51
|
Mathialagan RD, Abd Hamid Z, Ng QM, Rajab NF, Shuib S, Binti Abdul Razak SR. Bone Marrow Oxidative Stress and Acquired Lineage-Specific Genotoxicity in Hematopoietic Stem/Progenitor Cells Exposed to 1,4-Benzoquinone. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17165865. [PMID: 32823552 PMCID: PMC7459782 DOI: 10.3390/ijerph17165865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) are susceptible to benzene-induced genotoxicity. However, little is known about the mechanism of DNA damage response affecting lineage-committed progenitors for myeloid, erythroid, and lymphoid. Here, we investigated the genotoxicity of a benzene metabolite, 1,4-benzoquinone (1,4-BQ), in HSPCs using oxidative stress and lineage-directed approaches. Mouse bone marrow cells (BMCs) were exposed to 1,4-BQ (1.25–12 μM) for 24 h, followed by oxidative stress and genotoxicity assessments. Then, the genotoxicity of 1,4-BQ in lineage-committed progenitors was evaluated using colony forming cell assay following 7–14 days of culture. 1,4-BQ exposure causes significant decreases (p < 0.05) in glutathione level and superoxide dismutase activity, along with significant increases (p < 0.05) in levels of malondialdehyde and protein carbonyls. 1,4-BQ exposure induces DNA damage in BMCs by significantly (p < 0.05) increased percentages of DNA in tail at 7 and 12 μM and tail moment at 12 μM. We found crucial differences in genotoxic susceptibility based on percentages of DNA in tail between lineage-committed progenitors. Myeloid and pre-B lymphoid progenitors appeared to acquire significant DNA damage as compared with the control starting from a low concentration of 1,4-BQ exposure (2.5 µM). In contrast, the erythroid progenitor showed significant damage as compared with the control starting at 5 µM 1,4-BQ. Meanwhile, a significant (p < 0.05) increase in tail moment was only notable at 7 µM and 12 µM 1,4-BQ exposure for all progenitors. Benzene could mediate hematological disorders by promoting bone marrow oxidative stress and lineage-specific genotoxicity targeting HSPCs.
Collapse
Affiliation(s)
- Ramya Dewi Mathialagan
- Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 53000, Malaysia; (R.D.M.); (Q.M.N.)
| | - Zariyantey Abd Hamid
- Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 53000, Malaysia; (R.D.M.); (Q.M.N.)
- Correspondence:
| | - Qing Min Ng
- Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 53000, Malaysia; (R.D.M.); (Q.M.N.)
| | - Nor Fadilah Rajab
- Biomedical Science Programme and Center for Healthy Ageing & Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 53000, Malaysia;
| | - Salwati Shuib
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Siti Razila Binti Abdul Razak
- Oncological and Radiological Sciences Cluster, Advanced Medical & Dental Institute, Universiti Sains Malaysia, Kepala Batas Bertam, Pulau Pinang 13200, Malaysia;
| |
Collapse
|
52
|
Himburg HA, Roos M, Fang T, Zhang Y, Termini CM, Schlussel L, Kim M, Pang A, Kan J, Zhao L, Suh H, Sasine JP, Sapparapu G, Bowers PM, Schiller G, Chute JP. Chronic myeloid leukemia stem cells require cell-autonomous pleiotrophin signaling. J Clin Invest 2020; 130:315-328. [PMID: 31613796 DOI: 10.1172/jci129061] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/25/2019] [Indexed: 01/11/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) induce molecular remission in the majority of patients with chronic myelogenous leukemia (CML), but the persistence of CML stem cells hinders cure and necessitates indefinite TKI therapy. We report that CML stem cells upregulate the expression of pleiotrophin (PTN) and require cell-autonomous PTN signaling for CML pathogenesis in BCR/ABL+ mice. Constitutive PTN deletion substantially reduced the numbers of CML stem cells capable of initiating CML in vivo. Hematopoietic cell-specific deletion of PTN suppressed CML development in BCR/ABL+ mice, suggesting that cell-autonomous PTN signaling was necessary for CML disease evolution. Mechanistically, PTN promoted CML stem cell survival and TKI resistance via induction of Jun and the unfolded protein response. Human CML cells were also dependent on cell-autonomous PTN signaling, and anti-PTN antibody suppressed human CML colony formation and CML repopulation in vivo. Our results suggest that targeted inhibition of PTN has therapeutic potential to eradicate CML stem cells.
Collapse
Affiliation(s)
- Heather A Himburg
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Martina Roos
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Tiancheng Fang
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California, USA
| | - Yurun Zhang
- Molecular Biology Institute, UCLA, Los Angeles, California, USA
| | - Christina M Termini
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Lauren Schlussel
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Mindy Kim
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Amara Pang
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Jenny Kan
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Liman Zhao
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Hyung Suh
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Joshua P Sasine
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Gopal Sapparapu
- UCLA Clinical and Translational Science Institute, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Peter M Bowers
- UCLA Clinical and Translational Science Institute, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Gary Schiller
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - John P Chute
- Division of Hematology/Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center for Stem Cell Research and Regenerative Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
53
|
Kirtonia A, Pandya G, Sethi G, Pandey AK, Das BC, Garg M. A comprehensive review of genetic alterations and molecular targeted therapies for the implementation of personalized medicine in acute myeloid leukemia. J Mol Med (Berl) 2020; 98:1069-1091. [PMID: 32620999 DOI: 10.1007/s00109-020-01944-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/18/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is an extremely heterogeneous disease defined by the clonal growth of myeloblasts/promyelocytes not only in the bone marrow but also in peripheral blood and/or tissues. Gene mutations and chromosomal abnormalities are usually associated with aberrant proliferation and/or block in the normal differentiation of hematopoietic cells. So far, the combination of cytogenetic profiling and molecular and gene mutation analyses remains an essential tool for the classification, diagnosis, prognosis, and treatment for AML. This review gives an overview on how the development of novel innovative technologies has allowed us not only to detect the genetic alterations as early as possible but also to understand the molecular pathogenesis of AML to develop novel targeted therapies. We also discuss the remarkable advances made during the last decade to understand the AML genome both at primary and relapse diseases and how genetic alterations might influence the distinct biological groups as well as the clonal evolution of disease during the diagnosis and relapse. Also, the review focuses on how the persistence of epigenetic gene mutations during morphological remission is associated with relapse. It is suggested that along with the prognostic and therapeutic mutations, the novel molecular targeted therapies either approved by FDA or those under clinical trials including CART-cell therapy would be of immense importance in the effective management of AML.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology (AIB), Amity University, Gurgaon, Haryana, 122413, India
| | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
54
|
Tan Y, Wu Q, Zhou F. Targeting acute myeloid leukemia stem cells: Current therapies in development and potential strategies with new dimensions. Crit Rev Oncol Hematol 2020; 152:102993. [PMID: 32502928 DOI: 10.1016/j.critrevonc.2020.102993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
High relapse rate of acute myeloid leukemia (AML) is still a crucial problem despite considerable advances in anti-cancer therapies. One crucial cause of relapse is the existence of leukemia stem cells (LSCs) with self-renewal ability, which contribute to repeated treatment resistance and recurrence. Treatments targeting LSCs, especially in combination with existing chemotherapy regimens or hematopoietic stem cell transplantation might help achieve a higher complete remission rate and improve overall survival. Many novel agents of different therapeutic strategies that aim to modulate LSCs self-renewal, proliferation, apoptosis, and differentiation are under investigation. In this review, we summarize the latest advances of different therapies in development based on the biological characteristics of LSCs, with particular attention on natural products, synthetic compounds, antibody therapies, and adoptive cell therapies that promote the LSC eradication. We also explore the causes of AML recurrence and proposed potential strategies with new dimensions for targeting LSCs in the future.
Collapse
Affiliation(s)
- Yuxin Tan
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.
| |
Collapse
|
55
|
Network Pharmacology-Based Investigation of the System-Level Molecular Mechanisms of the Hematopoietic Activity of Samul-Tang, a Traditional Korean Herbal Formula. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9048089. [PMID: 32104198 PMCID: PMC7040423 DOI: 10.1155/2020/9048089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
Hematopoiesis is a dynamic process of the continuous production of diverse blood cell types to meet the body's physiological demands and involves complex regulation of multiple cellular mechanisms in hematopoietic stem cells, including proliferation, self-renewal, differentiation, and apoptosis. Disruption of the hematopoietic system is known to cause various hematological disorders such as myelosuppression. There is growing evidence on the beneficial effects of herbal medicines on hematopoiesis; however, their mechanism of action remains unclear. In this study, we conducted a network pharmacological-based investigation of the system-level mechanisms underlying the hematopoietic activity of Samul-tang, which is an herbal formula consisting of four herbal medicines, including Angelicae Gigantis Radix, Rehmanniae Radix Preparata, Paeoniae Radix Alba, and Cnidii Rhizoma. In silico analysis of the absorption-distribution-metabolism-excretion model identified 16 active phytochemical compounds contained in Samul-tang that may target 158 genes/proteins associated with myelosuppression to exert pharmacological effects. Functional enrichment analysis suggested that the targets of Samul-tang were significantly enriched in multiple pathways closely related to the hematopoiesis and myelosuppression development, including the PI3K-Akt, MAPK, IL-17, TNF, FoxO, HIF-1, NF-kappa B, and p53 signaling pathways. Our study provides novel evidence regarding the system-level mechanisms underlying the hematopoiesis-promoting effect of herbal medicines for hematological disorder treatment.
Collapse
|
56
|
Staehle HF, Heinemann J, Gruender A, Omlor AM, Pahl HL, Jutzi JS. Jmjd1c is dispensable for healthy adult hematopoiesis and Jak2V617F-driven myeloproliferative disease initiation in mice. PLoS One 2020; 15:e0228362. [PMID: 32017785 PMCID: PMC6999878 DOI: 10.1371/journal.pone.0228362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/13/2020] [Indexed: 11/30/2022] Open
Abstract
The histone demethylase JMJD1C is overexpressed in patients with myeloproliferative neoplasms (MPNs) and has been implicated in leukemic stem cell function of MLL-AF9 and HOXA9-driven leukemia. In the emerging field of histone demethylase inhibitors, JMJD1C therefore became a potential target. Depletion of Jmjd1c expression significantly reduced cytokine-independent growth in an MPN cell line, indicating a role for JMJD1C in MPN disease maintenance. Here, we investigated a potential role for the demethylase in MPN disease initiation. We introduced a Cre-inducible JAK2V617F mutation into Jmjd1c knockout mice. We show that Jmjd1c is dispensable, both for healthy hematopoiesis as well as for JAK2V617F-driven MPN disease initiation. Jmjd1c knockout mice did not show any significant changes in peripheral blood composition. Likewise, introduction of JAK2V617F into Jmjd1c-/- mice led to a similar MPN phenotype as JAK2V617F in a Jmjd1c wt background. This indicates that there is a difference between the role of JMJD1C in leukemic stem cells and in MPN. In the latter, JMJC domain-containing family members may serve redundant roles, compensating for the loss of individual proteins.
