1
|
Liu Y, Zheng L, Li Y, Ma L, Zheng N, Liu X, Zhao Y, Yu L, Liu N, Liu S, Zhang K, Zhou J, Wei M, Yang C, Yang G. Neratinib impairs function of m6A recognition on AML1-ETO pre-mRNA and induces differentiation of t (8;21) AML cells by targeting HNRNPA3. Cancer Lett 2024; 594:216980. [PMID: 38797229 DOI: 10.1016/j.canlet.2024.216980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/07/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Acute myeloid leukemia (AML) is frequently linked to genetic abnormalities, with the t (8; 21) translocation, resulting in the production of a fusion oncoprotein AML1-ETO (AE), being a prevalent occurrence. This protein plays a pivotal role in t (8; 21) AML's onset, advancement, and recurrence, making it a therapeutic target. However, the development of drug molecules targeting AML1-ETO are markedly insufficient, especially used in clinical treatment. In this study, it was uncovered that Neratinib could significantly downregulate AML1-ETO protein level, subsequently promoting differentiation of t (8; 21) AML cells. Based on "differentiated active" probes, Neratinib was identified as a functional inhibitor against HNRNPA3 through covalent binding. The further studies demonstrated that HNRNPA3 function as a putative m6A reader responsible for recognizing and regulating the alternative splicing of AML-ETO pre-mRNA. These findings not only contribute to a novel insight to the mechanism governing post-transcriptional modification of AML1-ETO transcript, but also suggest that Neratinib would be promising therapeutic potential for t (8; 21) AML treatment.
Collapse
MESH Headings
- Humans
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Quinolines/pharmacology
- Cell Differentiation/drug effects
- RUNX1 Translocation Partner 1 Protein/genetics
- RUNX1 Translocation Partner 1 Protein/metabolism
- RNA Precursors/metabolism
- RNA Precursors/genetics
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics
- Translocation, Genetic/drug effects
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Adenosine/pharmacology
- Alternative Splicing/drug effects
- Cell Line, Tumor
- Animals
- Mice
Collapse
Affiliation(s)
- Yulin Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Liting Zheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Ying Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Lan Ma
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Nan Zheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Xinhua Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Yanli Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Li Yu
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Xueyuan AVE 1098, Nanshan District, Shenzhen, Guangdong, 518000, PR China
| | - Ning Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Shuangwei Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Kun Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Jingfeng Zhou
- Department of Hematology and Oncology, International Cancer Center, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University Health Science Center, Xueyuan AVE 1098, Nanshan District, Shenzhen, Guangdong, 518000, PR China.
| | - Mingming Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Guang Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
2
|
Frankel NW, Deng H, Yucel G, Gainer M, Leemans N, Lam A, Li Y, Hung M, Lee D, Lee CT, Banicki A, Tian M, Almudhfar N, Naitmazi L, Roguev A, Lee S, Wong W, Gordley R, Lu TK, Garrison BS. Precision off-the-shelf natural killer cell therapies for oncology with logic-gated gene circuits. Cell Rep 2024; 43:114145. [PMID: 38669141 DOI: 10.1016/j.celrep.2024.114145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive disease with a poor prognosis (5-year survival rate of 30.5% in the United States). Designing cell therapies to target AML is challenging because no single tumor-associated antigen (TAA) is highly expressed on all cancer subpopulations. Furthermore, TAAs are also expressed on healthy cells, leading to toxicity risk. To address these targeting challenges, we engineer natural killer (NK) cells with a multi-input gene circuit consisting of chimeric antigen receptors (CARs) controlled by OR and NOT logic gates. The OR gate kills a range of AML cells from leukemic stem cells to blasts using a bivalent CAR targeting FLT3 and/or CD33. The NOT gate protects healthy hematopoietic stem cells (HSCs) using an inhibitory CAR targeting endomucin, a protective antigen unique to healthy HSCs. NK cells with the combined OR-NOT gene circuit kill multiple AML subtypes and protect primary HSCs, and the circuit also works in vivo.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Animals
- Mice
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Gene Regulatory Networks
- Hematopoietic Stem Cells/metabolism
- Cell Line, Tumor
- Precision Medicine/methods
- Cell- and Tissue-Based Therapy/methods
Collapse
Affiliation(s)
| | - Han Deng
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Gozde Yucel
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Marcus Gainer
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Nelia Leemans
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Alice Lam
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Yongshuai Li
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Michelle Hung
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Derrick Lee
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Chen-Ting Lee
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Andrew Banicki
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Mengxi Tian
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | | | | | - Assen Roguev
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA
| | | | | | | | - Timothy K Lu
- Senti Biosciences, Inc., South San Francisco, CA 94080, USA; Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | |
Collapse
|
3
|
Zarou MM, Rattigan KM, Sarnello D, Shokry E, Dawson A, Ianniciello A, Dunn K, Copland M, Sumpton D, Vazquez A, Helgason GV. Inhibition of mitochondrial folate metabolism drives differentiation through mTORC1 mediated purine sensing. Nat Commun 2024; 15:1931. [PMID: 38431691 PMCID: PMC10908830 DOI: 10.1038/s41467-024-46114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
Supporting cell proliferation through nucleotide biosynthesis is an essential requirement for cancer cells. Hence, inhibition of folate-mediated one carbon (1C) metabolism, which is required for nucleotide synthesis, has been successfully exploited in anti-cancer therapy. Here, we reveal that mitochondrial folate metabolism is upregulated in patient-derived leukaemic stem cells (LSCs). We demonstrate that inhibition of mitochondrial 1C metabolism through impairment of de novo purine synthesis has a cytostatic effect on chronic myeloid leukaemia (CML) cells. Consequently, changes in purine nucleotide levels lead to activation of AMPK signalling and suppression of mTORC1 activity. Notably, suppression of mitochondrial 1C metabolism increases expression of erythroid differentiation markers. Moreover, we find that increased differentiation occurs independently of AMPK signalling and can be reversed through reconstitution of purine levels and reactivation of mTORC1. Of clinical relevance, we identify that combination of 1C metabolism inhibition with imatinib, a frontline treatment for CML patients, decreases the number of therapy-resistant CML LSCs in a patient-derived xenograft model. Our results highlight a role for folate metabolism and purine sensing in stem cell fate decisions and leukaemogenesis.
Collapse
Affiliation(s)
- Martha M Zarou
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Kevin M Rattigan
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Daniele Sarnello
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Engy Shokry
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Amy Dawson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Angela Ianniciello
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Karen Dunn
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Mhairi Copland
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - David Sumpton
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Alexei Vazquez
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| | - G Vignir Helgason
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
4
|
Sun R, Wang C, Wang Y, Wu Y, Du P, Sun X, Li Q, Bi K, Jiang G. Role of miR‑let‑7c‑5p/c‑myc signaling axis in the committed differentiation of leukemic THP‑1 cells into monocytes/macrophages. Oncol Lett 2023; 26:403. [PMID: 37600342 PMCID: PMC10433716 DOI: 10.3892/ol.2023.13989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
In a preliminary experiment, it was found that c-myc expression was decreased following the differentiation of THP-1 cells into monocytes/macrophages induced by phorbol 12-myristate 13 acetate (PMA) + lipopolysaccharide (LPS) + interferon (IFN)-γ. The expression of miR-let-7c-5p was then found to be elevated by cross-sectional analysis using TargetScan and PubMed and differential microarray analysis. The present study aimed to investigate the role of the miR-let-7c-5p/c-myc signaling axis in the committed differentiation of THP-1 leukemic cells into monocytes/macrophages induced by PMA + LPS + IFN-γ. Human THP-1 leukemic cells were induced to differentiate into monocytes/macrophages by PMA + LPS + IFN-γ. Following induction for 48 h, the growth density of the THP-1 cells was observed directly under an inverted microscope, cell proliferation was measured using Cell Counting Kit-8 assay and the cell cycle and the expression of differentiation-related antigens (CD11b and CD14) were measured using flow cytometry. The mRNA expression of miR-let-7c-5p and c-myc was detected using reverse transcription-quantitative PCR and the protein expression of c-myc was detected using western blot analysis. Dual luciferase reporter gene analysis was used to detect the targeted binding of miR-let-7c-5p on the 3'UTR of c-myc. The relative expression of miR-let-7c-5p and c-myc genes in THP-1 cells induced by PMA + LPS + IFN-γ was found to be up- and downregulated respectively, and expression of miR-let-7c-5p was negatively correlated with the expression of c-myc gene. Dual luciferase reporter gene assays confirmed that miR-let-7c-5p targeted the 3'UTR of c-myc and inhibited luciferase activity. Following transfection with miR-let-7c-5p mimics, the expression of c-myc was markedly downregulated and the proliferative ability of the THP-1 cells was decreased, while the expression rate of CD11b and CD14 was significantly increased. The rescue experiment revealed that the effects of miR-let-7c-5p mimics on the proliferation and differentiation of THP-1 cells were attenuated by transfection with c-myc overexpression vector. Together, the findings of the present study demonstrated that miR-let-7c-5p can target the 3'UTR region of c-myc and that the miR-let-7c-5p/c-myc signaling axis is one of the critical pathways involved in the directional differentiation of leukemic cells into monocytes/macrophages.
