Ning Kon
- Institute for Cancer Genetics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Donglai Wang
- Institute for Cancer Genetics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Tongyuan Li
- Institute for Cancer Genetics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Le Jiang
- Institute for Cancer Genetics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Li Qiang
- Naomi Berrie Diabetes Center, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Wei Gu
- Institute for Cancer Genetics, College of Physicians and Surgeons of Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| |
Collapse
|
46
|
Ghiasi P, Hosseinkhani S, Ansari H, Aghdami N, Balalaei S, Pahlavan S, Baharvand H. Reversible permeabilization of the mitochondrial membrane promotes human cardiomyocyte differentiation from embryonic stem cells. J Cell Physiol 2018; 234:521-536. [PMID: 30071126 DOI: 10.1002/jcp.26758] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 04/23/2018] [Indexed: 01/07/2023]
Abstract
Cell death and differentiation appear to share similar cellular features. In this study, we aimed to investigate whether differentiation and mitochondrial cell death use a common pathway. We assessed the hallmarks of apoptosis during cardiomyocyte differentiation of human embryonic stem cells and found remarkable changes in P53, reactive oxygen species, apoptotic protease-activating factor 1, poly[ADP-ribose]polymerase 1, cellular adenosine triphosphate, and mitochondrial complex I activity. Furthermore, we observed reversible mitochondrial membrane permeabilization during cardiomyocyte differentiation accompanied by reversible loss of mitochondrial membrane potential, and these changes coincided with the fluctuating patterns of cytosolic cytochrome c accumulation and subsequent caspase-9 and -3/7 activation. Moreover, the use of apoptosis inhibitors (BCL2-associated X protein [BAX] inhibitor and caspase-3/7 inhibitor) during differentiation impaired cardiomyocyte development, resulting in substantial downregulation of T, MESP1, NKX2.5, and α-MHC. Additionally, although the expression of specific differentiation markers (T, MESP1, NKX2.5, MEF2C, GATA4, and SOX17) was enhanced in doxorubicin-induced human embryonic stem cells, the stemness-specific markers (OCT4 and NANOG) showed significant downregulation. With increasing doxorubicin concentration (0.03-0.6 µM; IC50 = 0.5 µM), we observed a marked increase in the expression of mesoderm and endoderm markers. In summary, we suggest that reversible mitochondrial outer membrane permeabilization promotes cardiomyocyte differentiation through an attenuated mitochondria-mediated apoptosis-like pathway.
Collapse
Affiliation(s)
- Parisa Ghiasi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Ansari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Balalaei
- Department of Chemistry, K. N. Toosi University of Technology, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
47
|
DNA damage responses and p53 in the aging process. Blood 2017; 131:488-495. [PMID: 29141944 DOI: 10.1182/blood-2017-07-746396] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022] Open
Abstract
The genome is constantly attacked by genotoxic insults. DNA damage has long been established as a cause of cancer development through its mutagenic consequences. Conversely, radiation therapy and chemotherapy induce DNA damage to drive cells into apoptosis or senescence as outcomes of the DNA damage response (DDR). More recently, DNA damage has been recognized as a causal factor for the aging process. The role of DNA damage in aging and age-related diseases is illustrated by numerous congenital progeroid syndromes that are caused by mutations in genome maintenance pathways. During the past 2 decades, understanding how DDR drives cancer development and contributes to the aging process has progressed rapidly. It turns out that the DDR factor p53 takes center stage during tumor development and also plays an important role in the aging process. Studies in metazoan models ranging from Caenorhabditis elegans to mammals have revealed cell-autonomous and systemic DDR mechanisms that orchestrate adaptive responses that augment maintenance of the aging organism amid gradually accumulating DNA damage.
Collapse
|
48
|
Alvarenga EM, Sousa NA, de Araújo S, Júnior JLP, Araújo AR, Iles B, Pacífico DM, Brito GAC, Souza EP, Sousa DP, Medeiros JVR. Carvacryl acetate, a novel semisynthetic monoterpene ester, binds to the TRPA1 receptor and is effective in attenuating irinotecan-induced intestinal mucositis in mice. J Pharm Pharmacol 2017; 69:1773-1785. [PMID: 28940490 DOI: 10.1111/jphp.12818] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/26/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVES We aimed to determine whether carvacryl acetate acts as a TRPA1 receptor agonist and its effects against irinotecan (CPT-11) induced intestinal mucositis in mice. METHODS TRPA1 structure was obtained from a protein databank, and the 3D structure of carvacryl acetate was determined. Appropriate binding conformations were discovered via automatic docking simulations. To determine the effect of carvacryl acetate in vivo, mice were treated with either DMSO 2%, CPT-11, carvacryl acetate followed by CPT-11, or HC-030031, a TRPA1 antagonist, followed by carvacryl acetate. Jejunum samples were taken and structural, inflammatory and antioxidant parameters were studied. KEY FINDINGS Eight amino acids residues in TRPA1 established stable interactions with carvacryl acetate, which led to pharmacological efficacy against CPT-11-induced intestinal mucositis via reduction of both neutropenia and bacteremia, increase in villi height and crypt depth, decrease in pro-inflammatory cytokines (interleukin-1β, keratinocyte chemoattractant and tumour necrosis factor-α) and decrease in malondialdehyde and nitric oxide metabolite levels in the jejunum. CONCLUSIONS Carvacryl acetate is a promising anti-inflammatory and antioxidant agent, a fact confirmed through observations of its interactions with TRPA1 in CPT-11-induced intestinal mucositis in mice.