Collapse
Affiliation(s)
- Hans F. Staehle
- Division of Molecular Hematology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| | - Johannes Heinemann
- Division of Molecular Hematology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| | - Albert Gruender
- Division of Molecular Hematology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| | - Anne M. Omlor
- Division of Molecular Hematology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| | - Heike Luise Pahl
- Division of Molecular Hematology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| | - Jonas Samuel Jutzi
- Division of Molecular Hematology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| |
Collapse
|
57
|
Abugomaa A, Elbadawy M, Yamawaki H, Usui T, Sasaki K. Emerging Roles of Cancer Stem Cells in Bladder Cancer Progression, Tumorigenesis, and Resistance to Chemotherapy: A Potential Therapeutic Target for Bladder Cancer. Cells 2020; 9:235. [PMID: 31963556 PMCID: PMC7016964 DOI: 10.3390/cells9010235] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
Bladder cancer (BC) is a complex and highly heterogeneous stem cell disease associated with high morbidity and mortality rates if it is not treated properly. Early diagnosis with personalized therapy and regular follow-up are the keys to a successful outcome. Cancer stem cells (CSCs) are the leading power behind tumor growth, with the ability of self-renewal, metastasis, and resistance to conventional chemotherapy. The fast-developing CSC field with robust genome-wide screening methods has found a platform for establishing more reliable therapies to target tumor-initiating cell populations. However, the high heterogeneity of the CSCs in BC disease remains a large issue. Therefore, in the present review, we discuss the various types of bladder CSC heterogeneity, important regulatory pathways, roles in tumor progression and tumorigenesis, and the experimental culture models. Finally, we describe the current stem cell-based therapies for BC disease.
Collapse
Affiliation(s)
- Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (A.A.); (M.E.); (K.S.)
- Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Dakahliya, Egypt
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (A.A.); (M.E.); (K.S.)
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan;
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (A.A.); (M.E.); (K.S.)
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (A.A.); (M.E.); (K.S.)
| |
Collapse
|
58
|
Mitchell K, Steidl U. Targeting Immunophenotypic Markers on Leukemic Stem Cells: How Lessons from Current Approaches and Advances in the Leukemia Stem Cell (LSC) Model Can Inform Better Strategies for Treating Acute Myeloid Leukemia (AML). Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036251. [PMID: 31451539 DOI: 10.1101/cshperspect.a036251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapies targeting cell-surface antigens in acute myeloid leukemia (AML) have been tested over the past 20 years with limited improvement in overall survival. Recent advances in the understanding of AML pathogenesis support therapeutic targeting of leukemia stem cells as the most promising avenue toward a cure. In this review, we provide an overview of the evolving leukemia stem cell (LSC) model, including evidence of the cell of origin, cellular and molecular disease architecture, and source of relapse in AML. In addition, we explore limitations of current targeted strategies utilized in AML and describe the various immunophenotypic antigens that have been proposed as LSC-directed therapeutic targets. We draw lessons from current approaches as well as from the (pre)-LSC model to suggest criteria that immunophenotypic targets should meet for more specific and effective elimination of disease-initiating clones, highlighting in detail a few targets that we suggest fit these criteria most completely.
Collapse
Affiliation(s)
- Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, New York 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
59
|
Dewi R, Hamid ZA, Rajab NF, Shuib S, Razak SA. Genetic, epigenetic, and lineage-directed mechanisms in benzene-induced malignancies and hematotoxicity targeting hematopoietic stem cells niche. Hum Exp Toxicol 2019; 39:577-595. [PMID: 31884827 DOI: 10.1177/0960327119895570] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Benzene is a known hematotoxic and leukemogenic agent with hematopoietic stem cells (HSCs) niche being the potential target. Occupational and environmental exposure to benzene has been linked to the incidences of hematological disorders and malignancies. Previous studies have shown that benzene may act via multiple modes of action targeting HSCs niche, which include induction of chromosomal and micro RNA aberrations, leading to genetic and epigenetic modification of stem cells and probable carcinogenesis. However, understanding the mechanism linking benzene to the HSCs niche dysregulation is challenging due to complexity of its microenvironment. The niche is known to comprise of cell populations accounted for HSCs and their committed progenitors of lymphoid, erythroid, and myeloid lineages. Thus, it is fundamental to address novel approaches via lineage-directed strategy to elucidate precise mechanism involved in benzene-induced toxicity targeting HSCs and progenitors of different lineages. Here, we review the key genetic and epigenetic factors that mediate hematotoxicological effects by benzene and its metabolites in targeting HSCs niche. Overall, the use of combined genetic, epigenetic, and lineage-directed strategies targeting the HSCs niche is fundamental to uncover the key mechanisms in benzene-induced hematological disorders and malignancies.
Collapse
Affiliation(s)
- R Dewi
- Biomedical Science Programme and Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Z Abdul Hamid
- Biomedical Science Programme and Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - N F Rajab
- Biomedical Science Programme and Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - S Shuib
- Department of Pathology, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Kuala Lumpur, Malaysia
| | - Sr Abdul Razak
- Oncological and Radiological Sciences Cluster, Advanced Medical & Dental Institute, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| |
Collapse
|
60
|
Vorbach S, Gründer A, Zhou F, Koellerer C, Jutzi JS, Simoni M, Riccetti L, Valk PJ, Sanders MA, Müller-Tidow C, Nofer JR, Pahl HL, Potì F. Enhanced expression of the sphingosine-1-phosphate-receptor-3 causes acute myelogenous leukemia in mice. Leukemia 2019; 34:721-734. [PMID: 31636343 DOI: 10.1038/s41375-019-0577-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/20/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemia (AML) carries a 10-100 fold lower mutational burden than other neoplastic entities. Mechanistic explanations for why a low number of mutations suffice to induce leukemogenesis are therefore required. Here we demonstrate that transgenic overexpression of the wild type sphingosine-1-phosphate receptor 3 (S1P3) in murine hematopoietic stem cells is sufficient to induce a transplantable myeloid leukemia. In contrast, S1P3 expression in more mature compartments does not cause malignant transformation. Treatment with the sphingosine phosphate receptor modulator Fingolimod, which prevents receptor signaling, normalized peripheral blood cell counts and reduced spleen sizes in S1P3 expressing mice. Gene expression analyses in AML patients revealed elevated S1P3 expression specifically in two molecular subclasses. Our data suggest a previously unrecognized contribution of wild type S1P3 signaling to leukemogenesis that warrants the exploration of S1P3 antagonists in preclinical AML models.
Collapse
Affiliation(s)
- Samuel Vorbach
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Albert Gründer
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Fengbiao Zhou
- Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Christoph Koellerer
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Jonas S Jutzi
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Manuela Simoni
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, Modena, Italy
| | - Laura Riccetti
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, Modena, Italy
| | - Peter J Valk
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mathijs A Sanders
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Carsten Müller-Tidow
- Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Jerzy-Roch Nofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Heike L Pahl
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| | - Francesco Potì
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, Modena, Italy.,Department of Medicine and Surgery-Unit of Neurosciences, University of Parma, Via Volturno 39/F, 43125, Parma, Italy
| |
Collapse
|
61
|
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
|
62
|
Jin J, Luo S, Ye X, Huang J. BCOR mutation and TLS-ERG expression in acute myeloid leukemia with monoclonal immunoglobulinemia. Onco Targets Ther 2019; 12:7123-7127. [PMID: 31564896 PMCID: PMC6730603 DOI: 10.2147/ott.s218805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 08/08/2019] [Indexed: 11/23/2022] Open
Abstract
Acute myeloid leukemia (AML) originates from the abnormal clonal proliferation of myeloblasts. Immunoglobulin is secreted by B cells. AML with monoclonal antibody often indicates a poor prognosis. Here we report a case of BCOR mutation and TLS-ERG expression in AML with monoclonal immunoglobulinemia. After chemotherapy, the patient achieved bone marrow complete remission. BCOR mutation and TLS-ERG fusion gene in patient's bone marrow were not detected, at the same time, peripheral blood monoclonal immunoglobulin also disappeared. BCOR mutation or TLS-ERG fusion gene expression is associated with poor prognosis, AML with monoclonal immunoglobulin may have the same prognostic significance.
Collapse
Affiliation(s)
- Jingxia Jin
- Department of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, People's Republic of China
| | - Shuna Luo
- Department of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, People's Republic of China
| | - XingNong Ye
- Department of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, People's Republic of China
| | - Jian Huang
- Department of Hematology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, People's Republic of China
| |
Collapse
|
63
|
Elbadawy M, Usui T, Mori T, Tsunedomi R, Hazama S, Nabeta R, Uchide T, Fukushima R, Yoshida T, Shibutani M, Tanaka T, Masuda S, Okada R, Ichikawa R, Omatsu T, Mizutani T, Katayama Y, Noguchi S, Iwai S, Nakagawa T, Shinohara Y, Kaneda M, Yamawaki H, Sasaki K. Establishment of a novel experimental model for muscle-invasive bladder cancer using a dog bladder cancer organoid culture. Cancer Sci 2019; 110:2806-2821. [PMID: 31254429 PMCID: PMC6726682 DOI: 10.1111/cas.14118] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/20/2019] [Accepted: 06/27/2019] [Indexed: 12/19/2022] Open
Abstract
In human and dogs, bladder cancer (BC) is the most common neoplasm affecting the urinary tract. Dog BC resembles human muscle-invasive BC in histopathological characteristics and gene expression profiles, and could be an important research model for this disease. Cancer patient-derived organoid culture can recapitulate organ structures and maintains the gene expression profiles of original tumor tissues. In a previous study, we generated dog prostate cancer organoids using urine samples, however dog BC organoids had never been produced. Therefore we aimed to generate dog BC organoids using urine samples and check their histopathological characteristics, drug sensitivity, and gene expression profiles. Organoids from individual BC dogs were successfully generated, expressed urothelial cell markers (CK7, CK20, and UPK3A) and exhibited tumorigenesis in vivo. In a cell viability assay, the response to combined treatment with a range of anticancer drugs (cisplatin, vinblastine, gemcitabine or piroxicam) was markedly different in each BC organoid. In RNA-sequencing analysis, expression levels of basal cell markers (CK5 and DSG3) and several novel genes (MMP28, CTSE, CNN3, TFPI2, COL17A1, and AGPAT4) were upregulated in BC organoids compared with normal bladder tissues or two-dimensional (2D) BC cell lines. These established dog BC organoids might be a useful tool, not only to determine suitable chemotherapy for BC diseased dogs but also to identify novel biomarkers in human muscle-invasive BC. In the present study, for the 1st time, dog BC organoids were generated and several specifically upregulated organoid genes were identified. Our data suggest that dog BC organoids might become a new tool to provide fresh insights into both dog BC therapy and diagnostic biomarkers.