Collapse
Affiliation(s)
- Ruijing Sun
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Chaozhe Wang
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yufang Wang
- Department of Laboratory Medicine, Fushan District People's Hospital, Yantai, Shandong 265500, P.R. China
| | - Yunhua Wu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pengchao Du
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaolin Sun
- Department of Laboratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Qing Li
- Department of Laboratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Kehong Bi
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250062, P.R. China
| | - Guosheng Jiang
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Department of Precision Molecular Laboratory Medicine, Zhangqiu District People's Hospital of Jinan Affiliated to Jining Medical University, Jinan, Shandong 250200, P.R. China
| |
Collapse
|
5
|
Park J, Luo Y, Park JW, Kim SH, Hong YJ, Lim Y, Seo YJ, Bae J, Seo SB. Downregulation of DNA methylation enhances differentiation of THP-1 cells and induces M1 polarization of differentiated macrophages. Sci Rep 2023; 13:13132. [PMID: 37573395 PMCID: PMC10423279 DOI: 10.1038/s41598-023-40362-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023] Open
Abstract
DNA methylation is an epigenetic modification that regulates gene expression and plays an essential role in hematopoiesis. UHRF1 and DNMT1 are both crucial for regulating genome-wide maintenance of DNA methylation. Specifically, it is well known that hypermethylation is crucial characteristic of acute myeloid leukemia (AML). However, the mechanism underlying how DNA methylation regulates the differentiation of AML cells, including THP-1 is not fully elucidated. In this study, we report that UHRF1 or DNMT1 depletion enhances the phorbol-12-myristate-13-acetate (PMA)-induced differentiation of THP-1 cells. Transcriptome analysis and genome-wide methylation array results showed that depleting UHRF1 or DNMT1 induced changes that made THP-1 cells highly sensitive to PMA. Furthermore, knockdown of UHRF1 or DNMT1 impeded solid tumor formation in xenograft mouse model. These findings suggest that UHRF1 and DNMT1 play a pivotal role in regulating differentiation and proliferation of THP-1 cells and targeting these proteins may improve the efficiency of differentiation therapy in AML patients.
Collapse
Affiliation(s)
- Junyoung Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yongyang Luo
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jin Woo Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Song Hyun Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ye Joo Hong
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Younghyun Lim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Young-Jin Seo
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jeehyeon Bae
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sang Beom Seo
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
6
|
Abokyi S, Ghartey-Kwansah G, Tse DYY. TFEB is a central regulator of the aging process and age-related diseases. Ageing Res Rev 2023; 89:101985. [PMID: 37321382 DOI: 10.1016/j.arr.2023.101985] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/25/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
Old age is associated with a greater burden of disease, including neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease, as well as other chronic diseases. Coincidentally, popular lifestyle interventions, such as caloric restriction, intermittent fasting, and regular exercise, in addition to pharmacological interventions intended to protect against age-related diseases, induce transcription factor EB (TFEB) and autophagy. In this review, we summarize emerging discoveries that point to TFEB activity affecting the hallmarks of aging, including inhibiting DNA damage and epigenetic modifications, inducing autophagy and cell clearance to promote proteostasis, regulating mitochondrial quality control, linking nutrient-sensing to energy metabolism, regulating pro- and anti-inflammatory pathways, inhibiting senescence and promoting cell regenerative capacity. Furthermore, the therapeutic impact of TFEB activation on normal aging and tissue-specific disease development is assessed in the contexts of neurodegeneration and neuroplasticity, stem cell differentiation, immune responses, muscle energy adaptation, adipose tissue browning, hepatic functions, bone remodeling, and cancer. Safe and effective strategies of activating TFEB hold promise as a therapeutic strategy for multiple age-associated diseases and for extending lifespan.
Collapse
Affiliation(s)
- Samuel Abokyi
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China; Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China.
| | - George Ghartey-Kwansah
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Dennis Yan-Yin Tse
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China; Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR of China; Centre for Eye and Vision Research, 17W Hong Kong Science Park, Hong Kong SAR of China.
| |
Collapse
|
7
|
Kaiser AM, Gatto A, Hanson KJ, Zhao RL, Raj N, Ozawa MG, Seoane JA, Bieging-Rolett KT, Wang M, Li I, Trope WL, Liou DZ, Shrager JB, Plevritis SK, Newman AM, Van Rechem C, Attardi LD. p53 governs an AT1 differentiation programme in lung cancer suppression. Nature 2023; 619:851-859. [PMID: 37468633 PMCID: PMC11288504 DOI: 10.1038/s41586-023-06253-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 05/24/2023] [Indexed: 07/21/2023]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide1. Mutations in the tumour suppressor gene TP53 occur in 50% of lung adenocarcinomas (LUADs) and are linked to poor prognosis1-4, but how p53 suppresses LUAD development remains enigmatic. We show here that p53 suppresses LUAD by governing cell state, specifically by promoting alveolar type 1 (AT1) differentiation. Using mice that express oncogenic Kras and null, wild-type or hypermorphic Trp53 alleles in alveolar type 2 (AT2) cells, we observed graded effects of p53 on LUAD initiation and progression. RNA sequencing and ATAC sequencing of LUAD cells uncovered a p53-induced AT1 differentiation programme during tumour suppression in vivo through direct DNA binding, chromatin remodelling and induction of genes characteristic of AT1 cells. Single-cell transcriptomics analyses revealed that during LUAD evolution, p53 promotes AT1 differentiation through action in a transitional cell state analogous to a transient intermediary seen during AT2-to-AT1 cell differentiation in alveolar injury repair. Notably, p53 inactivation results in the inappropriate persistence of these transitional cancer cells accompanied by upregulated growth signalling and divergence from lung lineage identity, characteristics associated with LUAD progression. Analysis of Trp53 wild-type and Trp53-null mice showed that p53 also directs alveolar regeneration after injury by regulating AT2 cell self-renewal and promoting transitional cell differentiation into AT1 cells. Collectively, these findings illuminate mechanisms of p53-mediated LUAD suppression, in which p53 governs alveolar differentiation, and suggest that tumour suppression reflects a fundamental role of p53 in orchestrating tissue repair after injury.
Collapse
Affiliation(s)
- Alyssa M Kaiser
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alberto Gatto
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathryn J Hanson
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard L Zhao
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nitin Raj
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael G Ozawa
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - José A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Kathryn T Bieging-Rolett
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mengxiong Wang
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Irene Li
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Winston L Trope
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Douglas Z Liou
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph B Shrager
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Capucine Van Rechem
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
8
|
Cheng X, Liang H, Li Q, Wang J, Liu J, Zhang Y, Ru Y, Zhou Y. Raman spectroscopy differ leukemic cells from their healthy counterparts and screen biomarkers in acute leukemia. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 281:121558. [PMID: 35843058 DOI: 10.1016/j.saa.2022.121558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Precision medicine is important in the treatment of acute leukemia (AL). The target therapies of AL provide an opportunity to reduce the mortality of AL. How AL cells differ from their healthy counterparts is the basis for the development of therapies and the outcome of AL patients. Therefore, a label-free and noninvasive single-cell Raman platform was used to characterize cell molecular profiles and found potential biomarkers from three healthy people and twelve AL patients with more than 90% accuracy. We analyzed myeloblasts, abnormal promyelocytes, monoblasts and B-ALL cells respectively, compared with their healthy counterparts, which could be distinguished by their intrinsic phenotypic Raman spectra using orthogonal partial least squares discriminate analysis (OPLS-DA). Most importantly, we selected statistically significant markers of the four leukemia models. Further analysis of leukemic granulocytes, we found that a combination of the 1003, 1341 and 1579 cm-1 Raman peaks could discriminate myeloblasts and abnormal promyelocytes from normal granulocytes. The assignments of 1579 cm-1 gave us a clue to find potential important variables myeloperoxidase related with AL diagnosis. Our study demonstrates the capability of the Raman platform to characterize leukemia cells with non-invasively probing metabolites. The biomarker we identified could be extensible to other blood cells and potentially have a high impact on leukemia therapy.
Collapse
Affiliation(s)
- Xuelian Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Haoyue Liang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Qing Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Jing Wang
- Nankai University, National Demonstration Center for Experimental Chemistry Education, Tianjin 300071, China
| | - Jing Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yun Zhang
- Department of Clinical Laboratory, The District People's Hospital of Zhangqiu, Jinan 250000, China
| | - Yongxin Ru
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China.
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China.
| |
Collapse
|
9
|
Genome-Wide Gene Expression Profiling Defines the Mechanism of Anticancer Effect of Colorectal Cancer Cell-Derived Conditioned Medium on Acute Myeloid Leukemia. Genes (Basel) 2022; 13:genes13050883. [PMID: 35627268 PMCID: PMC9171579 DOI: 10.3390/genes13050883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common type of leukemia in adults, accounting for 30% of all adult leukemia cases. While there have been recent improvements in the prognosis of the disease, the prognosis remains grim, and further understanding of AML and the development of new therapeutic agents is critical. This study aimed to investigate the potential interaction between colorectal cancer (CRC) cells and AML cells. Unexpectedly, we found that CRC cell-derived conditioned medium (CM) showed anticancer activities in AML cells by inducing apoptosis and differentiation. Mechanistic studies suggest that these phenotypes are closely associated with the suppression of PI3K/AKT/mTOR and MAPK survival signaling, the upregulation of myeloid differentiation-promoting transcription factors c/EBPα and PU.1, and the augmentation of executioner caspases-3/7. Importantly, bioinformatic analyses of our gene expression profiling data, including that derived from principal component analysis (PCA), volcano plots, boxplots, heat maps, kyoto encyclopedia of genes and genomes (KEGG) pathways, and receiver operating characteristic (ROC) curves, which evaluate gene expression profiling data, provided deeper insight into the mechanism in which CRC-CM broadly modulates apoptosis-, cell cycle arrest-, and differentiation-related gene expression, such as BMF, PLSCR3, CDKN1C, and ID2, among others, revealing the genes that exert anticancer effects in AML cells at the genomic level. Collectively, our data suggest that it may be worthwhile to isolate and identify the molecules with tumor-suppressive effects in the CM, which may help to improve the prognosis of patients with AML.