Collapse
Affiliation(s)
- Elenice M Alvarenga
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Nayara A Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Simone de Araújo
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Parnaíba, PI, Brazil
| | - José L P Júnior
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Alyne R Araújo
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, PI, Brazil
| | - Bruno Iles
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Dvison M Pacífico
- Department of Morphology, Faculty of Medicine, Postgraduate Program in Morphofunctional Sciences, Federal University Ceará, Fortaleza, CE, Brazil
| | - Gerly Anne C Brito
- Department of Morphology, Faculty of Medicine, Postgraduate Program in Morphofunctional Sciences, Federal University Ceará, Fortaleza, CE, Brazil
| | - Emmanuel P Souza
- Department of Morphology, Faculty of Medicine, Postgraduate Program in Morphofunctional Sciences, Federal University Ceará, Fortaleza, CE, Brazil
| | - Damião P Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Jand Venes R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Parnaíba, PI, Brazil
| |
Collapse
|
49
|
Abramowitz J, Neuman T, Perlman R, Ben-Yehuda D. Gene and protein analysis reveals that p53 pathway is functionally inactivated in cytogenetically normal Acute Myeloid Leukemia and Acute Promyelocytic Leukemia. BMC Med Genomics 2017; 10:18. [PMID: 28340577 PMCID: PMC5423421 DOI: 10.1186/s12920-017-0249-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
Background Mechanisms that inactivate the p53 pathway in Acute Myeloid Leukemia (AML), other than rare mutations, are still not well understood. Methods We performed a bioinformatics study of the p53 pathway function at the gene expression level on our collection of 1153 p53-pathway related genes. Publically available Affymetrix data of 607 de-novo AML patients at diagnosis were analyzed according to the patients cytogenetic, FAB and molecular mutations subtypes. We further investigated the functional status of the p53 pathway in cytogenetically normal AML (CN-AML) and Acute Promyelocytic Leukemia (APL) patients using bioinformatics, Real-Time PCR and immunohistochemistry. Results We revealed significant and differential alterations of p53 pathway-related gene expression in most of the AML subtypes. We found that p53 pathway-related gene expression was not correlated with the accepted grouping of AML subtypes such as by cytogenetically-based prognosis, morphological stage or by the type of molecular mutation. Our bioinformatic analysis revealed that p53 is not functional in CN-AML and APL blasts at inducing its most important functional outcomes: cell cycle arrest, apoptosis, DNA repair and oxidative stress defense. We revealed transcriptional downregulation of important p53 acetyltransferases in both CN-AML and APL, accompanied by increased Mdmx protein expression and inadequate Chk2 protein activation. Conclusions Our bioinformatic analysis demonstrated that p53 pathway is differentially inactivated in different AML subtypes. Focused gene and protein analysis of p53 pathway in CN-AML and APL patients imply that functional inactivation of p53 protein can be attributed to its impaired acetylation. Our analysis indicates the need in further accurate evaluation of p53 pathway functioning and regulation in distinct subtypes of AML. Electronic supplementary material The online version of this article (doi:10.1186/s12920-017-0249-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Abramowitz
- Department of Hematology, Hadassah-Hebrew University Medical Center, P.O. Box 12000, Jerusalem, 91120, Israel.
| | - Tzahi Neuman
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Riki Perlman
- Department of Hematology, Hadassah-Hebrew University Medical Center, P.O. Box 12000, Jerusalem, 91120, Israel
| | - Dina Ben-Yehuda
- Department of Hematology, Hadassah-Hebrew University Medical Center, P.O. Box 12000, Jerusalem, 91120, Israel
| |
Collapse
|
50
|
Liao J, Liu PP, Hou G, Shao J, Yang J, Liu K, Lu W, Wen S, Hu Y, Huang P. Regulation of stem-like cancer cells by glutamine through β-catenin pathway mediated by redox signaling. Mol Cancer 2017; 16:51. [PMID: 28245869 PMCID: PMC5331650 DOI: 10.1186/s12943-017-0623-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/23/2017] [Indexed: 12/11/2022] Open
Abstract
Background Cancer stem cells (CSCs) are thought to play an important role in tumor recurrence and drug resistance, and present a major challenge in cancer therapy. The tumor microenvironment such as growth factors, nutrients and oxygen affect CSC generation and proliferation by providing the necessary energy sources and growth signals. The side population (SP) analysis has been used to detect the stem-like cancer cell populations based on their high expression of ABCG2 that exports Hoechst-33342 and certain cytotoxic drugs from the cells. The purpose of this research is to investigate the effect of a main nutrient molecule, glutamine, on SP cells and the possible underlying mechanism(s). Methods Biochemical assays and flow cytometric analysis were used to evaluate the effect of glutamine on stem-like side population cells in vitro. Molecular analyses including RNAi interfering, qRT-PCR, and immunoblotting were employed to investigate the molecular signaling in response to glutamine deprivation and its influence on tumor formation capacity in vivo. Results We show that glutamine supports the maintenance of the stem cell phenotype by promoting glutathione synthesis and thus maintaining redox balance for SP cells. A deprivation of glutamine in the culture medium significantly reduced the proportion of SP cells. L-asparaginase, an enzyme that catalyzes the hydrolysis of asparagine and glutamine to aspartic acid and glutamate, respectively, mimics the effect of glutamine withdrawal and also diminished the proportion of SP cells. Mechanistically, glutamine deprivation increases intracellular ROS levels, leading to down-regulation of the β-catenin pathway. Conclusion Glutamine plays a significant role in maintaining the stemness of cancer cells by a redox-mediated mechanism mediated by β-catenin. Inhibition of glutamine metabolism or deprivation of glutamine by L-asparaginase may be a new strategy to eliminate CSCs and overcome drug resistance. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0623-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianwei Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Pan-Pan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Guoxin Hou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jiajia Shao
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 Waihuan East Road, Guangzhou, 510006, China
| | - Jing Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Kaiyan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yumin Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China. .,Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|