Collapse
Affiliation(s)
- Mohamed Elbadawy
- Laboratory of Veterinary PharmacologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
- Department of PharmacologyFaculty of Veterinary MedicineBenha UniversityToukhEgypt
| | - Tatsuya Usui
- Laboratory of Veterinary PharmacologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Takashi Mori
- Laboratory of Veterinary Clinical OncologyFaculty of Applied Biological SciencesGifu UniversityGifuJapan
- Center for Highly Advanced Integration of Nano and Life SciencesGifu University (G‐CHAIN)GifuJapan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast, and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast, and Endocrine SurgeryYamaguchi University Graduate School of MedicineUbeJapan
- Department of Translational Research and Developmental Therapeutics against CancerSchool of MedicineYamaguchi UniversityUbeJapan
| | - Rina Nabeta
- Department of Veterinary SurgeryFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Tsuyoshi Uchide
- Department of Veterinary SurgeryFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Ryuji Fukushima
- Animal Medical CenterFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Toshinori Yoshida
- Laboratory of Veterinary PathologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Makoto Shibutani
- Laboratory of Veterinary PathologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Takaharu Tanaka
- Laboratory of Veterinary PathologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Sosuke Masuda
- Laboratory of Veterinary PathologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Rena Okada
- Laboratory of Veterinary PathologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Ryo Ichikawa
- Laboratory of Veterinary PathologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Tsutomu Omatsu
- Research and Education Center for Prevention of Global Infectious Disease of AnimalsTokyo University of Agriculture and TechnologyFuchuJapan
| | - Tetsuya Mizutani
- Research and Education Center for Prevention of Global Infectious Disease of AnimalsTokyo University of Agriculture and TechnologyFuchuJapan
| | - Yukie Katayama
- Research and Education Center for Prevention of Global Infectious Disease of AnimalsTokyo University of Agriculture and TechnologyFuchuJapan
| | - Shunsuke Noguchi
- Laboratory of Veterinary RadiologyGraduate School of Life and Environmental SciencesOsaka Prefecture UniversitySanoJapan
| | - Satomi Iwai
- Laboratory of Small Animal Surgery 2School of Veterinary MedicineKitasato UniversityTowadaJapan
| | - Takayuki Nakagawa
- Laboratory of Veterinary SurgeryGraduate School of Agricultural and Life SciencesUniversity of TokyoTokyoJapan
| | - Yuta Shinohara
- Laboratory of Veterinary PharmacologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
- Pet Health & Food DivisionIskara Industry CO., LTDTokyoJapan
| | - Masahiro Kaneda
- Laboratory of Veterinary AnatomyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary PharmacologySchool of Veterinary MedicineKitasato UniversityTowadaJapan
| | - Kazuaki Sasaki
- Laboratory of Veterinary PharmacologyDepartment of Veterinary MedicineFaculty of AgricultureTokyo University of Agriculture and TechnologyFuchuJapan
| |
Collapse
|
64
|
Loizou E, Banito A, Livshits G, Ho YJ, Koche RP, Sánchez-Rivera FJ, Mayle A, Chen CC, Kinalis S, Bagger FO, Kastenhuber ER, Durham BH, Lowe SW. A Gain-of-Function p53-Mutant Oncogene Promotes Cell Fate Plasticity and Myeloid Leukemia through the Pluripotency Factor FOXH1. Cancer Discov 2019; 9:962-979. [PMID: 31068365 DOI: 10.1158/2159-8290.cd-18-1391] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/20/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Mutations in the TP53 tumor suppressor gene are common in many cancer types, including the acute myeloid leukemia (AML) subtype known as complex karyotype AML (CK-AML). Here, we identify a gain-of-function (GOF) Trp53 mutation that accelerates CK-AML initiation beyond p53 loss and, surprisingly, is required for disease maintenance. The Trp53R172H mutation (TP53R175H in humans) exhibits a neomorphic function by promoting aberrant self-renewal in leukemic cells, a phenotype that is present in hematopoietic stem and progenitor cells (HSPC) even prior to their transformation. We identify FOXH1 as a critical mediator of mutant p53 function that binds to and regulates stem cell-associated genes and transcriptional programs. Our results identify a context where mutant p53 acts as a bona fide oncogene that contributes to the pathogenesis of CK-AML and suggests a common biological theme for TP53 GOF in cancer. SIGNIFICANCE: Our study demonstrates how a GOF p53 mutant can hijack an embryonic transcription factor to promote aberrant self-renewal. In this context, mutant Trp53 functions as an oncogene to both initiate and sustain myeloid leukemia and suggests a potential convergent activity of mutant Trp53 across cancer types.This article is highlighted in the In This Issue feature, p. 813.
Collapse
Affiliation(s)
- Evangelia Loizou
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Graduate School of Medical Sciences, New York, New York
| | - Ana Banito
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geulah Livshits
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Francisco J Sánchez-Rivera
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Allison Mayle
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chi-Chao Chen
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Savvas Kinalis
- Center for Genomic Medicine, Rigshopitalet, University of Copenhagen, Copenhagen, Denmark
| | - Frederik O Bagger
- Center for Genomic Medicine, Rigshopitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedicine and Swiss Institute of Bioinformatics, UKBB Universitats-Kinderspital, Basel, Switzerland
| | - Edward R Kastenhuber
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York.,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benjamin H Durham
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York. .,Howard Hughes Medical Institute, New York, New York
| |
Collapse
|
65
|
Metabolic Imaging Reveals a Unique Preference of Symmetric Cell Division and Homing of Leukemia-Initiating Cells in an Endosteal Niche. Cell Metab 2019; 29:950-965.e6. [PMID: 30581117 DOI: 10.1016/j.cmet.2018.11.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 07/23/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023]
Abstract
The metabolic properties of leukemia-initiating cells (LICs) in distinct bone marrow niches and their relationships to cell-fate determinations remain largely unknown. Using a metabolic imaging system with a highly responsive genetically encoded metabolic sensor, SoNar, we reveal that SoNar-high cells are more glycolytic, enriched for higher LIC frequency, and develop leukemia much faster than SoNar-low counterparts in an MLL-AF9-induced murine acute myeloid leukemia model. SoNar-high cells mainly home to and locate in the hypoxic endosteal niche and maintain their activities through efficient symmetric division. SoNar can indicate the dynamics of metabolic changes of LICs in the endosteal niche. SoNar-high human leukemia cells or primary samples have enhanced clonogenic capacities in vitro or leukemogenesis in vivo. PDK2 fine-tunes glycolysis, homing, and symmetric division of LICs. These findings provide a unique angle for the study of metabolisms in stem cells, and may lead to development of novel strategies for cancer treatment.
Collapse
|
66
|
Piya S, Mu H, Bhattacharya S, Lorenzi PL, Davis RE, McQueen T, Ruvolo V, Baran N, Wang Z, Qian Y, Crews CM, Konopleva M, Ishizawa J, You MJ, Kantarjian H, Andreeff M, Borthakur G. BETP degradation simultaneously targets acute myelogenous leukemia stem cells and the microenvironment. J Clin Invest 2019; 129:1878-1894. [PMID: 30829648 DOI: 10.1172/jci120654] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Anti-leukemic effect of BET/BRD4 (BETP) protein inhibition has been largely attributed to transcriptional downregulation of cellular anabolic/anti-apoptotic processes but its effect on bone marrow microenvironment, a sanctuary favoring persistence of leukemia stem/progenitor cells, is unexplored. Sustained degradation of BETP with small-molecule BET proteolysis-targeting chimera (PROTAC), ARV-825, resulted in marked downregulation of surface CXCR4 and CD44, key proteins in leukemia-microenvironment interaction, in AML cells. Abrogation of surface CXCR4 expression impaired SDF-1α directed migration and was mediated through transcriptional down-regulation of PIM1 kinase that in turn phosphorylates CXCR4 and facilitates its surface localization. Down-regulation of CD44/CD44v8-10 impaired cystine uptake, lowered intracellular reduced glutathione and increased oxidative stress. More importantly, BETP degradation markedly decreased CD34+CD38-CD90-CD45RA+ leukemic stem cell population and alone or in combination with Cytarabine, prolonged survival in mouse model of human leukemia including AML-PDX. Gene expression profiling and single cell proteomics confirmed down regulation of the gene signatures associated with 'stemness' in AML and Wnt/β-catenin, Myc pathways. Hence, BETP degradation by ARV-825 simultaneously targets cell intrinsic signaling, stromal interactions and metabolism in AML.
Collapse
Affiliation(s)
- Sujan Piya
- Section of Molecular Hematology and Therapy.,Department of Leukemia
| | - Hong Mu
- Section of Molecular Hematology and Therapy.,Department of Leukemia
| | | | | | - R Eric Davis
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Teresa McQueen
- Section of Molecular Hematology and Therapy.,Department of Leukemia
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy.,Department of Leukemia
| | - Natalia Baran
- Section of Molecular Hematology and Therapy.,Department of Leukemia
| | - Zhiqiang Wang
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yimin Qian
- Arvinas Inc., New Haven, Connecticut, USA
| | - Craig M Crews
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | | | | | - M James You
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Michael Andreeff
- Section of Molecular Hematology and Therapy.,Department of Leukemia
| | | |
Collapse
|
67
|
Poljsak B, Kovac V, Dahmane R, Levec T, Starc A. Cancer Etiology: A Metabolic Disease Originating from Life's Major Evolutionary Transition? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7831952. [PMID: 31687086 PMCID: PMC6800902 DOI: 10.1155/2019/7831952] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/21/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022]
Abstract
A clear understanding of the origins of cancer is the basis of successful strategies for effective cancer prevention and management. The origin of cancer at the molecular and cellular levels is not well understood. Is the primary cause of the origin of cancer the genomic instability or impaired energy metabolism? An attempt was made to present cancer etiology originating from life's major evolutionary transition. The first evolutionary transition went from simple to complex cells when eukaryotic cells with glycolytic energy production merged with the oxidative mitochondrion (The Endosymbiosis Theory first proposed by Lynn Margulis in the 1960s). The second transition went from single-celled to multicellular organisms once the cells obtained mitochondria, which enabled them to obtain a higher amount of energy. Evidence will be presented that these two transitions, as well as the decline of NAD+ and ATP levels, are the root of cancer diseases. Restoring redox homeostasis and reactivation of mitochondrial oxidative metabolism are important factors in cancer prevention.
Collapse
Affiliation(s)
- B. Poljsak
- 1Faculty of Health Sciences, University of Ljubljana, Laboratory of Oxidative Stress Research, Ljubljana, Slovenia
| | - V. Kovac
- 1Faculty of Health Sciences, University of Ljubljana, Laboratory of Oxidative Stress Research, Ljubljana, Slovenia
| | - R. Dahmane
- 2Faculty of Health Sciences, University of Ljubljana, Chair of Biomedicine in Health Care, Ljubljana, Slovenia
| | - T. Levec
- 3Faculty of Health Sciences, University of Ljubljana, Chair of Public Health, Ljubljana, Slovenia
| | - A. Starc
- 3Faculty of Health Sciences, University of Ljubljana, Chair of Public Health, Ljubljana, Slovenia
| |
Collapse
|
68
|
Vu LP, Cheng Y, Kharas MG. The Biology of m 6A RNA Methylation in Normal and Malignant Hematopoiesis. Cancer Discov 2018; 9:25-33. [PMID: 30578356 DOI: 10.1158/2159-8290.cd-18-0959] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 11/16/2022]
Abstract
Hematopoietic development and differentiation are highly regulated processes, and recent studies focusing on m6A mRNA methylation have uncovered how this mark controls cell fate in both normal and malignant hematopoietic states. In this review, we focus on how writers, readers, and erasers of RNA methylation can mediate distinct phenotypes on mRNAs and on cells. Targeting the RNA methylation program has emerged as a potential novel therapeutic strategy, and we explore the role for these regulators in both normal and dysregulated cell contexts. SIGNIFICANCE: RNA methylation is required for cancer cell survival in solid tumors and in acute myeloid leukemia, and targeting this pathway has been proposed as a new therapeutic strategy in cancer. However, understanding the role for RNA methylation in both normal and malignant states is essential for understanding the potential consequences for therapeutic intervention.