Collapse
|
10
|
Josa-Culleré L, Madden KS, Cogswell TJ, Jackson TR, Carter TS, Zhang D, Trevitt G, Davies SG, Vyas P, Wynne GM, Milne TA, Russell AJ. A Phenotypic Screen Identifies a Compound Series That Induces Differentiation of Acute Myeloid Leukemia Cells In Vitro and Shows Antitumor Effects In Vivo. J Med Chem 2021; 64:15608-15628. [PMID: 34672555 DOI: 10.1021/acs.jmedchem.1c00574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Induction of differentiation is a promising therapeutic strategy against acute myeloid leukemia. However, current differentiation therapies are effective only to specific patient populations. To identify novel differentiation agents with wider efficacy, we developed a phenotypic high-throughput screen with a range of genetically diverse cell lines. From the resulting hits, one chemical scaffold was optimized in terms of activity and physicochemical properties to yield OXS007417, a proof-of-concept tool compound, which was also able to decrease tumor volume in a murine in vivo xenograft model.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Differentiation/drug effects
- Cell Survival/drug effects
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Female
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Structure
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Phenotype
- Structure-Activity Relationship
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Laia Josa-Culleré
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Katrina S Madden
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Thomas J Cogswell
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Thomas R Jackson
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Tom S Carter
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Douzi Zhang
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Graham Trevitt
- XenoGesis Ltd., BioCity Nottingham, Pennyfoot Street, Nottingham NG1 1GF, Nottinghamshire, U.K
| | - Stephen G Davies
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Graham M Wynne
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Thomas A Milne
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| |
Collapse
|
11
|
Sabatier M, Boet E, Zaghdoudi S, Guiraud N, Hucteau A, Polley N, Cognet G, Saland E, Lauture L, Farge T, Sahal A, Pancaldi V, Chu-Van E, Castelli F, Bertoli S, Bories P, Récher C, Boutzen H, Mansat-De Mas V, Stuani L, Sarry JE. Activation of Vitamin D Receptor Pathway Enhances Differentiating Capacity in Acute Myeloid Leukemia with Isocitrate Dehydrogenase Mutations. Cancers (Basel) 2021; 13:5243. [PMID: 34680392 PMCID: PMC8533831 DOI: 10.3390/cancers13205243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/30/2022] Open
Abstract
Relapses and resistance to therapeutic agents are major barriers in the treatment of acute myeloid leukemia (AML) patients. These unfavorable outcomes emphasize the need for new strategies targeting drug-resistant cells. As IDH mutations are present in the preleukemic stem cells and systematically conserved at relapse, targeting IDH mutant cells could be essential to achieve a long-term remission in the IDH mutant AML subgroup. Here, using a panel of human AML cell lines and primary AML patient specimens harboring IDH mutations, we showed that the production of an oncometabolite (R)-2-HG by IDH mutant enzymes induces vitamin D receptor-related transcriptional changes, priming these AML cells to differentiate with pharmacological doses of ATRA and/or VD. This activation occurs in a CEBPα-dependent manner. Accordingly, our findings illuminate potent and cooperative effects of IDH mutations and the vitamin D receptor pathway on differentiation in AML, revealing a novel therapeutic approach easily transferable/immediately applicable to this subgroup of AML patients.
Collapse
Affiliation(s)
- Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Sonia Zaghdoudi
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Nathan Guiraud
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Alexis Hucteau
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Nathaniel Polley
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Guillaume Cognet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Laura Lauture
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Vera Pancaldi
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
| | - Emeline Chu-Van
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, 91191 Gif-sur-Yvette, France; (E.C.-V.); (F.C.)
| | - Florence Castelli
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, 91191 Gif-sur-Yvette, France; (E.C.-V.); (F.C.)
| | - Sarah Bertoli
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
- Département d’Hématologie, University of Toulouse, CEDEX 6, 31013 Toulouse, France
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, CEDEX 9, 31059 Toulouse, France
| | - Pierre Bories
- Réseau Régional de Cancérologie Onco-Occitanie, CEDEX 9, 31059 Toulouse, France;
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
- Département d’Hématologie, University of Toulouse, CEDEX 6, 31013 Toulouse, France
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, CEDEX 9, 31059 Toulouse, France
| | - Héléna Boutzen
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Véronique Mansat-De Mas
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
- Département d’Hématologie, University of Toulouse, CEDEX 6, 31013 Toulouse, France
| | - Lucille Stuani
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| |
Collapse
|
12
|
Diab D, Pinon A, Ouk C, Hage-Sleiman R, Diab-Assaf M, Liagre B, Leger DY. Involvement of autophagy in diosgenin‑induced megakaryocyte differentiation in human erythroleukemia cells. Mol Med Rep 2021; 24:746. [PMID: 34458927 PMCID: PMC8436216 DOI: 10.3892/mmr.2021.12386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/21/2021] [Indexed: 11/06/2022] Open
Abstract
Natural agents have been used to restart the process of differentiation that is inhibited during leukemic transformation of hematopoietic stem or progenitor cells. Autophagy is a housekeeping pathway that maintains cell homeostasis against stress by recycling macromolecules and organelles and plays an important role in cell differentiation. In the present study, an experimental model was established to investigate the involvement of autophagy in the megakaryocyte differentiation of human erythroleukemia (HEL) cells induced by diosgenin [also known as (25R)‑Spirosten‑5‑en‑3b‑ol]. It was demonstrated that Atg7 expression was upregulated from day 1 of diosgenin‑induced differentiation and was accompanied by a significant elevation in the conversion of light chain 3 A/B (LC3‑A/B)‑I to LC3‑A/B‑II. Autophagy was modulated before or after the induction of megakaryocyte differentiation using 3‑methyladenine (3‑MA, autophagy inhibitor) and metformin (Met, autophagy initiation activator). 3‑MA induced a significant accumulation of the LC3 A/B‑II form at day 8 of differentiation. It was revealed that 3‑MA had a significant repressive effect on the nuclear (polyploidization) and membrane glycoprotein V [(GpV) expression] maturation. On the other hand, autophagy activation increased GpV genomic expression, but did not change the nuclear maturation profile after HEL cells treatment with Met. It was concluded that autophagy inhibition had a more prominent effect on the diosgenin‑differentiated cells than autophagy activation.
Collapse
Affiliation(s)
- Dima Diab
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| | - Aline Pinon
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| | - Catherine Ouk
- BISCEm Flow Cytometry/Microscopy Unit, University of Limoges, 87025 Limoges, France
| | - Rouba Hage-Sleiman
- Department of Biology, Faculty of Sciences, Lebanese University, Hadath El Jebbeh, Beyrouth 21219, Lebanon
| | - Mona Diab-Assaf
- Doctoral School of Sciences and Technology, Lebanese University, Hadath El Jebbeh, Beyrouth 21219, Lebanon
| | - Bertrand Liagre
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| | - David Yannick Leger
- PEIRENE Laboratory EA 7500, Faculty of Pharmacy, University of Limoges, 87025 Limoges, France
| |
Collapse
|
13
|
Hunsu VO, Facey COB, Fields JZ, Boman BM. Retinoids as Chemo-Preventive and Molecular-Targeted Anti-Cancer Therapies. Int J Mol Sci 2021; 22:7731. [PMID: 34299349 PMCID: PMC8304138 DOI: 10.3390/ijms22147731] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Retinoic acid (RA) agents possess anti-tumor activity through their ability to induce cellular differentiation. However, retinoids have not yet been translated into effective systemic treatments for most solid tumors. RA signaling is mediated by the following two nuclear retinoic receptor subtypes: the retinoic acid receptor (RAR) and the retinoic X receptor (RXR), and their isoforms. The identification of mutations in retinoid receptors and other RA signaling pathway genes in human cancers offers opportunities for target discovery, drug design, and personalized medicine for distinct molecular retinoid subtypes. For example, chromosomal translocation involving RARA occurs in acute promyelocytic leukemia (APL), and all-trans retinoic acid (ATRA) is a highly effective and even curative therapeutic for APL patients. Thus, retinoid-based target discovery presents an important line of attack toward designing new, more effective strategies for treating other cancer types. Here, we review retinoid signaling, provide an update on retinoid agents and the current clinical research on retinoids in cancer, and discuss how the retinoid pathway genotype affects the ability of retinoid agents to inhibit the growth of colorectal cancer (CRC) cells. We also deliberate on why retinoid agents have not shown clinical efficacy against solid tumors and discuss alternative strategies that could overcome the lack of efficacy.
Collapse
Affiliation(s)
- Victoria O. Hunsu
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (V.O.H.); (C.O.B.F.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (V.O.H.); (C.O.B.F.)
| | | | - Bruce M. Boman
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (V.O.H.); (C.O.B.F.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
14
|
Zhao M, Wang J, Qu M, Zhao Y, Wang H, Ke Y, Liu Y, Lei ZN, Liu HM, Hu Z, Wei L, Chen ZS. OGP46 Induces Differentiation of Acute Myeloid Leukemia Cells via Different Optimal Signaling Pathways. Front Cell Dev Biol 2021; 9:652972. [PMID: 33748146 PMCID: PMC7969801 DOI: 10.3389/fcell.2021.652972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/09/2021] [Indexed: 11/13/2022] Open
Abstract
Acute myelogenous leukemia (AML) is characterized by blockage of cell differentiation leading to the accumulation of immature cells, which is the most prevalent form of acute leukemia in adults. It is well known that all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) are the preferred drugs for acute promyelocytic leukemia (APL). However, they can lead to irreversible resistance which may be responsible for clinical failure after complete remission (CR). Moreover, the differentiation therapy of ATRA-based treatment has not been effective against AML with t(8;21) translocation. Here we aimed to identify the differentiation effect of OGP46 on AML cell lines (HL-60, NB4, and Kasumi-1) and explore its possible mechanisms. We found that OGP46 has significant inhibitory activity against these cells by triggering cell differentiation with cell-cycle exit at G1/G0 and inhibited the colony-formation capacity of the AML cells. It was shown that OGP46 induced the differentiation of NB4 cells via the transcriptional misregulation in cancer signaling pathway by PML-RARα depletion, while it was attributed to the hematopoietic cell lineage and phagosome pathway in Kasumi-1 cells, which are all critical pathways in cell differentiation. These results highlight that OGP46 is an active agent not only in the APL cell line NB4 but also in AML-M2 cell lines, especially Kasumi-1 with t(8;21) translocation. Therefore, OGP46 may be a potential compound for surmounting the differentiation blockage in AML.
Collapse
Affiliation(s)
- Min Zhao
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China.,School of Pharmacy, Weifang Medical University, Weifang, China
| | - Jiangyun Wang
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China.,School of Pharmacy, Weifang Medical University, Weifang, China
| | - Mei Qu
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China.,School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yao Zhao
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Haihua Wang
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yu Ke
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Ying Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States.,School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Hong-Min Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zhenbo Hu
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Liuya Wei
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China.,School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, United States
| |
Collapse
|
15
|
Zuo S, Sun L, Wang Y, Chen B, Wang J, Ge X, Lu Y, Yang N, Shen P. Establishment of a novel mesenchymal stem cell-based regimen for chronic myeloid leukemia differentiation therapy. Cell Death Dis 2021; 12:208. [PMID: 33627636 PMCID: PMC7904926 DOI: 10.1038/s41419-021-03499-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/25/2022]
Abstract
Chronic myeloid leukemia (CML) is characterized by the accumulation of malignant and immature white blood cells which spread to the peripheral blood and other tissues/organs. Despite the fact that current tyrosine kinase inhibitors (TKIs) are capable of achieving the complete remission by reducing the tumor burden, severe adverse effects often occur in CML patients treated with TKIs. The differentiation therapy exhibits therapeutic potential to improve cure rates in leukemia, as evidenced by the striking success of all-trans-retinoic acid in acute promyelocytic leukemia treatment. However, there is still a lack of efficient differentiation therapy strategy in CML. Here we showed that MPL, which encodes the thrombopoietin receptor driving the development of hematopoietic stem/progenitor cells, decreased along with the progression of CML. We first elucidated that MPL signaling blockade impeded the megakaryocytic differentiation and contributed to the progression of CML. While allogeneic human umbilical cord-derived mesenchymal stem cells (UC-MSCs) treatment efficiently promoted megakaryocytic lineage differentiation of CML cells through restoring the MPL expression and activating MPL signaling. UC-MSCs in combination with eltrombopag, a non-peptide MPL agonist, further activated JAK/STAT and MAPK signaling pathways through MPL and exerted a synergetic effect on enhancing CML cell differentiation. The established combinational treatment not only markedly reduced the CML burden but also significantly eliminated CML cells in a xenograft CML model. We provided a new molecular insight of thrombopoietin (TPO) and MPL signaling in MSCs-mediated megakaryocytic differentiation of CML cells. Furthermore, a novel anti-CML treatment regimen that uses the combination of UC-MSCs and eltrombopag shows therapeutic potential to overcome the differentiation blockade in CML.
Collapse
MESH Headings
- Animals
- Benzoates/pharmacology
- Cell Lineage
- Coculture Techniques
- Gene Expression Regulation, Leukemic
- Humans
- Hydrazines/pharmacology
- Janus Kinases/metabolism
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/surgery
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/metabolism
- Mice, Nude
- Mitogen-Activated Protein Kinases/metabolism
- Pyrazoles/pharmacology
- Receptors, Thrombopoietin/agonists
- Receptors, Thrombopoietin/metabolism
- STAT Transcription Factors/metabolism
- Signal Transduction
- Thrombopoiesis/drug effects
- Umbilical Cord/cytology
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Shiman Zuo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China
| | - Luchen Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China
| | - Yuxin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jingyue Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China
| | - Xiangyu Ge
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yan Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China.
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of life science, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
16
|
Zhao X, Liu HQ, Wang LN, Yang L, Liu XL. Current and emerging molecular and epigenetic disease entities in acute myeloid leukemia and a critical assessment of their therapeutic modalities. Semin Cancer Biol 2020; 83:121-135. [PMID: 33242577 DOI: 10.1016/j.semcancer.2020.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Acute myeloid leukemia (AML) is the most frequently diagnosed acute leukemia, and its incidence increases with age. Although the etiology of AML remains unknown, exposure to genotoxic agents or some prior hematologic disorders could lead to the development of this condition. The pathogenesis of AML involves the development of malignant transformation of hematopoietic stem cells that undergo successive genomic alterations, ultimately giving rise to a full-blown disease. From the disease biology perspective, AML is considered to be extremely complex with significant genetic, epigenetic, and phenotypic variations. Molecular and cytogenetic alterations in AML include mutations in those subsets of genes that are involved in normal cell proliferation, maturation and survival, thus posing significant challenge to targeting these pathways without attendant toxicity. In addition, multiple malignant cells co-exist in the majority of AML patients. Individual subclones are characterized by unique genetic and epigenetic abnormalities, which contribute to the differences in their response to treatment. As a result, despite a dramatic progress in our understanding of the pathobiology of AML, not much has changed in therapeutic approaches to treat AML in the past four decades. Dose and regimen modifications with improved supportive care have contributed to improved outcomes by reducing toxicity-related side effects. Several drug candidates are currently being developed, including targeted small-molecule inhibitors, cytotoxic chemotherapies, monoclonal antibodies and epigenetic drugs. This review summarizes the current state of affairs in the pathobiological and therapeutic aspects of AML.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, China
| | - Huan-Qiu Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Li-Na Wang
- Department of Paediatrics, The First Hospital of Jilin University, Changchun, China
| | - Le Yang
- Department of Endocrinology, The People's Hospital of Jilin Province, Changchun, China.
| | - Xiao-Liang Liu
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
17
|
Nair R, Salinas-Illarena A, Baldauf HM. New strategies to treat AML: novel insights into AML survival pathways and combination therapies. Leukemia 2020; 35:299-311. [PMID: 33122849 DOI: 10.1038/s41375-020-01069-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022]
Abstract
The effective treatment of acute myeloid leukemia (AML) is very challenging. Due to the immense heterogeneity of this disease, treating it using a "one size fits all" approach is ineffective and only benefits a subset of patients. Instead, there is a shift towards more personalized treatment based on the patients' genomic signature. This shift has facilitated the increased revelation of novel insights into pathways that lead to the survival and propagation of AML cells. These AML survival pathways are involved in drug resistance, evasion of the immune system, reprogramming metabolism, and impairing differentiation. In addition, based on the reports of enhanced clinical efficiencies when combining drugs or treatments, deeper investigation into possible pathways, which can be targeted together to increase treatment response in a wider group of patients, is warranted. In this review, not only is a comprehensive summary of targets involved in these pathways provided, but also insights into the potential of targeting these molecules in combination therapy will be discussed.
Collapse
Affiliation(s)
- Ramya Nair
- Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Alejandro Salinas-Illarena
- Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
| | - Hanna-Mari Baldauf
- Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.
| |
Collapse
|
18
|
Opydo-Chanek M, Cichoń I, Rak A, Kołaczkowska E, Mazur L. The pan-Bcl-2 inhibitor obatoclax promotes differentiation and apoptosis of acute myeloid leukemia cells. Invest New Drugs 2020; 38:1664-1676. [PMID: 32367199 PMCID: PMC7575496 DOI: 10.1007/s10637-020-00931-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022]
Abstract
One of the key features of acute myeloid leukemia (AML) is the arrest of differentiation at the early progenitor stage of myelopoiesis. Therefore, the identification of new agents that could overcome this differentiation block and force leukemic cells to enter the apoptotic pathway is essential for the development of new treatment strategies in AML. Regarding this, herein we report the pro-differentiation activity of the pan-Bcl-2 inhibitor, obatoclax. Obatoclax promoted differentiation of human AML HL-60 cells and triggered their apoptosis in a dose- and time-dependent manner. Importantly, obatoclax-induced apoptosis was associated with leukemic cell differentiation. Moreover, decreased expression of Bcl-2 protein was observed in obatoclax-treated HL-60 cells. Furthermore, differentiation of these cells was accompanied by the loss of their proliferative capacity, as shown by G0/G1 cell cycle arrest. Taken together, these findings indicate that the anti-AML effects of obatoclax involve not only the induction of apoptosis but also differentiation of leukemic cells. Therefore, obatoclax represents a promising treatment for AML that warrants further exploration.