Collapse
Affiliation(s)
- Ly P Vu
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuanming Cheng
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael G Kharas
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
69
|
Abstract
Bones provide both skeletal scaffolding and space for hematopoiesis in its marrow. Previous work has shown that these functions were tightly regulated by the nervous system. The central and peripheral nervous systems tightly regulate compact bone remodeling, its metabolism, and hematopoietic homeostasis in the bone marrow (BM). Accumulating evidence indicates that the nervous system, which fine-tunes inflammatory responses and alterations in neural functions, may regulate autoimmune diseases. Neural signals also influence the progression of hematological malignancies such as acute and chronic myeloid leukemias. Here, we review the interplay of the nervous system with bone, BM, and immunity, and discuss future challenges to target hematological diseases through modulation of activity of the nervous system.
Collapse
Affiliation(s)
- Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
70
|
Bellmunt J. Stem-Like Signature Predicting Disease Progression in Early Stage Bladder Cancer. The Role of E2F3 and SOX4. Biomedicines 2018; 6:biomedicines6030085. [PMID: 30072631 PMCID: PMC6164884 DOI: 10.3390/biomedicines6030085] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022] Open
Abstract
The rapid development of the cancer stem cells (CSC) field, together with powerful genome-wide screening techniques, have provided the basis for the development of future alternative and reliable therapies aimed at targeting tumor-initiating cell populations. Urothelial bladder cancer stem cells (BCSCs) that were identified for the first time in 2009 are heterogenous and originate from multiple cell types; including urothelial stem cells and differentiated cell types—basal, intermediate stratum and umbrella cells Some studies hypothesize that BCSCs do not necessarily arise from normal stem cells but might derive from differentiated progenies following mutational insults and acquisition of tumorigenic properties. Conversely, there is data that normal bladder tissues can generate CSCs through mutations. Prognostic risk stratification by identification of predictive markers is of major importance in the management of urothelial cell carcinoma (UCC) patients. Several stem cell markers have been linked to recurrence or progression. The CD44v8-10 to standard CD44-ratio (total ratio of all CD44 alternative splicing isoforms) in urothelial cancer has been shown to be closely associated with tumor progression and aggressiveness. ALDH1, has also been reported to be associated with BCSCs and a worse prognosis in a large number of studies. UCC include low-grade and high-grade non-muscle invasive bladder cancer (NMIBC) and high-grade muscle invasive bladder cancer (MIBC). Important genetic defects characterize the distinct pathways in each one of the stages and probably grades. As an example, amplification of chromosome 6p22 is one of the most frequent changes seen in MIBC and might act as an early event in tumor progression. Interestingly, among NMIBC there is a much higher rate of amplification in high-grade NMIBC compared to low grade NMIBC. CDKAL1, E2F3 and SOX4 are highly expressed in patients with the chromosomal 6p22 amplification aside from other six well known genes (ID4, MBOAT1, LINC00340, PRL, and HDGFL1). Based on that, SOX4, E2F3 or 6q22.3 amplifications might represent potential targets in this tumor type. Focusing more in SOX4, it seems to exert its critical regulatory functions upstream of the Snail, Zeb, and Twist family of transcriptional inducers of EMT (epithelial–mesenchymal transition), but without directly affecting their expression as seen in several cell lines of the Cancer Cell Line Encyclopedia (CCLE) project. SOX4 gene expression correlates with advanced cancer stages and poor survival rate in bladder cancer, supporting a potential role as a regulator of the bladder CSC properties. SOX4 might serve as a biomarker of the aggressive phenotype, also underlying progression from NMIBC to MIBC. The amplicon in chromosome 6 contains SOX4 and E2F3 and is frequently found amplified in bladder cancer. These genes/amplicons might be a potential target for therapy. As an existing hypothesis is that chromatin deregulation through enhancers or super-enhancers might be the underlying mechanism responsible of this deregulation, a potential way to target these transcription factors could be through epigenetic modifiers.
Collapse
Affiliation(s)
- Joaquim Bellmunt
- Department of Medical Oncology, Hospital del Mar, IMIM (PSMAR-Hospital del Mar Research Institute), 08003 Barcelona, Spain.
- Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
71
|
Han SW, Kim YY, Kang WJ, Kim HC, Ku SY, Kang BC, Yun JW. The Use of Normal Stem Cells and Cancer Stem Cells for Potential Anti-Cancer Therapeutic Strategy. Tissue Eng Regen Med 2018; 15:365-380. [PMID: 30603561 PMCID: PMC6171655 DOI: 10.1007/s13770-018-0128-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/08/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Despite recent advance in conventional cancer therapies including surgery, radiotherapy, chemotherapy, and immunotherapy to reduce tumor size, unfortunately cancer mortality and metastatic cancer incidence remain high. Along with a deeper understanding of stem cell biology, cancer stem cell (CSC) is important in targeted cancer therapy. Herein, we review representative patents using not only normal stem cells as therapeutics themselves or delivery vehicles, but also CSCs as targets for anti-cancer strategy. METHODS Relevant patent literatures published between 2005 and 2017 are discussed to present developmental status and experimental results on using normal stem cells and CSCs for cancer therapy and explore potential future directions in this field. RESULTS Stem cells have been considered as important element of regenerative therapy by promoting tissue regeneration. Particularly, there is a growing trend to use stem cells as a target drug-delivery system to reduce undesirable side effects in non-target tissues. Noteworthy, studies on CSC-specific markers for distinguishing CSCs from normal stem cells and mature cancer cells have been conducted as a selective anti-cancer therapy with few side effects. Many researchers have also reported the development of various substances with anticancer effects by targeting CSCs from cancer tissues. CONCLUSION There has been a continuing increase in the number of studies on therapeutic stem cells and CSC-specific markers for selective diagnosis and therapy of cancer. This review focuses on the current status in the use of normal stem cells and CSCs for targeted cancer therapy. Future direction is also proposed.
Collapse
Affiliation(s)
- Seung-Woo Han
- Department of Biotechnology, The Catholic University of Korea, 43 Jibongro, Bucheon, 14662 Republic of Korea
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| | - Woo-Ju Kang
- Department of Biotechnology, The Catholic University of Korea, 43 Jibongro, Bucheon, 14662 Republic of Korea
| | - Hyoung-Chin Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do 28116 Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| | - Byeong-Cheol Kang
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
- Biomedical Center for Animal Resource and Development, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
- Designed Animal and Transplantation Research Institute, Institute of GreenBio Science Technology, Seoul National University, 1447 Pyeongchang-daero, Daehwa-myeon, Pyeongchang-gun, Gangwon-do 25354 Republic of Korea
| | - Jun-Won Yun
- Department of Biotechnology, The Catholic University of Korea, 43 Jibongro, Bucheon, 14662 Republic of Korea
| |
Collapse
|
72
|
Djema W, Bonnet C, Mazenc F, Clairambault J, Fridman E, Hirsch P, Delhommeau F. Control in dormancy or eradication of cancer stem cells: Mathematical modeling and stability issues. J Theor Biol 2018; 449:103-123. [PMID: 29678688 DOI: 10.1016/j.jtbi.2018.03.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/17/2018] [Accepted: 03/31/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Modeling and analysis of cell population dynamics enhance our understanding of cancer. Here we introduce and explore a new model that may apply to many tissues. ANALYSES An age-structured model describing coexistence between mutated and ordinary stem cells is developed and explored. The model is transformed into a nonlinear time-delay system governing the dynamics of healthy cells, coupled to a nonlinear differential-difference system describing dynamics of unhealthy cells. Its main features are highlighted and an advanced stability analysis of several steady states is performed, through specific Lyapunov-like functionals for descriptor-type systems. RESULTS We propose a biologically based model endowed with rich dynamics. It incorporates a new parameter representing immunoediting processes, including the case where proliferation of cancer cells is locally kept under check by the immune cells. It also considers the overproliferation of cancer stem cells, modeled as a subpopulation of mutated cells that is constantly active in cell division. The analysis that we perform here reveals the conditions of existence of several steady states, including the case of cancer dormancy, in the coupled model of interest. Our study suggests that cancer dormancy may result from a plastic sensitivity of mutated cells to their shared environment, different from that - fixed - of healthy cells, and this is related to an action (or lack of action) of the immune system. Next, the stability analysis that we perform is essentially oriented towards the determination of sufficient conditions, depending on all the model parameters, that ensure either a regionally (i.e., locally) stable dormancy steady state or eradication of unhealthy cells. Finally, we discuss some biological interpretations, with regards to our findings, in light of current and emerging therapeutics. These final insights are particularly formulated in the paradigmatic case of hematopoiesis and acute leukemia, which is one of the best known malignancies for which it is always hard, in presence of a clinical and histological remission, to decide between cure and dormancy of a tumoral clone.
Collapse
Affiliation(s)
- Walid Djema
- Inria Saclay, CentraleSupélec, Université Paris-Saclay & Inria Sophia-Antipolis, Biocore and McTao teams, Université Côte d'Azur (UCA), France.
| | - Catherine Bonnet
- Inria Saclay, Disco team, Université Paris-Saclay, CentraleSupélec, L2S (CNRS), France.
| | - Frédéric Mazenc
- Inria Saclay, Disco team, Université Paris-Saclay, CentraleSupélec, L2S (CNRS), France.
| | - Jean Clairambault
- Inria, Mamba team and Sorbonne Université, Paris 6, UPMC, Laboratoire Jacques-Louis Lions, Paris, France.
| | - Emilia Fridman
- Department of Electrical Engineering and Systems at the School of Electrical Engineering, Tel-Aviv, Israel.
| | - Pierre Hirsch
- Sorbonne Université, GRC n7, Groupe de Recherche Clinique sur les Myéloproliferations Aiguës et Chroniques, AP-HP, Hôpital Saint-Antoine, Paris F-75012, France.
| | - François Delhommeau
- Sorbonne Université, GRC n7, Groupe de Recherche Clinique sur les Myéloproliferations Aiguës et Chroniques, AP-HP, Hôpital Saint-Antoine, Paris F-75012, France.
| |
Collapse
|
73
|
Alteration in the Cytokine Secretion of Bone Marrow Stromal Cells from Patients with Chronic Myelomonocytic Leukemia Contribute to Impaired Hematopoietic Supportive Activity. Stem Cells Int 2018; 2018:5921392. [PMID: 30123289 PMCID: PMC6079359 DOI: 10.1155/2018/5921392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/03/2018] [Accepted: 05/15/2018] [Indexed: 12/20/2022] Open
Abstract
Bone marrow stromal cells (BMSCs) represent an important cellular component of the bone marrow microenvironment, which play an important role in supporting and regulating the proliferation and differentiation of hematopoietic stem/progenitor cells (HSPCs). We have previously reported that the ability of BMSCs derived from CMML patients (CMML-BMSCs) in supporting the expansion of cord blood (CB) CD34+ cells was significantly reduced compared to BMSCs derived from healthy donors (HD-BMSCs). In addition, CMML-BMSCs led to a skewed differentiation of CB CD34+ cells favoring myeloid lineage compared with HD-BMSCs. To assess whether the altered cytokine secretion was one of the mechanisms to mediate the impaired hematopoietic supportive activity of CMML-BMSCs, a transwell coculture followed by cytokine array was performed. We showed that noncontacted coculture with CMML-BMSCs preferentially promoted the differentiation of CB CD34+ cells toward myeloid lineage. The expression levels of multiple cytokines (IL-6, IL-8, and GRO-β) were markedly reduced in CMML-BMSCs compared with HD-BMSCs. By supplementing IL-6, IL-8, or GRO-β, the hematopoietic supportive activity of CMML-BMSCs was partially restored. These results suggested that BMSCs may contribute to the pathogenesis of CMML by altering their cytokine secretion, which will shed light on the further investigation to develop novel therapeutic strategies for CMML patients.