Collapse
Affiliation(s)
- Małgorzata Opydo-Chanek
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| | - Iwona Cichoń
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Agnieszka Rak
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Elżbieta Kołaczkowska
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Lidia Mazur
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| |
Collapse
|
19
|
Bolis M, Paroni G, Fratelli M, Vallerga A, Guarrera L, Zanetti A, Kurosaki M, Garattini SK, Gianni’ M, Lupi M, Pattini L, Barzago MM, Terao M, Garattini E. All-Trans Retinoic Acid Stimulates Viral Mimicry, Interferon Responses and Antigen Presentation in Breast-Cancer Cells. Cancers (Basel) 2020; 12:cancers12051169. [PMID: 32384653 PMCID: PMC7281473 DOI: 10.3390/cancers12051169] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 05/02/2020] [Indexed: 12/16/2022] Open
Abstract
All-trans retinoic acid (ATRA), a recognized differentiating agent, has significant potential in the personalized/stratified treatment of breast cancer. The present study reports on the molecular mechanisms underlying the anti-tumor activity of ATRA in breast cancer. The work is based on transcriptomic experiments performed on ATRA-treated breast cancer cell-lines, short-term tissue cultures of patient-derived mammary-tumors and a xenograft model. ATRA upregulates gene networks involved in interferon-responses, immune-modulation and antigen-presentation in retinoid-sensitive cells and tumors characterized by poor immunogenicity. ATRA-dependent upregulation of these gene networks is caused by a viral mimicry process, involving the activation of endogenous retroviruses. ATRA induces a non-canonical type of viral mimicry, which results in increased expression of the IRF1 (Interferon Responsive Factor 1) transcription factor and the DTX3L (Deltex-E3-Ubiquitin-Ligase-3L) downstream effector. Functional knockdown studies indicate that IRF1 and DTX3L are part of a negative feedback loop controlling ATRA-dependent growth inhibition of breast cancer cells. The study is of relevance from a clinical/therapeutic perspective. In fact, ATRA stimulates processes controlling the sensitivity to immuno-modulatory drugs, such as immune-checkpoint-inhibitors. This suggests that ATRA and immunotherapeutic agents represent rational combinations for the personalized treatment of breast cancer. Remarkably, ATRA-sensitivity seems to be relatively high in immune-cold mammary tumors, which are generally resistant to immunotherapy.
Collapse
Affiliation(s)
- Marco Bolis
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
- Functional Cancer Genomics Laboratory, Institute of Oncology Research, USI, University of Southern Switzerland, 6500 Bellinzona, Switzerland
- Bioinformatics Core Unit Institute of Oncology Research, Swiss Institute of Bioinformatics, 1000 Lausanne, Switzerland
| | - Gabriela Paroni
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Maddalena Fratelli
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Arianna Vallerga
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Luca Guarrera
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Adriana Zanetti
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Mami Kurosaki
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Silvio Ken Garattini
- Department of Oncology, Azienda Ospedaliera di Udine, DAME, Dipartimento di Area Medica Università degli Studi di Udine, 33100 Udine, Italy;
| | - Maurizio Gianni’
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Monica Lupi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via La Masa 19, 20156 Milano, Italy;
| | - Linda Pattini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20156 Milano, Italy;
| | - Maria Monica Barzago
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Mineko Terao
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156 Milano, Italy; (M.B.); (G.P.); (M.F.); (A.V.); (L.G.); (A.Z.); (M.K.); (M.G.); (M.M.B.); (M.T.)
- Correspondence: ; Tel.: +39-02-3901-4533
| |
Collapse
|
20
|
Sami SA, Darwish NHE, Barile ANM, Mousa SA. Current and Future Molecular Targets for Acute Myeloid Leukemia Therapy. Curr Treat Options Oncol 2020; 21:3. [PMID: 31933183 DOI: 10.1007/s11864-019-0694-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OPINION STATEMENT Acute myeloid leukemia (AML) disease prognosis is poor and there is a high risk of chemo-resistant relapse for both young and old patients. Thus, there is a demand for alternative and target-specific drugs to improve the 5-year survival rate. Current treatment mainstays include chemotherapy, or mutation-specific targeting molecules including FLT3 inhibitors, IDH inhibitors, and monoclonal antibodies. Efforts to devise new, targeted therapy have included recent advances in methods for high-throughput genomic screening and the availability of computer-assisted techniques for the design of novel agents predicted to specifically inhibit mutant molecules involved in leukemogenesis. Crosstalk between the leukemia cells and the bone marrow microenvironment through cell surface molecules, such as the integrins αvβ3 and αvβ5, might influence drug response and AML progression. This review article focuses on current AML treatment options, new AML targeted therapies, the role of integrins in AML progression, and a potential therapeutic agent-integrin αvβ3 antagonist.
Collapse
Affiliation(s)
- Shaheedul A Sami
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA
| | - Noureldien H E Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA.,Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amanda N M Barile
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA.
| |
Collapse
|
21
|
Ilnicka A, Gocek E, Łopatecka J, Marcinkowska E. Regulation of FOXP3 expression in myeloid cells in response to all-trans-retinoic acid, interleukin 2 and transforming growth factor β. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 96:18-26. [PMID: 30826380 DOI: 10.1016/j.dci.2019.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 06/09/2023]
Abstract
FoxP3 is a transcription factor essential for differentiation and function of T regulatory cells (Tregs). There are two major subsets of Tregs: natural Tregs (nTregs) generated in thymus and inducible Tregs (iTregs) produced in peripheral immune system. It has been documented that iTreg development is dependent on soluble mediators including interleukin 2 (IL2), transforming growth factor β (TGFβ) and all-trans-retinoic acid (ATRA). In our experiments we performed a gene expression array, followed by Real-time PCR experiments to study expression of genes regulated by ATRA in cells of myeloid origin. Our experiments revealed that ATRA alone, but also a cocktail of mediators consisting of IL2, TGFβ and ATRA, upregulate expression of FOXP3 gene in normal and leukemic myeloid cells. Our results indicate that signaling pathways which are used at the late steps of T cell differentiation, are also active in the cells of myeloid lineage.
Collapse
Affiliation(s)
- Aleksandra Ilnicka
- Department of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Elżbieta Gocek
- Department of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Justyna Łopatecka
- Department of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Ewa Marcinkowska
- Department of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
22
|
Torres J, Monti R, Moore AL, Seimiya M, Jiang Y, Beerenwinkel N, Beisel C, Beira JV, Paro R. A switch in transcription and cell fate governs the onset of an epigenetically-deregulated tumor in Drosophila. eLife 2018; 7:32697. [PMID: 29560857 PMCID: PMC5862528 DOI: 10.7554/elife.32697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/04/2018] [Indexed: 12/20/2022] Open
Abstract
Tumor initiation is often linked to a loss of cellular identity. Transcriptional programs determining cellular identity are preserved by epigenetically-acting chromatin factors. Although such regulators are among the most frequently mutated genes in cancer, it is not well understood how an abnormal epigenetic condition contributes to tumor onset. In this work, we investigated the gene signature of tumors caused by disruption of the Drosophila epigenetic regulator, polyhomeotic (ph). In larval tissue ph mutant cells show a shift towards an embryonic-like signature. Using loss- and gain-of-function experiments we uncovered the embryonic transcription factor knirps (kni) as a new oncogene. The oncogenic potential of kni lies in its ability to activate JAK/STAT signaling and block differentiation. Conversely, tumor growth in ph mutant cells can be substantially reduced by overexpressing a differentiation factor. This demonstrates that epigenetically derailed tumor conditions can be reversed when targeting key players in the transcriptional network. When an animal is developing as an embryo, different cells start to specialize into the specific cell types needed to form the tissues and organs of the body. How an individual cell commits to become a certain type of cell is mostly determined by which of the genes in its DNA are active. In animal cells, DNA is wrapped around proteins called histones, and one way that cells can maintain their distinct pattern of gene activity is via chemical tags on the histones. These tags can switch nearby genes on or off, and are added or removed by other proteins called epigenetic regulators. The epigenetic tags are also stably inherited when the cell divides, meaning that a cell’s identity can be maintained over many cell generations. If epigenetic regulators fail to work properly or get disrupted, the pattern of gene activity in a cell becomes altered. As a consequence, that cell can lose its identity and will often turn into a cancer cell. In fact, mutations in epigenetic regulators are found in several human cancers. It is not yet understood how these changes in gene expression lead cells to become cancerous. Torres et al. have now analyzed an epigenetic regulator called Polyhomeotic in developing larvae of the fruit fly, Drosophila melanogaster. The results show that when Polyhomeotic is not produced the fly larvae develop tumors. Moreover, the mutant cells without Polyhomeotic had different gene expression profiles compared to normal cells. This in turn caused the mutant cells, which had previously committed to a certain fate, to become more like the unspecialized cells found in early embryos. Torres et al. next showed that, among the genes that were incorrectly regulated when Polyhomeotic’s activity was compromised, one gene called knirps was switched on by mistake, which led the mutant cells to become tumor cells. When the activity of knirps was reduced instead, almost no tumors grew. Additionally, Torres et al. found that the protein encoded by knirps activates a signaling pathway that keeps tumor cells unspecialized by blocking their normal progress to a more mature and specialized state – a process known as differentiation. Experimentally raising the levels of a different molecule that ultimately promotes differentiation caused the tumor cells to grow less. These findings suggest that tumors caused when epigenetic regulation goes awry may be reversed by targeting key genes such as knirps. Further work is now needed to test whether these findings will also extend to humans. Forcing cancer cells from a highly dividing, non-specialized state into a dead-end, mature state may lead to new ways to treat cancer.
Collapse
Affiliation(s)
- Joana Torres
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Remo Monti
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ariane L Moore
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Makiko Seimiya
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Yanrui Jiang
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jorge V Beira
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Renato Paro
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|
23
|
Baik H, Boulanger M, Hosseini M, Kowalczyk J, Zaghdoudi S, Salem T, Sarry JE, Hicheri Y, Cartron G, Piechaczyk M, Bossis G. Targeting the SUMO Pathway Primes All- trans Retinoic Acid-Induced Differentiation of Nonpromyelocytic Acute Myeloid Leukemias. Cancer Res 2018; 78:2601-2613. [PMID: 29487199 DOI: 10.1158/0008-5472.can-17-3361] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/18/2018] [Accepted: 02/23/2018] [Indexed: 11/16/2022]
Abstract
Differentiation therapies using all-trans retinoic acid (ATRA) are highly efficient at treating acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia (AML). However, their efficacy, if any, is limited in the case of non-APL AML. We report here that inhibition of SUMOylation, a posttranslational modification related to ubiquitination, restores the prodifferentiation and antiproliferative activities of retinoids in non-APL AML. Controlled inhibition of SUMOylation with the pharmacologic inhibitors 2-D08 or anacardic acid, or via overexpression of SENP deSUMOylases, enhanced the ATRA-induced expression of key genes involved in differentiation, proliferation, and apoptosis in non-APL AML cells. This activated ATRA-induced terminal myeloid differentiation and reduced cell proliferation and viability, including in AML cells resistant to chemotherapeutic drugs. Conversely, enhancement of SUMOylation via overexpression of the SUMO-conjugating enzyme Ubc9 dampened expression of ATRA-responsive genes and prevented differentiation. Thus, inhibition of the SUMO pathway is a promising strategy to sensitize patients with non-APL AML to retinoids and improve the treatment of this poor-prognosis cancer.Significance: SUMOylation silences key ATRA-responsive genes in nonpromyelocytic acute myeloid leukemias. Cancer Res; 78(10); 2601-13. ©2018 AACR.
Collapse
Affiliation(s)
- Hayeon Baik
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Mathias Boulanger
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Mohsen Hosseini
- Centre de Recherche en Cancérologie de Toulouse, Inserm and Université de Toulouse, Toulouse, France
| | - Julie Kowalczyk
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Département d'Hématologie Clinique, CHU de Montpellier, Montpellier, France
| | - Sonia Zaghdoudi
- Centre de Recherche en Cancérologie de Toulouse, Inserm and Université de Toulouse, Toulouse, France
| | - Tamara Salem
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Jean-Emmanuel Sarry
- Centre de Recherche en Cancérologie de Toulouse, Inserm and Université de Toulouse, Toulouse, France
| | - Yosr Hicheri
- Département d'Hématologie Clinique, CHU de Montpellier, Montpellier, France
| | - Guillaume Cartron
- Département d'Hématologie Clinique, CHU de Montpellier, Montpellier, France
| | - Marc Piechaczyk
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.
| | - Guillaume Bossis
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
24
|
De Nicola M, Bruni E, Traversa E, Ghibelli L. Slow release of etoposide from dextran conjugation shifts etoposide activity from cytotoxicity to differentiation: A promising tool for dosage control in anticancer metronomic therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2005-2014. [PMID: 28535989 DOI: 10.1016/j.nano.2017.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/30/2017] [Accepted: 05/08/2017] [Indexed: 11/27/2022]
Abstract
Drug conjugation, improving drug stability, solubility and body permanence, allows achieving impressive results in tumor control. Here, we show that conjugation may provide a straightforward method to administer drugs by the emerging anticancer metronomic approach, presently consisting of low, repeated doses of cytotoxic drugs used in traditional chemotherapy, thus reducing toxicity without reducing efficiency; however, low dose maintenance in tumor sites is difficult. We show that conjugating the antitumor drug etoposide to dextran via pH-sensitive bond produces slow releasing, apoptosis-proficient conjugates rapidly internalized into acidic lysosomes; importantly, release of active etoposide requires cell internalization and acidic pH. Conjugation, without impairing etoposide-induced complete elimination of tumor cells, shifted the mode of apoptosis from cytotoxicity- to differentiation-related; interestingly, high conjugate doses acted as low doses of free etoposide, thus mimicking the effect of metronomic therapy. This indicates slow release as a promising novel strategy for stabilizing low drug levels in metronomic regimens.
Collapse
Affiliation(s)
- Milena De Nicola
- Dipartimento di Biologia, Università di Roma Tor Vergata, Roma, Italy; Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Roma, Italy.
| | - Emanuele Bruni
- Dipartimento di Biologia, Università di Roma Tor Vergata, Roma, Italy.
| | - Enrico Traversa
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Roma, Italy; International Research Center for Renewable Energy (IRCRE), Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Lina Ghibelli
- Dipartimento di Biologia, Università di Roma Tor Vergata, Roma, Italy.
| |
Collapse
|
25
|
Gao J, Fan M, Xiang G, Wang J, Zhang X, Guo W, Wu X, Sun Y, Gu Y, Ge H, Tan R, Qiu H, Shen Y, Xu Q. Diptoindonesin G promotes ERK-mediated nuclear translocation of p-STAT1 (Ser727) and cell differentiation in AML cells. Cell Death Dis 2017; 8:e2765. [PMID: 28471454 PMCID: PMC5520695 DOI: 10.1038/cddis.2017.159] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/26/2017] [Accepted: 03/13/2017] [Indexed: 12/22/2022]
Abstract
Exploration of a new differentiation therapy that extends the range of differentiation for treating acute myeloid leukemia (AML) is attractive to researchers and clinicians. Here we report that diptoindonesin G (Dip G), a natural resveratrol aneuploid, exerts antiproliferative activity by inducing G2/M phase arrest and cell differentiation in AML cell lines and primary AML cells. Gene-profiling experiments showed that treating human leukemia HL-60 cells with Dip G was associated with a remarkable upregulation of STAT1 target gene expression, including IFIT3 and CXCL10. Mechanistically, Dip G activated ERK, which caused phosphorylation of STAT1 at Ser727 and selectively enhanced the interaction of p-STAT1 (Ser727) and p-ERK, further promoting their nuclear translocation. The nuclear translocation of p-STAT1 and p-ERK enhanced the transactivation of STAT1-targeted genes in AML cells. Furthermore, in vivo treatment of HL-60 xenografts demonstrated that Dip G significantly inhibited tumor growth and reduced tumor weight by inducing cell differentiation. Taken together, these results shed light on an essential role for ERK-mediated nuclear translocation of p-STAT1 (Ser727) and its full transcriptional activity in Dip G-induced differentiation of AML cells. Furthermore, these results demonstrate that Dip G could be used as a differentiation-inducing agent for AML therapy, particularly for non-acute promyelocytic leukemia therapy.
Collapse
Affiliation(s)
- Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Minmin Fan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Gang Xiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Jujuan Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yanhong Gu
- Department of Clinical Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Huiming Ge
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Renxiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China.,Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing University, Nanjing 210093, China
| | - Hongxia Qiu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China.,Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing University, Nanjing 210093, China
| |
Collapse
|
26
|
Sunami Y, Araki M, Kan S, Ito A, Hironaka Y, Imai M, Morishita S, Ohsaka A, Komatsu N. Histone Acetyltransferase p300/CREB-binding Protein-associated Factor (PCAF) Is Required for All- trans-retinoic Acid-induced Granulocytic Differentiation in Leukemia Cells. J Biol Chem 2017; 292:2815-2829. [PMID: 28053092 DOI: 10.1074/jbc.m116.745398] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/30/2016] [Indexed: 01/01/2023] Open
Abstract
Differentiation therapy with all-trans-retinoic acid (ATRA) improves the treatment outcome of acute promyelocytic leukemia (APL); however, the molecular mechanism by which ATRA induces granulocytic differentiation remains unclear. We previously reported that the inhibition of the NAD-dependent histone deacetylase (HDAC) SIRT2 induces granulocytic differentiation in leukemia cells, suggesting the involvement of protein acetylation in ATRA-induced leukemia cell differentiation. Herein, we show that p300/CREB-binding protein-associated factor (PCAF), a histone acetyltransferase (HAT), is a prerequisite for ATRA-induced granulocytic differentiation in leukemia cells. We found that PCAF expression was markedly increased in leukemia cell lines (NB4 and HL-60) and primary APL cells during ATRA-induced granulocytic differentiation. Consistent with these results, the expression of PCAF was markedly up-regulated in the bone marrow cells of APL patients who received ATRA-containing chemotherapy. The knockdown of PCAF inhibited ATRA-induced granulocytic differentiation in leukemia cell lines and primary APL cells. Conversely, the overexpression of PCAF induced the expression of the granulocytic differentiation marker CD11b at the mRNA level. Acetylome analysis identified the acetylated proteins after ATRA treatment, and we found that histone H3, a known PCAF acetylation substrate, was preferentially acetylated by the ATRA treatment. Furthermore, we have demonstrated that PCAF is required for the acetylation of histone H3 on the promoter of ATRA target genes, such as CCL2 and FGR, and for the expression of these genes in ATRA-treated leukemia cells. These results strongly support our hypothesis that PCAF is induced and activated by ATRA, and the subsequent acetylation of PCAF substrates promotes granulocytic differentiation in leukemia cells. Targeting PCAF and its downstream acetylation targets could serve as a novel therapeutic strategy to overcome all subtypes of AML.