Collapse
|
74
|
Abstract
Purpose of review This article provides a brief overview of cancer-preventive phytochemicals specifically targeting pancreatic cancer (PC) stem cells for prevention and treatment. Recent findings Cancer stem cells (CSCs) represent a small proportion of the total cells of a given tumor, and contribute to tumor growth, recurrence, metastasis, and treatment resistance. Many intertwined pathways, including hedgehog, Wnt Signaling, and NOTCH, have been shown to play a role in the formation of CSCs. Recently, numerous chemopreventive agents, such as genistein, (-)-epigallocatechin-3-gallate (EGCG), sulforaphane, curcumin, resveratrol, and quercetin, have been shown to target CSCs mediated through the inhibition of multiple signalling pathways, to avoid toxicity and the side effects of chemical compounds. Summary A growing body of research suggests that CSCs are the drivers in treatment resistance, cancer recurrence, and metastasis, in addition to tumor initiation and heterogeneity. Patient survival depends on these CSCs, which are one cause of tumor recurrence after surgery and chemotherapy. Therefore, target selection; an improved understanding of CSC biology, the genetic and molecular profiles of CSCs, and their key signaling pathways, and; appropriate clinical trials endpoints that are designed to target CSCs will help in the development of drugs that will specifically target this small population of stem cells.
Collapse
|
75
|
LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. Hemasphere 2018; 2:e54. [PMID: 31723778 PMCID: PMC6745991 DOI: 10.1097/hs9.0000000000000054] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022] Open
Abstract
Supplemental Digital Content is available in the text Despite recent advances, the myeloproliferative neoplasms (MPNs) are attended by considerable morbidity and mortality. Janus kinase (Jak) inhibitors such as ruxolitinib manage symptoms but do not substantially change the natural history of the disease. In this report, we show the effects of IMG-7289, an irreversible inhibitor of the epigenetically active lysine-specific demethylase 1 (LSD1) in mouse models of MPN. Once-daily treatment with IMG-7289 normalized or improved blood cell counts, reduced spleen volumes, restored normal splenic architecture, and reduced bone marrow fibrosis. Most importantly, LSD1 inhibition lowered mutant allele burden and improved survival. IMG-7289 selectively inhibited proliferation and induced apoptosis of JAK2V617F cells by concomitantly increasing expression and methylation of p53, and, independently, the pro-apoptotic factor PUMA and by decreasing the levels of its antiapoptotic antagonist BCLXL. These data provide a molecular understanding of the disease-modifying activity of the LSD1 inhibitor IMG-7289 that is currently undergoing clinical evaluation in patients with high-risk myelofibrosis. Moreover, low doses of IMG-7289 and ruxolitinib synergize in normalizing the MPN phenotype in mice, offering a rationale for investigating combination therapy.
Collapse
|
76
|
Mitchell K, Barreyro L, Todorova TI, Taylor SJ, Antony-Debré I, Narayanagari SR, Carvajal LA, Leite J, Piperdi Z, Pendurti G, Mantzaris I, Paietta E, Verma A, Gritsman K, Steidl U. IL1RAP potentiates multiple oncogenic signaling pathways in AML. J Exp Med 2018; 215:1709-1727. [PMID: 29773641 PMCID: PMC5987926 DOI: 10.1084/jem.20180147] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/19/2018] [Accepted: 04/09/2018] [Indexed: 01/02/2023] Open
Abstract
The surface molecule interleukin-1 receptor accessory protein (IL1RAP) is consistently overexpressed across multiple genetic subtypes of acute myeloid leukemia (AML) and other myeloid malignancies, including at the stem cell level, and is emerging as a novel therapeutic target. However, the cell-intrinsic functions of IL1RAP in AML cells are largely unknown. Here, we show that targeting of IL1RAP via RNA interference, genetic deletion, or antibodies inhibits AML pathogenesis in vitro and in vivo, without perturbing healthy hematopoietic function or viability. Furthermore, we found that the role of IL1RAP is not restricted to the IL-1 receptor pathway, but that IL1RAP physically interacts with and mediates signaling and pro-proliferative effects through FLT3 and c-KIT, two receptor tyrosine kinases with known key roles in AML pathogenesis. Our study provides a new mechanistic basis for the efficacy of IL1RAP targeting in AML and reveals a novel role for this protein in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Laura Barreyro
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | | | - Samuel J Taylor
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | | | | | - Luis A Carvajal
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Joana Leite
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Zubair Piperdi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Gopichand Pendurti
- Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY
| | - Ioannis Mantzaris
- Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY
| | - Elisabeth Paietta
- Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY
| | - Amit Verma
- Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY
- Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY
| | - Kira Gritsman
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY
- Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY
- Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
77
|
Desai A, Zhang Y, Park Y, Dawson DM, Larusch GA, Kasturi L, Wald D, Ready JM, Gerson SL, Markowitz SD. A second-generation 15-PGDH inhibitor promotes bone marrow transplant recovery independently of age, transplant dose and granulocyte colony-stimulating factor support. Haematologica 2018; 103:1054-1064. [PMID: 29472361 PMCID: PMC6058768 DOI: 10.3324/haematol.2017.178376] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/20/2018] [Indexed: 01/09/2023] Open
Abstract
Hematopoietic stem cell transplantation following myeloablative chemotherapy is a curative treatment for many hematopoietic malignancies. However, profound granulocytopenia during the interval between transplantation and marrow recovery exposes recipients to risks of fatal infection, a significant source of transplant-associated morbidity and mortality. We have previously described the discovery of a small molecule, SW033291, that potently inhibits the prostaglandin degrading enzyme 15-PGDH, increases bone marrow prostaglandin E2, and accelerates hematopoietic recovery following murine transplant. Here we describe the efficacy of (+)-SW209415, a second-generation 15-PGDH inhibitor, in an expanded range of models relevant to human transplantation. (+)-SW209415 is 10,000-fold more soluble, providing the potential for intravenous delivery, while maintaining potency in inhibiting 15-PGDH, increasing in vivo prostaglandin E2, and accelerating hematopoietic regeneration following transplantation. In additional models, (+)-SW209415: (i) demonstrated synergy with granulocyte colony-stimulating factor, the current standard of care; (ii) maintained efficacy as transplant cell dose was escalated; (iii) maintained efficacy when transplant donors and recipients were aged; and (iv) potentiated homing in xenotransplants using human hematopoietic stem cells. (+)-SW209415 showed no adverse effects, no potentiation of in vivo growth of human myeloma and leukemia xenografts, and, on chronic high-dose administration, no toxicity as assessed by weight, blood counts and serum chemistry. These studies provide independent chemical confirmation of the activity of 15-PGDH inhibitors in potentiating hematopoietic recovery, extend the range of models in which inhibiting 15-PGDH demonstrates activity, allay concerns regarding potential for adverse effects from increasing prostaglandin E2, and thereby, advance 15-PGDH as a therapeutic target for potentiating hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Amar Desai
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yongyou Zhang
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Youngsoo Park
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Dawn M Dawson
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Gretchen A Larusch
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lakshmi Kasturi
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David Wald
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stanton L Gerson
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Sanford D Markowitz
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| |
Collapse
|
78
|
Xu H, Wen Q. Downregulation of miR‑135a predicts poor prognosis in acute myeloid leukemia and regulates leukemia progression via modulating HOXA10 expression. Mol Med Rep 2018; 18:1134-1140. [PMID: 29845297 DOI: 10.3892/mmr.2018.9066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/18/2018] [Indexed: 11/06/2022] Open
Abstract
MicroRNA‑135a (miR‑135a) has been shown to exert important roles in various human cancer types, such as glioblastoma, thyroid carcinoma and renal carcinoma. However, the function of miR‑135a in acute myeloid leukemia (AML) remains largely unknown. In the present study, it was demonstrated that miR‑135a expression was significantly downregulated in AML cells compared with normal control cells. Furthermore, the downregulation of miR‑135a in patients with AML predicted poor prognosis. Through functional experiments, overexpression of miR‑135a was demonstrated to significantly inhibit the proliferation and cell cycle of AML cells, while it promoted cellular apoptosis. miR‑135a directly targeted HOXA10 in AML cells. miR‑135a overexpression significantly suppressed the mRNA and protein levels of HOXA10 in AML cells. Moreover, there was an inverse association between miR‑135a expression and HOXA10 level in AML samples. Additionally, by ectopic expression of HOXA10, restoration of HOXA10 significantly abolished the effects of miR‑135a overexpression on AML cell proliferation, cell cycle and apoptosis. In conclusion, the present study demonstrated that miR‑135a serves as a tumor suppressor in AML by targeting HOXA10, and miR‑135a may be a promising prognostic biomarker for AML patients.
Collapse
Affiliation(s)
- Hongwei Xu
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Quan Wen
- General Internal Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
79
|
Arthur DE, Uzairu A, Mamza P, Abechi SE, Shallangwa GA. Structure-based optimization of tyrosine kinase inhibitors: a molecular docking study. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s13721-018-0170-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
80
|
Jiao J, Luo M, Wang R. Feedback regulation in a stem cell model with acute myeloid leukaemia. BMC SYSTEMS BIOLOGY 2018; 12:43. [PMID: 29745850 PMCID: PMC5998901 DOI: 10.1186/s12918-018-0561-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background The haematopoietic lineages with leukaemia lineages are considered in this paper. In particular, we mainly consider that haematopoietic lineages are tightly controlled by negative feedback inhibition of end-product. Actually, leukemia has been found 100 years ago. Up to now, the exact mechanism is still unknown, and many factors are thought to be associated with the pathogenesis of leukemia. Nevertheless, it is very necessary to continue the profound study of the pathogenesis of leukemia. Here, we propose a new mathematical model which include some negative feedback inhibition from the terminally differentiated cells of haematopoietic lineages to the haematopoietic stem cells and haematopoietic progenitor cells in order to describe the regulatory mechanisms mentioned above by a set of ordinary differential equations. Afterwards, we carried out detailed dynamical bifurcation analysis of the model, and obtained some meaningful results. Results In this work, we mainly perform the analysis of the mathematic model by bifurcation theory and numerical simulations. We have not only incorporated some new negative feedback mechanisms to the existing model, but also constructed our own model by using the modeling method of stem cell theory with probability method. Through a series of qualitative analysis and numerical simulations, we obtain that the weak negative feedback for differentiation probability is conducive to the cure of leukemia. However, with the strengthening of negative feedback, leukemia will be more difficult to be cured, and even induce death. In contrast, strong negative feedback for differentiation rate of progenitor cells can promote healthy haematopoiesis and suppress leukaemia. Conclusions These results demonstrate that healthy progenitor cells are bestowed a competitive advantage over leukaemia stem cells. Weak g1, g2, and h1 enable the system stays in the healthy state. However, strong h2 can promote healthy haematopoiesis and suppress leukaemia.
Collapse
Affiliation(s)
- Jianfeng Jiao
- Department of Mathematics, Shanghai University, Shangda Road No.99, Shanghai, 200444, China
| | - Min Luo
- Department of Mathematics, Shanghai University, Shangda Road No.99, Shanghai, 200444, China
| | - Ruiqi Wang
- Department of Mathematics, Shanghai University, Shangda Road No.99, Shanghai, 200444, China.
| |
Collapse
|
81
|
Si X, Zhang X, Hao X, Li Y, Chen Z, Ding Y, Shi H, Bai J, Gao Y, Cheng T, Yang FC, Zhou Y. Upregulation of miR-99a is associated with poor prognosis of acute myeloid leukemia and promotes myeloid leukemia cell expansion. Oncotarget 2018; 7:78095-78109. [PMID: 27801668 PMCID: PMC5363646 DOI: 10.18632/oncotarget.12947] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/14/2016] [Indexed: 01/05/2023] Open
Abstract
Leukemia stem cells (LSCs) can resist available treatments that results in disease progression and/or relapse. To dissect the microRNA (miRNA) expression signature of relapse in acute myeloid leukemia (AML), miRNA array analysis was performed using enriched LSCs from paired bone marrow samples of an AML patient at different disease stages. We identified that miR-99a was significantly upregulated in the LSCs obtained at relapse compared to the LSCs collected at the time of initial diagnosis. We also found that miR-99a was upregulated in LSCs compared to non-LSCs in a larger cohort of AML patients, and higher expression levels of miR-99a were significantly correlated with worse overall survival and event-free survival in these AML patients. Ectopic expression of miR-99a led to increased colony forming ability and expansion in myeloid leukemia cells after exposure to chemotherapeutic drugs in vitro and in vivo, partially due to overcoming of chemotherapeutic agent-mediated cell cycle arrest. Gene profiling and bioinformatic analyses indicated that ectopic expression of miR-99a significantly upregulated genes that are critical for LSC maintenance, cell cycle, and downstream targets of E2F and MYC. This study suggests that miR-99a has a novel role and potential use as a biomarker in myeloid leukemia progression.
Collapse
Affiliation(s)
- Xiaohui Si
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiaoyun Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xing Hao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yunan Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zizhen Chen
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yahui Ding
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hui Shi
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jie Bai
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China.,Collaborative Innovation Center for Cancer Medicine, Tianjin, China
| | - Feng-Chun Yang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.,Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China
| |
Collapse
|
82
|
Kumar R, Godavarthy PS, Krause DS. The bone marrow microenvironment in health and disease at a glance. J Cell Sci 2018; 131:131/4/jcs201707. [PMID: 29472498 DOI: 10.1242/jcs.201707] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The bone marrow microenvironment (BMM) is the 'domicile' of hematopoietic stem cells, as well as of malignant processes that can develop there. Multiple and complex interactions with the BMM influence hematopoietic stem cell (HSC) physiology, but also the pathophysiology of hematological malignancies. Reciprocally, hematological malignancies alter the BMM, in order to render it more hospitable for malignant progression. In this Cell Science at a Glance article and accompanying poster, we highlight concepts of the normal and malignant hematopoietic stem cell niches. We present the intricacies of the BMM in malignancy and provide approaches for targeting the interactions between malignant cells and their BMM. This is done in an effort to augment existing treatment strategies in the future.
Collapse
Affiliation(s)
- Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, D-60596 Frankfurt am Main, Germany
| | - P Sonika Godavarthy
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, D-60596 Frankfurt am Main, Germany
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str. 42-44, D-60596 Frankfurt am Main, Germany
| |
Collapse
|
83
|
Chen C, Zhuang Y, Chen X, Chen X, Li D, Fan Y, Xu J, Chen Y, Wu L. Hsp90 N- and C-terminal double inhibition synergistically suppresses Bcr-Abl-positive human leukemia cells. Oncotarget 2018; 8:10025-10036. [PMID: 28036294 PMCID: PMC5354638 DOI: 10.18632/oncotarget.14324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/15/2016] [Indexed: 12/23/2022] Open
Abstract
Heat shock protein 90 (Hsp90) contains amino (N)–terminal domain, carboxyl(C)-terminal domain, and middle domains, which activate Hsp90 chaperone function cooperatively in tumor cells. One terminal occupancy might influence another terminal binding with inhibitor. The Bcr-Abl kinase is one of the Hsp90 clients implicated in the pathogenesis of chronic myeloid leukemia (CML). Present studies demonstrate that double inhibition of the N- and C-terminal termini can disrupt Hsp90 chaperone function synergistically, but not antagonistically, in Bcr-Abl-positive human leukemia cells. Furthermore, both the N-terminal inhibitor 17-AAG and the C-terminal inhibitor cisplatin (CP) have the capacity to suppress progenitor cells; however, only CP is able to inhibit leukemia stem cells (LSCs) significantly, which implies that the combinational treatment is able to suppress human leukemia in different mature states.
Collapse
Affiliation(s)
- Chun Chen
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Yingting Zhuang
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Xianling Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, China
| | - Xiaole Chen
- Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Deptartment of Biopharmaceutics, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Ding Li
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Yingjuan Fan
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Jianhua Xu
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| | - Yuanzhong Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, China
| | - Lixian Wu
- Deptartment of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Institute of Materia Medica, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China.,Fuijan Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, China
| |
Collapse
|
84
|
Plesa A, Dumontet C, Mattei E, Tagoug I, Hayette S, Sujobert P, Tigaud I, Pages MP, Chelghoum Y, Baracco F, Labussierre H, Ducastelle S, Paubelle E, Nicolini FE, Elhamri M, Campos L, Plesa C, Morisset S, Salles G, Bertrand Y, Michallet M, Thomas X. High frequency of CD34+CD38-/low immature leukemia cells is correlated with unfavorable prognosis in acute myeloid leukemia. World J Stem Cells 2017; 9:227-234. [PMID: 29321824 PMCID: PMC5746643 DOI: 10.4252/wjsc.v9.i12.227] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 10/17/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the importance of the CD34+CD38- cell population when compared to the CD34+CD38+/low and CD34+CD38+/high leukemic cell sub-populations and to determine its correlations with leukemia characteristics and known prognostic factors, as well as with response to therapy and survival.
METHODS Two hundred bone marrow samples were obtained at diagnosis from 200 consecutive patients with newly diagnosed acute myeloid leukemia (AML) were studied between September 2008 and December 2010 at our Institution (Hematology Department, Lyon, France). The CD34/CD38 cell profile was analyzed by multiparameter flowcytometry approach using 8C panels and FACS CANTO and Diva software (BD Bioscience).
RESULTS We analyzed CD34 and CD38 expression in bone marrow samples of 200 AML patients at diagnosis, and investigated the prognostic value of the most immature CD34+CD38- population. Using a cut-off value of 1% of CD34+CD38- from total “bulk leukemic cells” we found that a high (> 1%) level of CD34+CD38- blasts at diagnosis was correlated with advanced age, adverse cytogenetics as well as with a lower rate of complete response after induction and shorter disease-free survival. In a multivariate analysis considering age, leukocytosis, the % of CD34+ blasts cells and the standardized cytogenetic and molecular risk subgroups, a percentage of CD34+CD38- leukemic cells > 1% was an independent predictor of DFS [HR = 2.8 (1.02-7.73), P = 0.04] and OS [HR = 2.65 (1.09-6.43), P = 0.03].
CONCLUSION Taken together, these results show that a CD34/CD38 “backbone” for leukemic cell analysis by multicolour flowcytometry at diagnosis provides useful prognostic information.
Collapse
Affiliation(s)
- Adriana Plesa
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
- CRCL, INSERM 1052/CNRS 5286, Lyon FR-69008, France
| | - Charles Dumontet
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
- CRCL, INSERM 1052/CNRS 5286, Lyon FR-69008, France
| | - Eve Mattei
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Ines Tagoug
- CRCL, INSERM 1052/CNRS 5286, Lyon FR-69008, France
| | - Sandrine Hayette
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Pierre Sujobert
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Isabelle Tigaud
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Marie Pierre Pages
- Laboratory of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Youcef Chelghoum
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Fiorenza Baracco
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Helene Labussierre
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Sophie Ducastelle
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Etienne Paubelle
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | | | - Mohamed Elhamri
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Lydia Campos
- Laboratory of Hematology, Nord Hospital, Saint Etienne 42055, France
| | - Claudiu Plesa
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Stéphane Morisset
- Statistical and Clinical Research, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Gilles Salles
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Yves Bertrand
- Department of Pediatric Hematology and BMT, IHOP Lyon, Lyon 69001, France
| | - Mauricette Michallet
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| | - Xavier Thomas
- Department of Hematology, Hospices Civils de Lyon, Pierre - Bénite Cedex 69495, France
| |
Collapse
|
85
|
El-Badawy A, Ghoneim MA, Gabr MM, Salah RA, Mohamed IK, Amer M, El-Badri N. Cancer cell-soluble factors reprogram mesenchymal stromal cells to slow cycling, chemoresistant cells with a more stem-like state. Stem Cell Res Ther 2017; 8:254. [PMID: 29115987 PMCID: PMC5688803 DOI: 10.1186/s13287-017-0709-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/24/2017] [Accepted: 10/23/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) play different roles in modulating tumor progression, growth, and metastasis. MSCs are recruited to the tumor site in large numbers and subsequently have an important microenvironmental role in modulating tumor progression and drug sensitivity. However, the effect of the tumor microenvironment on MSC plasticity remains poorly understood. Herein, we report a paracrine effect of cancer cells, in which they secrete soluble factors that promote a more stem-like state in bone marrow mesenchymal stem cells (BM-MSCs). METHODS The effect of soluble factors secreted from MCF7, Hela, and HepG2 cancer cell lines on BM-MSCs was assessed using a Transwell indirect coculture system. After 5 days of coculture, BM-MSCs were characterized by flow cytometry for surface marker expression, by qPCR for gene expression profile, and by confocal immunofluorescence for marker expression. We then measured the sensitivity of cocultured BM-MSCs to chemotherapeutic agents, their cell cycle profile, and their response to DNA damage. The sphere formation, invasive properties, and in-vivo performance of BM-MSCs after coculture with cancer cells were also measured. RESULTS Indirect coculture of cancer cells and BM-MSCs, without direct cell contact, generated slow cycling, chemoresistant spheroid stem cells that highly expressed markers of pluripotency, cancer cells, and cancer stem cells (CSCs). They also displayed properties of a side population and enhanced sphere formation in culture. Accordingly, these cells were termed cancer-induced stem cells (CiSCs). CiSCs showed a more mesenchymal phenotype that was further augmented upon TGF-β stimulation and demonstrated a high expression of the β-catenin pathway and ALDH1A1. CONCLUSIONS These findings demonstrate that MSCs, recruited to the tumor microenvironment in large numbers, may display cellular plasticity, acquire a more stem-like state, and acquire some properties of CSCs upon exposure to cancer cell-secreted factors. These acquired characteristics may contribute to tumor progression, survival, and metastasis. Our findings provide new insights into the interactions between MSCs and cancer cells, with the potential to identify novel molecular targets for cancer therapy.