Collapse
Affiliation(s)
| | - Marito Araki
- Department of Transfusion Medicine and Stem Cell Regulation, and
| | - Shin Kan
- From the Department of Hematology.,Leading Center for the Development and Research of Cancer Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan and
| | - Akihiro Ito
- the Chemical Genetics Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | - Misa Imai
- Leading Center for the Development and Research of Cancer Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan and
| | - Soji Morishita
- Department of Transfusion Medicine and Stem Cell Regulation, and
| | - Akimichi Ohsaka
- Department of Transfusion Medicine and Stem Cell Regulation, and
| | | |
Collapse
|
27
|
Afzali M, Baharara J, Nezhad Shahrokhabadi K, Amini E. Evaluation of the Cytotoxic Effect of the Brittle Star (Ophiocoma Erinaceus) Dichloromethane Extract and Doxorubicin on EL4 Cell Line. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2017; 16:216-226. [PMID: 29844793 PMCID: PMC5963663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Leukemia is a blood disease that creates from inhibition of differentiation and increased proliferation rate. The nature has been known as a rich source of medically useful substances. High diversity of bioactive molecules, extracted from marine invertebrates, makes them as ideal candidates for cancer research. The study has been done to investigate cytotoxic effects of dichloromethane brittle star extract and doxorubicin on EL4 cancer cells. Blood cancer EL4 cells were cultured and treated at different concentrations of brittle star (Ophiocoma erinaceus) dichloromethane extract at 24, 48 and 72 h. Cell toxicity was studied using MTT assay. Cell morphology was examined using an invert microscope. Further, apoptosis was examined using Annexin V-FITC, propodium iodide, DAPI, and Acridine orange/propodium iodide staining. Eventually, the apoptosis pathways were analyzed using measurement of Caspase-3 and -9 activity. The statistical analysis was performed using SPSS, ANOVA software, and Tukey's test. P<0.05 was considered to be significant. MTT assay and morphological observations showed that dichloromethane extract can inhibit cell growth in a dose dependent. The results considered 32 µg/mL of the extract as IC50. Also, doxorubicin suppressed EL4 proliferation as IC50=32 µg/mL. All experiments related to apoptosis analysis confirmed that dichloromethane brittle star extract and doxorubicin have a cytotoxic effect on EL4 cells inIC50 concentration. The study showed that dichloromethane brittle star extract is as an adjunct to doxorubicin in treatment of leukemia cells.
Collapse
Affiliation(s)
- Mahbubeh Afzali
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Javad Baharara
- Research Center for Animal Development Applied Biology & Biology Department, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | | | - Elaheh Amini
- Department of Cellular & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
28
|
Tian P, Yan L. Inhibition of MicroRNA-149-5p Induces Apoptosis of Acute Myeloid Leukemia Cell Line THP-1 by Targeting Fas Ligand (FASLG). Med Sci Monit 2016; 22:5116-5123. [PMID: 28013316 PMCID: PMC5207011 DOI: 10.12659/msm.899114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background This study was aimed to reveal the role of miR-149-5p in acute myeloid leukemia (AML) cells apoptosis and the possible mechanism involved. Material/Methods The expression of miR-149-5p in leukemia cell lines, as well as the blood and bone marrow (BM) samples from leukemia patients, were monitored by reverse-transcription polymerase chain reaction (RT-PCR). AML cell line THP-1 was transfected with miR-149-5p mimic or inhibitor, and then cell apoptosis was determined using the APO Percentage assay kit. The target of miR-149-5p was predicted by using the microRNA.org database, and verified by RT-PCR, Western blot, and Dual-Luciferase reporter assays. Further, small interfering RNA (siRNA) against the target gene was co-transfected with miR-149-5p inhibitor, and then the cell apoptosis and the expression of apoptosis-related proteins were assessed. Results MiR-149-5p was significantly up-regulated in leukemia cell lines and samples from leukemia patients (P<0.01 or P<0.001), especially in THP-1 cells and samples from AML patients. Cell apoptosis was significantly decreased by miR-149-5p overexpression (P<0.01) and increased by miR-149-5p suppression (P<0.05). Fas Ligand (FASLG) was a direct target of miR-149-5p, and was negatively regulated by miR-149-5p. More importantly, the inductive effects of miR-149-5p suppression on cell apoptosis were abrogated by si-FASLG (P<0.01). Furthermore, the up-regulative effects of miR-149-5p suppression on the phosphorylated form of Fas-associated via death domain (p-FADD), caspase-8, caspase-2, caspase-3, and the cleaved forms of these caspases were abrogated by si-FASLG. Conclusions Inhibition of miR-149-5p can induce apoptosis in THP-1 cells. These inductive effects might be via targeting FASLG and activating FADD and caspases.
Collapse
Affiliation(s)
- Peijun Tian
- Department of Haematology, Ankang City Central Hospitalg, Ankang, Shaanxi, China (mainland)
| | - Li Yan
- Tumor Department of Hematology, Shaanxi Province Chinese Medicine Hospital, Ankang, Shaanxi, China (mainland)
| |
Collapse
|
29
|
Identification of H7 as a novel peroxiredoxin I inhibitor to induce differentiation of leukemia cells. Oncotarget 2016; 7:3873-83. [PMID: 26716647 PMCID: PMC4826176 DOI: 10.18632/oncotarget.6763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/28/2015] [Indexed: 01/02/2023] Open
Abstract
Identifying novel targets to enhance leukemia-cell differentiation is an urgent requirment. We have recently proposed that inhibiting the antioxidant enzyme peroxiredoxin I (Prdx I) may induce leukemia-cell differentiation. However, this concept remains to be confirmed. In this work, we identified H7 as a novel Prdx I inhibitor through virtual screening, in vitro activity assay, and surface plasmon resonance assay. Cellular thermal shift assay showed that H7 directly bound to Prdx I but not to Prdxs II–V in cells. H7 treatment also increased reactive oxygen species (ROS) level and cell differentiation in leukemia cells, as reflected by the upregulation of the cell surface differentiation marker CD11b/CD14 and the morphological maturation of cells. The differentiation-induction effect of H7 was further observed in some non-acute promyelocytic leukemia (APL) and primary leukemia cells apart from APL NB4 cells. Moreover, the ROS scavenger N-acetyl cysteine significantly reversed the H7-induced cell differentiation. We demonstrated as well that H7-induced cell differentiation was associated with the activation of the ROS-Erk1/2-C/EBPβ axis. Finally, we showed H7 treatment induced cell differentiation in an APL mouse model. All of these data confirmed that Prdx I was novel target for inducing leukemia-cell differentiation and that H7 was a novel lead compound for optimizing Prdx I inhibition.
Collapse
|
30
|
Sierra-Rivera CA, Franco-Molina MA, Mendoza-Gamboa E, Zapata-Benavides P, Santaolalla-Tapia J, Coronado-Cerda EE, Tamez-Guerra RS, Rodríguez-Padilla C. Effect of bovine dialyzable leukocyte extract on induction of cell differentiation and death in K562 human chronic myelogenous leukemia cells. Oncol Lett 2016; 12:4449-4460. [PMID: 28101208 PMCID: PMC5228060 DOI: 10.3892/ol.2016.5285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/22/2016] [Indexed: 01/08/2023] Open
Abstract
Differentiation induction therapy is an attractive approach in leukemia treatment due to the fact that in blast crisis stage, leukemic cells lose their differentiation capacity. Therefore, it has been proposed as a therapeutic strategy to induce terminal differentiation of leukemic blast cells into a specific lineage, leading to prevention of high proliferation rates. The aim of the present study was to demonstrate the potential of cell differentiation and death induced by bovine dialyzable leukocyte extract (bDLE) in the K562 cell line. For this purpose K562 and MOLT-3 human leukemic cell lines and primary human monocytes and murine peritoneal macrophages were exposed to bDLE, phorbol myristate acetate (PMA) and dimethyl sulfoxide for 96 h, and the viability, proliferation and cell cycle were evaluated. To determine the lineage that led to cell differentiation, Romanowsky staining was performed to observe the morphological changes following the treatments, and the expression of the surface markers cluster of differentiation (CD)14+, CD68+, CD163+ and CD42a+, as well as the phagocytic activity, and the production of nitric oxide (NO) (assessed by colorimetric assay), cytokines [interleukin (IL)-1β, IL-6, IL-8 and tumor necrosis factor-α] and chemokines [chemokine (C-C motif) ligand (CCL)2, CCL5 and chemokine (C-X-C motif) ligand 8] in cell supernatants was assessed by flow cytometry. The results of the present study reveal that high doses of bDLE increase the cell death in K562 and MOLT-3 lines, without affecting the viability of human monocytes and murine peritoneal macrophages. Furthermore, low doses of bDLE induce differentiation in K562 cells towards a monocyte/macrophage lineage with an M2 phenotype, and induced moderately upregulated expression of CD42+, a megakaryocytic marker. Cell cycle arrest in the S and G2/M phases was observed in bDLE-treated K562 cells, which demonstrated similar phagocytic activity, NO levels and cytokine and chemokine production to that of PMA-treated cells. The present study demonstrates that bDLE exhibits an antileukemia effect, suggesting that it may be an effective candidate for leukemia treatment.