Collapse
Affiliation(s)
- Ahmed El-Badawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | | | - Mahmoud M. Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Ihab K. Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Marwa Amer
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| |
Collapse
|
86
|
Hanekamp D, Denys B, Kaspers GJL, Te Marvelde JG, Schuurhuis GJ, De Haas V, De Moerloose B, de Bont ES, Zwaan CM, de Jong A, Depreter B, Lammens T, Philippé J, Cloos J, van der Velden VHJ. Leukaemic stem cell load at diagnosis predicts the development of relapse in young acute myeloid leukaemia patients. Br J Haematol 2017; 183:512-516. [PMID: 29076143 DOI: 10.1111/bjh.14991] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Diana Hanekamp
- Department of Haematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Barbara Denys
- Department of Laboratory Medicine, Haematology, Ghent University Hospital, Ghent, Belgium
| | - Gertjan J L Kaspers
- Department of Paediatric Oncology/Haematology, VU University Medical Center, Amsterdam, the Netherlands.,Princess Máxima Center for Paediatric Oncology, Utrecht, the Netherlands
| | - Jeroen G Te Marvelde
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | | | - Barbara De Moerloose
- Department of Paediatric Haematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Eveline S de Bont
- Department of Paediatric Oncology/Haematology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - C Michel Zwaan
- Department of Paediatric Oncology/Haematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Anja de Jong
- Dutch Childhood Oncology Group, The Hague, the Netherlands
| | - Barbara Depreter
- Department of Paediatric Haematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium.,Department of Clinical Biology, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Tim Lammens
- Department of Paediatric Haematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Jan Philippé
- Department of Laboratory Medicine, Haematology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium.,Department of Clinical Biology, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Jacqueline Cloos
- Department of Haematology, VU University Medical Center, Amsterdam, the Netherlands.,Department of Paediatric Oncology/Haematology, VU University Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
87
|
Wiehle L, Raddatz G, Pusch S, Gutekunst J, von Deimling A, Rodríguez-Paredes M, Lyko F. mIDH-associated DNA hypermethylation in acute myeloid leukemia reflects differentiation blockage rather than inhibition of TET-mediated demethylation. Cell Stress 2017; 1:55-67. [PMID: 31225434 PMCID: PMC6551656 DOI: 10.15698/cst2017.10.106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Isocitrate dehydrogenases 1 and 2 (IDH1/2) are recurrently mutated in acute myeloid leukemia (AML), but their mechanistic role in leukemogenesis is poorly understood. The inhibition of TET enzymes by D-2-hydroxyglutarate (D-2-HG), which is produced by mutant IDH1/2 (mIDH1/2), has been suggested to promote epigenetic deregulation during tumorigenesis. In addition, mIDH also induces a differentiation block in various cell culture and mouse models. Here we analyze the genomic methylation patterns of AML patients with mIDH using Infinium 450K data from a large AML cohort and found that mIDH is associated with pronounced DNA hypermethylation at tens of thousands of CpGs. Interestingly, however, myeloid leukemia cells overexpressing mIDH, cells that were cultured in the presence of D-2-HG or TET2 mutant AML patients did not show similar methylation changes. In further analyses, we also characterized the methylation landscapes of myeloid progenitor cells and analyzed their relationship to mIDH-associated hypermethylation. Our findings identify the differentiation state of myeloid cells, rather than inhibition of TET-mediated DNA demethylation, as a major factor of mIDH-associated hypermethylation in AML. Furthermore, our results are also important for understanding the mode of action of currently developed mIDH inhibitors.
Collapse
Affiliation(s)
- Laura Wiehle
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center; 69120 Heidelberg, Germany
| | - Günter Raddatz
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center; 69120 Heidelberg, Germany
| | - Stefan Pusch
- German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Julian Gutekunst
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center; 69120 Heidelberg, Germany
| | - Andreas von Deimling
- German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Manuel Rodríguez-Paredes
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center; 69120 Heidelberg, Germany
| | - Frank Lyko
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center; 69120 Heidelberg, Germany
| |
Collapse
|
88
|
Rajaraman R, Guernsey D, Rajaraman M, Rajaraman S. Neosis - A Parasexual Somatic Reduction Division in Cancer. INT J HUM GENET 2017. [DOI: 10.1080/09723757.2007.11885983] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- R. Rajaraman
- Department of Medicine, Division of Hematology, QEII Health Sciences Center, Dalhousie University, Halifax, NS. B3H 1X5, Canada
| | - D.L. Guernsey
- Department of Pathology QEII Health Sciences Center, Dalhousie University, Halifax, NS. B3H 1X5, Canada
| | - M.M. Rajaraman
- Nova Scotia Cancer Centre, Department of Radiation Oncology, QEII Health Sciences Center, Dalhousie University, Halifax, NS. B3H 1X5, Canada
| | - S.R. Rajaraman
- Department of Radiation Oncology, QEII Health Sciences Center, Dalhousie University, Downtown Clinic, Windsor, ON, N9A 1G5, Canada
| |
Collapse
|
89
|
Jung HJ. Chemical Proteomic Approaches Targeting Cancer Stem Cells: A Review of Current Literature. Cancer Genomics Proteomics 2017; 14:315-327. [PMID: 28870999 PMCID: PMC5611518 DOI: 10.21873/cgp.20042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 12/24/2022] Open
Abstract
Cancer stem cells (CSCs) have been proposed as central drivers of tumor initiation, progression, recurrence, and therapeutic resistance. Therefore, identifying stem-like cells within cancers and understanding their properties is crucial for the development of effective anticancer therapies. Recently, chemical proteomics has become a powerful tool to efficiently determine protein networks responsible for CSC pathophysiology and comprehensively elucidate molecular mechanisms of drug action against CSCs. This review provides an overview of major methodologies utilized in chemical proteomic approaches. In addition, recent successful chemical proteomic applications targeting CSCs are highlighted. Future direction of potential CSC research by integrating chemical genomic and proteomic data obtained from a single biological sample of CSCs are also suggested in this review.
Collapse
Affiliation(s)
- Hye Jin Jung
- Department of BT-Convergent Pharmaceutical Engineering, Sun Moon University, Asan, Republic of Korea
| |
Collapse
|
90
|
Carvajal LA, Steidl U. Eliminating Cancer Stem Cells in CML with Combination Transcriptional Therapy. Cell Stem Cell 2017; 19:6-8. [PMID: 27392220 DOI: 10.1016/j.stem.2016.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Leukemia stem cells (LSCs) are resistant to current therapies used to treat chronic myeloid leukemia (CML). Abraham et al. (2016) have identified a molecular network critical for CML LSC survival and propose that simultaneously targeting two of their major transcriptional regulators, p53 and c-Myc, may facilitate their eradication.
Collapse
Affiliation(s)
- Luis A Carvajal
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine (Oncology), Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, NY 10467, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
91
|
Li Y, Lin K, Yang Z, Han N, Quan X, Guo X, Li C. Bladder cancer stem cells: clonal origin and therapeutic perspectives. Oncotarget 2017; 8:66668-66679. [PMID: 29029546 PMCID: PMC5630446 DOI: 10.18632/oncotarget.19112] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/17/2017] [Indexed: 12/21/2022] Open
Abstract
In this article, we review the origin and therapeutic perspectives of bladder cancer stem cells (BCSCs), which are integral to the initiation, high recurrence and chemoresistance of bladder cancer. BCSCs are heterogenous and originate from multiple cell types, including urothelial stem cells and differentiated cell types, including basal, intermediate stratum and umbrella cells. Cell surface markers, including CD44, CD67LR, EMA, ALDH1A1 and BCMab1, are used to identify and isolate BCSCs. The Hedgehog, Notch, Wnt and JAK-STAT signaling pathways play key roles in maintaining the stemness, self-renewal and proliferative potential of BCSCs. High expression of ABC transporters, acetaldehyde dehydrogenase, antioxidants and apoptosis resistance proteins in BCSCs play a critical role in chemoresistance. Consequently, a greater understanding of the biology of BCSCs will be important for identifying effective therapeutic targets to improve clinical outcomes for bladder cancer patients.
Collapse
Affiliation(s)
- Yi Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Kaisu Lin
- Department of Oncology, the Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, China
| | - Zhao Yang
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ning Han
- Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofang Quan
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Chong Li
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Beijing Jianlan Institute of Medicine, Beijing, China
| |
Collapse
|
92
|
Galán-Díez M, Kousteni S. The osteoblastic niche in hematopoiesis and hematological myeloid malignancies. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:53-62. [PMID: 29098141 PMCID: PMC5662025 DOI: 10.1007/s40610-017-0055-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review focuses on evidence highlighting the bidirectional crosstalk between the hematopoietic stem cell (HSC) and their surrounding stromal cells, with a particular emphasis on cells of the osteoblast lineage. The role and molecular functions of osteoblasts in normal hematopoiesis and in myeloid hematological malignancies is discussed. RECENT FINDINGS Cells of the osteoblast lineage have emerged as potent regulators of HSC expansion that regulate their recruitment and, depending on their stage of differentiation, their activity, proliferation and differentiation along the lymphoid, myeloid and erythroid lineages. In addition, mutations in mature osteoblasts or their progenitors induce myeloid malignancies. Conversely, signals from myelodysplastic cells can remodel the osteoblastic niche to favor self-perpetuation. SUMMARY Understanding cellular crosstalk between osteoblastic cells and HSCs in the bone marrow microenvironment is of fundamental importance for developing therapies against benign and malignant hematological diseases.
Collapse
Affiliation(s)
- Marta Galán-Díez
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, New York 10032, USA
| | - Stavroula Kousteni
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, New York 10032, USA
| |
Collapse
|
93
|
Owen HC, Appiah S, Hasan N, Ghali L, Elayat G, Bell C. Phytochemical Modulation of Apoptosis and Autophagy: Strategies to Overcome Chemoresistance in Leukemic Stem Cells in the Bone Marrow Microenvironment. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 135:249-278. [PMID: 28807161 DOI: 10.1016/bs.irn.2017.02.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Advances in scientific research and targeted treatment regimes have improved survival rates for many cancers over the past few decades. However, for some types of leukemia, including acute lymphoblastic and acute myeloid leukemia, mortality rates have continued to rise, with chemoresistance in leukemic stem cells (LSCs) being a major contributing factor. Most cancer drug therapies act by inducing apoptosis in dividing cells but are ineffective in targeting quiescent LSCs. Niches in the bone marrow, known as leukemic niches, behave as "sanctuaries" where LSCs acquire drug resistance. This review explores the role of the bone marrow environment in the maintenance of LSCs and its contribution to chemoresistance and considers current research on the potential use of phytochemicals to overcome chemoresistance through the modulation of signaling pathways involved in the survival and death of leukemic clonal cells and/or leukemic stem cells. Phytochemicals from traditional Chinese medicine, namely baicalein, chrysin, wogonin (constituents of Scutellaria baicalensis; huáng qín; ), curcumin (a constituent of Curcuma longa, jiāng huáng, ), and resveratrol (a constituent of Polygonum cuspidatum; hŭ zhàng, ) have been shown to induce apoptosis in leukemic cell lines, with curcumin and resveratrol also causing cell death via the induction of autophagy (a nonapoptotic pathway). In order to be effective in eliminating LSCs, it is important to target signaling pathways (such as Wnt/β-catenin, Notch, and Hedgehog). Resveratrol has been reported to induce apoptosis in leukemic cells through the inhibition of the Notch and Sonic hedgehog signaling pathways, therefore showing potential to affect LSCs. While these findings are of interest, there is a lack of reported research on the modulatory effect of phytochemicals on the autophagic cell death pathway in leukemia, and on the signaling pathways involved in the maintenance of LSCs, highlighting the need for further work in these areas.
Collapse
Affiliation(s)
- Helen C Owen
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom.
| | - Sandra Appiah
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom.
| | - Noor Hasan
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| | - Lucy Ghali
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| | - Ghada Elayat
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| | - Celia Bell
- Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, United Kingdom
| |
Collapse
|
94
|
Zhou ZL, Ma J, Tong MH, Chan BP, Wong AST, Ngan AHW. Nanomechanical measurement of adhesion and migration of leukemia cells with phorbol 12-myristate 13-acetate treatment. Int J Nanomedicine 2016; 11:6533-6545. [PMID: 27994457 PMCID: PMC5153271 DOI: 10.2147/ijn.s118065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The adhesion and traction behavior of leukemia cells in their microenvironment is directly linked to their migration, which is a prime issue affecting the release of cancer cells from the bone marrow and hence metastasis. In assessing the effectiveness of phorbol 12-myristate 13-acetate (PMA) treatment, the conventional batch-cell transwell-migration assay may not indicate the intrinsic effect of the treatment on migration, since the treatment may also affect other cellular behavior, such as proliferation or death. In this study, the pN-level adhesion and traction forces between single leukemia cells and their microenvironment were directly measured using optical tweezers and traction-force microscopy. The effects of PMA on K562 and THP1 leukemia cells were studied, and the results showed that PMA treatment significantly increased cell adhesion with extracellular matrix proteins, bone marrow stromal cells, and human fibroblasts. PMA treatment also significantly increased the traction of THP1 cells on bovine serum albumin proteins, although the effect on K562 cells was insignificant. Western blots showed an increased expression of E-cadherin and vimentin proteins after the leukemia cells were treated with PMA. The study suggests that PMA upregulates adhesion and thus suppresses the migration of both K562 and THP1 cells in their microenvironment. The ability of optical tweezers and traction-force microscopy to measure directly pN-level cell–protein or cell–cell contact was also demonstrated.
Collapse
Affiliation(s)
| | - Jing Ma
- School of Biological Sciences, University of Hong Kong, Hong Kong, People's Republic of China
| | | | | | - Alice Sze Tsai Wong
- School of Biological Sciences, University of Hong Kong, Hong Kong, People's Republic of China
| | | |
Collapse
|
95
|
Olivos DJ, Mayo LD. Emerging Non-Canonical Functions and Regulation by p53: p53 and Stemness. Int J Mol Sci 2016; 17:ijms17121982. [PMID: 27898034 PMCID: PMC5187782 DOI: 10.3390/ijms17121982] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/15/2023] Open
Abstract
Since its discovery nearly 40 years ago, p53 has ascended to the forefront of investigated genes and proteins across diverse research disciplines and is recognized most exclusively for its role in cancer as a tumor suppressor. Levine and Oren (2009) reviewed the evolution of p53 detailing the significant discoveries of each decade since its first report in 1979. In this review, we will highlight the emerging non-canonical functions and regulation of p53 in stem cells. We will focus on general themes shared among p53's functions in non-malignant stem cells and cancer stem-like cells (CSCs) and the influence of p53 on the microenvironment and CSC niche. We will also examine p53 gain of function (GOF) roles in stemness. Mutant p53 (mutp53) GOFs that lead to survival, drug resistance and colonization are reviewed in the context of the acquisition of advantageous transformation processes, such as differentiation and dedifferentiation, epithelial-to-mesenchymal transition (EMT) and stem cell senescence and quiescence. Finally, we will conclude with therapeutic strategies that restore wild-type p53 (wtp53) function in cancer and CSCs, including RING finger E3 ligases and CSC maintenance. The mechanisms by which wtp53 and mutp53 influence stemness in non-malignant stem cells and CSCs or tumor-initiating cells (TICs) are poorly understood thus far. Further elucidation of p53's effects on stemness could lead to novel therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lindsey D Mayo
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
96
|
MacLean AL, Lo Celso C, Stumpf MP. Concise Review: Stem Cell Population Biology: Insights from Hematopoiesis. Stem Cells 2016; 35:80-88. [DOI: 10.1002/stem.2508] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/19/2016] [Accepted: 08/21/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Adam L. MacLean
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| | - Cristina Lo Celso
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| | - Michael P.H. Stumpf
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| |
Collapse
|
97
|
He L, Gu J, Lim LY, Yuan ZX, Mo J. Nanomedicine-Mediated Therapies to Target Breast Cancer Stem Cells. Front Pharmacol 2016; 7:313. [PMID: 27679576 PMCID: PMC5020043 DOI: 10.3389/fphar.2016.00313] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidences have suggested the existence of breast cancer stem cells (BCSCs), which possess the potential of both self-renewal and differentiation. The origin of BCSCs might have relationship to the development of normal mammary stem cells. BCSCs are believed to play a key role in the initiation, recurrence and chemo-/radiotherapy resistances of breast cancer. Therefore, elimination of BCSCs is crucial for breast cancer therapy. However, conventional chemo and radiation therapies cannot eradicate BCSCs effectively. Fortunately, nanotechnology holds great potential for specific and efficient anti-BCSCs treatment. “Smart” nanocarriers can distinguish BCSCs from the other breast cancer cells and selectively deliver therapeutic agents to the BCSCs. Emerging findings suggest that BCSCs in breast cancer could be successfully inhibited and even eradicated by functionalized nanomedicines. In this review, we focus on origin of BCSCs, strategies used to target BCSCs, and summarize the nanotechnology-based delivery systems that have been applied for eliminating BCSCs in breast cancer.
Collapse
Affiliation(s)
- Lili He
- College of Pharmacy, Southwest University for Nationalities Chengdu, China
| | - Jian Gu
- College of Pharmacy, Southwest University for Nationalities Chengdu, China
| | - Lee Y Lim
- Pharmacy, School of Medicine and Pharmacology, The University of Western Australia, Crawley WA, Australia
| | - Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education Guangzhou, China
| |
Collapse
|
98
|
Aldehyde dehydrogenases inhibition eradicates leukemia stem cells while sparing normal progenitors. Blood Cancer J 2016; 6:e469. [PMID: 27611922 PMCID: PMC5056970 DOI: 10.1038/bcj.2016.78] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022] Open
Abstract
The vast majority of patients with acute myeloid leukemia (AML) achieve complete remission (CR) after standard induction chemotherapy. However, the majority subsequently relapse and die of the disease. A leukemia stem cell (LSC) paradigm has been invoked to explain this failure of CR to reliably translate into cure. Indeed, LSCs are highly enriched in CD34+CD38− leukemic cells that exhibit positive aldehyde dehydrogenase activity (ALDH+) on flow cytometry, these LSCs are resistant to currently existing treatments in AML such as cytarabine and anthracycline that, at the cost of great toxicity on normal cells, are highly active against the leukemic bulk, but spare the LSCs responsible for relapse. To try to combat the LSC population selectively, a well-characterized ALDH inhibitor by the trivial name of dimethyl ampal thiolester (DIMATE) was assessed on sorted CD34+CD38− subpopulations from AML patients and healthy patients. ALDH activity and cell viability were monitored by flow cytometry. From enzyme kinetic studies DIMATE is an active enzyme-dependent, competitive, irreversible inhibitor of ALDH1. On cells in culture, DIMATE is a powerful inhibitor of ALDHs 1 and 3, has a major cytotoxic activity on human AML cell lines. Moreover, DIMATE is highly active against leukemic populations enriched in LSCs, but, unlike conventional chemotherapy, DIMATE is not toxic for healthy hematopoietic stem cells which retained, after treatment, their self-renewing and multi-lineage differentiation capacity in immunodeficient mice, xenografted with human leukemic cells. DIMATE eradicates specifically human AML cells and spares healthy mouse hematologic cells.
Collapse
|
99
|
MicroRNA-139-5p regulates proliferation of hematopoietic progenitors and is repressed during BCR-ABL-mediated leukemogenesis. Blood 2016; 128:2117-2129. [PMID: 27605510 DOI: 10.1182/blood-2016-02-702464] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as important regulators of the immune system. However, despite this prominence, our understanding of the function of miRNAs in the early hematopoietic stages is incomplete. In this study, we found that miR-139-5p negatively regulated the proliferation of hematopoietic stem cells and progenitor cells and that downregulation of miR-139-5p expression was associated with hematopoietic malignancy, such as chronic myeloid leukemia (CML). Knockdown of miR-139-5p resulted in myeloid-biased differentiation with expansion of myeloid progenitor cells. In contrast, miR-139-5p expression inhibited the proliferation of hematopoietic progenitors and resulted in the remission of a CML-like disease that is induced by breakpoint cluster region-Abelson (BCR-ABL) transformation. We also found that Brg1 is a functional target of miR-139-5p and that Brg1 is involved in BCR-ABL-induced leukemogenesis. Thus, our results identify miR-139-5p as a key regulator of cellular proliferation during early hematopoiesis and suggest that it is a potent antileukemic molecule.
Collapse
|
100
|
Chakraborty SN, Leng X, Perazzona B, Sun X, Lin YH, Arlinghaus RB. Combination of JAK2 and HSP90 inhibitors: an effective therapeutic option in drug-resistant chronic myelogenous leukemia. Genes Cancer 2016; 7:201-208. [PMID: 27551334 PMCID: PMC4979592 DOI: 10.18632/genesandcancer.111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Recent studies suggest that JAK2 serves as a novel therapeutic target in Bcr-Abl+ chronic myelogenous leukemia (CML). We have reported the existence of an HSP90- associated high molecular weight network complex (HMWNC) that is composed of HSP90 client proteins BCR-ABL, JAK2, and STAT3 in wild type Bcr-Abl+ leukemic cells. Here we showed that the HSP90-HMWNC is present in leukemia cells from CML patients in blast stage, and in Imatinib (IM)-resistant 32Dp210 (T315I) leukemia cells. We found that the HSP90-HMWNC could be disassembled by depleting JAK2 with either Jak2-specific shRNA or treatment with JAK2 inhibitors (TG101209 or Ruxolitinib) and HSP90 inhibitor (AUY922). Combinational treatment with JAK2 and HSP90 inhibitors diminished the activation of BCR-ABL, JAK2 and its downstream targets. As a result, the IM-resistant 32Dp210 T315I cells underwent apoptosis. When administered in mice bearing 32Dp210 T315I leukemia, combinational therapy using Ruxolitinib and AUY922 prolonged the survival significantly. Thus, a combination of JAK2 and HSP90 inhibitors could be a powerful strategy for the treatment of CML, especially in IM-resistant patients.
Collapse
Affiliation(s)
- Sandip N Chakraborty
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Xiaohong Leng
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Bastianella Perazzona
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Xiaoping Sun
- Department of Laboratory Medicine, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Yu-Hsi Lin
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| |
Collapse
|