Collapse
Affiliation(s)
- Crystel A Sierra-Rivera
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| | - Moisés A Franco-Molina
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| | - Edgar Mendoza-Gamboa
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| | - Pablo Zapata-Benavides
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| | - Jesús Santaolalla-Tapia
- Laboratory of Stem Cell Biology, Faculty of Medicine, University Autonomous of Morelos, Cuernavaca, Morelos 62350, Mexico
| | - Erika E Coronado-Cerda
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| | - Reyes S Tamez-Guerra
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| | - Cristina Rodríguez-Padilla
- Laboratory of Immunology and Virology, Faculty of Biological Sciences, University Autonomous of Nuevo León (UANL), San Nicolás de los Garza, Nuevo León 66450, Mexico
| |
Collapse
|
31
|
Heo SK, Noh EK, Gwon GD, Kim JY, Jo JC, Choi Y, Koh S, Baek JH, Min YJ, Kim H. Radotinib inhibits acute myeloid leukemia cell proliferation via induction of mitochondrial-dependent apoptosis and CDK inhibitors. Eur J Pharmacol 2016; 789:280-290. [PMID: 27477352 DOI: 10.1016/j.ejphar.2016.07.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/22/2016] [Accepted: 07/27/2016] [Indexed: 12/27/2022]
Abstract
Radotinib is a BCR-ABL1 tyrosine kinase inhibitor approved for the second-line treatment of chronic myeloid leukemia. However, effects of radotinib on acute myeloid leukemia (AML) are unclear. In the present study, we observed that radotinib exerted cytotoxic effects on AML cells. Of the various AML cell lines examined (NB4, HL60, HEL 92.1.7, and THP-1), Kasumi-1 was the most sensitive to radotinib. Results of microarray analysis showed that 417 and 595 genes associated with apoptosis and cell cycle regulation, respectively, were differently expressed (i.e., showed >2-fold difference in expression). Radotinib-induced apoptosis involved the mitochondrial pathway. Moreover, radotinib increased the apoptosis of and induced caspase-3 activity in both Kasumi-1 cells and bone marrow cells (BMCs) obtained from patients with AML. Radotinib also increased cleaved caspase-3, caspase-7, and caspase-9 levels and decreased the number of proliferating Kasumi-1 cells and BMCs from patients with AML. In addition, radotinib induced G0/G1 phase arrest by inducing CDKIs p21 and p27 and by inhibiting CDK2, CDK4, and CDK6. These results indicate that radotinib induces caspase-dependent apoptosis and G0/G1 phase arrest in AML cells by regulating CDKI-CDK-cyclin cascade. Moreover, these results indicate that radotinib inhibits AML cell proliferation by inducing mitochondria-dependent apoptosis and CDKIs p21 and p27. To our knowledge, this is the first study to show that radotinib can be potentially used for the anti-leukemic therapy of patients with AML.
Collapse
Affiliation(s)
- Sook-Kyoung Heo
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Republic of Korea
| | - Eui-Kyu Noh
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea
| | - Gi-Dong Gwon
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Republic of Korea
| | - Jeong Yi Kim
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Republic of Korea
| | - Jae-Cheol Jo
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea
| | - Yunsuk Choi
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea
| | - SuJin Koh
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea
| | - Jin Ho Baek
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea
| | - Young Joo Min
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea
| | - Hawk Kim
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Republic of Korea; Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-714, Republic of Korea.
| |
Collapse
|
32
|
King YA, Chiu YJ, Chen HP, Kuo DH, Lu CC, Yang JS. Endoplasmic reticulum stress contributes to arsenic trioxide-induced intrinsic apoptosis in human umbilical and bone marrow mesenchymal stem cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:314-328. [PMID: 25258189 DOI: 10.1002/tox.22046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 09/04/2014] [Indexed: 06/03/2023]
Abstract
Arsenic trioxide is an old drug and has been used for a long time in traditional Chinese and Western medicines. However, the cancer treatment of arsenic trioxide has heart and vascular toxicity. The cytotoxic effects of arsenic trioxide and its molecular mechanism in human umbilical mesenchymal stem cells (HUMSC) and human bone marrow-derived mesenchymal stem cells (HMSC-bm) were investigated in this study. Our results showed that arsenic trioxide significantly reduced the viability of HUMSC and HMSC-bm in a concentration- and time-dependent manner. Arsenic trioxide is able to induce apoptotic cell death in HUMSC and HMSC-bm, as shown from the results of morphological examination, flow cytometric analyses, DAPI staining and comet assay. The appearance of arsenic trioxide also led to an increase of intracellular free calcium (Ca(2+) ) concentration and the disruption of mitochondrial membrane potential (ΔΨm). The caspase-9 and caspase-3 activities were time-dependently increased in arsenic trioxide-treated HUMSC and HMSC-bm. In addition, the proteomic analysis and DNA microarray were carried out to investigate the expression level changes of genes and proteins affected by arsenic trioxide treatment in HUMSC. Our results suggest that arsenic trioxide induces a prompt induction of ER stress and mitochondria-modulated apoptosis in HUMSC and HMSC-bm. A framework was proposed for the effect of arsenic trioxide cytotoxicity by targeting ER stress.
Collapse
Affiliation(s)
- Yih-An King
- Department of Dermatology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yu-Jen Chiu
- Division of Reconstructive and Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hao-Ping Chen
- Department of Biochemistry, Tzu Chi University, Hualien, Taiwan
| | - Daih-Huang Kuo
- Department of Pharmacy and Graduate Institute of Pharmaceutical Technology, Tajen University, Pingtung, Taiwan
| | - Chi-Cheng Lu
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | | |
Collapse
|
33
|
Kühn K, Cott C, Bohler S, Aigal S, Zheng S, Villringer S, Imberty A, Claudinon J, Römer W. The interplay of autophagy and β-Catenin signaling regulates differentiation in acute myeloid leukemia. Cell Death Discov 2015; 1:15031. [PMID: 27551462 PMCID: PMC4979480 DOI: 10.1038/cddiscovery.2015.31] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/03/2015] [Accepted: 08/07/2015] [Indexed: 12/18/2022] Open
Abstract
The major feature of leukemic cells is an arrest of differentiation accompanied by highly active proliferation. In many subtypes of acute myeloid leukemia, these features are mediated by the aberrant Wnt/β-Catenin pathway. In our study, we established the lectin LecB as inducer of the differentiation of the acute myeloid leukemia cell line THP-1 and used it for the investigation of the involved processes. During differentiation, functional autophagy and low β-Catenin levels were essential. Corresponding to this, a high β-Catenin level stabilized proliferation and inhibited autophagy, resulting in low differentiation ability. Initiated by LecB, β-Catenin was degraded, autophagy became active and differentiation took place within hours. Remarkably, the reduction of β-Catenin sensitized THP-1 cells to the autophagy-stimulating mTOR inhibitors. As downmodulation of E-Cadherin was sufficient to significantly reduce LecB-mediated differentiation, we propose E-Cadherin as a crucial interaction partner in this signaling pathway. Upon LecB treatment, E-Cadherin colocalized with β-Catenin and thereby prevented the induction of β-Catenin target protein expression and proliferation. That way, our study provides for the first time a link between E-Cadherin, the aberrant Wnt/β-Catenin signaling, autophagy and differentiation in acute myeloid leukemia. Importantly, LecB was a valuable tool to elucidate the underlying molecular mechanisms of acute myeloid leukemia pathogenesis and may help to identify novel therapy approaches.
Collapse
Affiliation(s)
- K Kühn
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| | - C Cott
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| | - S Bohler
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| | - S Aigal
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany; International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - S Zheng
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| | - S Villringer
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| | - A Imberty
- Centre de Recherches sur les Macromolécules Végétales (CERMAV), CNRS and Université Grenoble Alpes , 601 rue de la chimie, 38000 Grenoble, France
| | - J Claudinon
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| | - W Römer
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraβe 1, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraβe 18, 79104 Freiburg, Germany
| |
Collapse
|
34
|
Baurska H, Kiełbiński M, Biecek P, Haus O, Jaźwiec B, Kutner A, Marcinkowska E. Monocytic differentiation induced by side-chain modified analogs of vitamin D in ex vivo cells from patients with acute myeloid leukemia. Leuk Res 2014; 38:638-47. [PMID: 24703772 DOI: 10.1016/j.leukres.2014.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/31/2014] [Accepted: 03/11/2014] [Indexed: 12/20/2022]
Abstract
The differentiation-inducing potential of side-chain modified analogs of vitamins D, compared to the reference compound, 1,25-dihydroxyvitamin D3, was studied in blast cells from patients with acute myeloid leukemia and in cell lines. Analogs PRI-1906 and PRI-1907 showed increased cell-differentiation activities, PRI-1907 even at a very low concentration. Our study revealed a high variability of individual patients' blasts in their susceptibility to vitamin D analogs. The blasts of the patients with normal karyotype and with mutated NPM1 reacted to analogs with stronger differentiation than the blasts of the remaining patients, while the blasts with mutated FLT3 receptor reacted with weaker differentiation than the remaining blasts.
Collapse
Affiliation(s)
- Hanna Baurska
- Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Marek Kiełbiński
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wrocław Medical University, Pasteura 4, 50-367 Wrocław, Poland
| | - Przemysław Biecek
- Institute of Applied Mathematics and Mechanics, University of Warsaw, Krakowskie Przedmieście 26/28, Warszawa, Poland
| | - Olga Haus
- Department of Clinical Genetics, Collegium Medicum, Nicolaus Copernicus University, Skłodowska-Curie 9, 85-094 Bydgoszcz, Poland
| | - Bożena Jaźwiec
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wrocław Medical University, Pasteura 4, 50-367 Wrocław, Poland
| | - Andrzej Kutner
- Pharmaceutical Research Institute, Rydygiera 8, 01-793 Warszawa, Poland
| | - Ewa Marcinkowska
- Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland.
| |
Collapse
|