1
|
Cabrera-Serrano AJ, Sánchez-Maldonado JM, González-Olmedo C, Carretero-Fernández M, Díaz-Beltrán L, Gutiérrez-Bautista JF, García-Verdejo FJ, Gálvez-Montosa F, López-López JA, García-Martín P, Pérez EM, Sánchez-Rovira P, Reyes-Zurita FJ, Sainz J. Crosstalk Between Autophagy and Oxidative Stress in Hematological Malignancies: Mechanisms, Implications, and Therapeutic Potential. Antioxidants (Basel) 2025; 14:264. [PMID: 40227235 PMCID: PMC11939785 DOI: 10.3390/antiox14030264] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy is a fundamental cellular process that maintains homeostasis by degrading damaged components and regulating stress responses. It plays a crucial role in cancer biology, including tumor progression, metastasis, and therapeutic resistance. Oxidative stress, similarly, is key to maintaining cellular balance by regulating oxidants and antioxidants, with its disruption leading to molecular damage. The interplay between autophagy and oxidative stress is particularly significant, as reactive oxygen species (ROS) act as both inducers and by-products of autophagy. While autophagy can function as a tumor suppressor in early cancer stages, it often shifts to a pro-tumorigenic role in advanced disease, aiding cancer cell survival under adverse conditions such as hypoxia and nutrient deprivation. This dual role is mediated by several signaling pathways, including PI3K/AKT/mTOR, AMPK, and HIF-1α, which coordinate the balance between autophagic activity and ROS production. In this review, we explore the mechanisms by which autophagy and oxidative stress interact across different hematological malignancies. We discuss how oxidative stress triggers autophagy, creating a feedback loop that promotes tumor survival, and how autophagic dysregulation leads to increased ROS accumulation, exacerbating tumorigenesis. We also examine the therapeutic implications of targeting the autophagy-oxidative stress axis in cancer. Current strategies involve modulating autophagy through specific inhibitors, enhancing ROS levels with pro-oxidant compounds, and combining these approaches with conventional therapies to overcome drug resistance. Understanding the complex relationship between autophagy and oxidative stress provides critical insights into novel therapeutic strategies aimed at improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Carmen González-Olmedo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - María Carretero-Fernández
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - Leticia Díaz-Beltrán
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Juan Francisco Gutiérrez-Bautista
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Servicio de Análisis Clínicos e Inmunología, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Department of Biochemistry, Molecular Biology and Immunology III, University of Granada, 18016 Granada, Spain
| | - Francisco José García-Verdejo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Gálvez-Montosa
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - José Antonio López-López
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Paloma García-Martín
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Eva María Pérez
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Pedro Sánchez-Rovira
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Jesús Reyes-Zurita
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
2
|
Shivhare K, Satija NK. Acute Myeloid Leukemia-Osteoblast Interaction Mediated Autophagy Induction Protects Against Cytarabine Induced Apoptosis. Cell Biochem Funct 2025; 43:e70055. [PMID: 39936300 DOI: 10.1002/cbf.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
High rate of relapse, following chemotherapy, in acute myeloid leukemia (AML) is a major concern. The chemoprotection conferred by the bone marrow microenvironment has lately been recognized, in addition to autophagy-mediated chemoresistance. Thus, the present study explored the effect of osteoblast on autophagy in AML and its impact on sensitivity to cytarabine (Ara-C) in the context of endosteal niche. Co-culture of KG1-a, HL60, or THP-1 AML cells with osteoblastic Saos-2 cell line induced autophagy in AML cell lines under direct contact. HL60 cells when co-culture with Saos-2 demonstrated more resistance to Ara-C induced apoptosis, which was reversed upon chloroquine treatment. Similarly, inhibition of autophagy in AML cell by knocking down Beclin-1 enhanced HL60 sensitivity to Ara-C. An interesting observation was upregulation of autophagy even in Saos-2 cells upon co-culture with AML cell, and increase in HL60 apoptosis in response to Ara-C on Beclin-1 knockdown in osteoblast cell. This highlights that autophagy plays a chemoprotective role in the endosteal niche in AML against Ara-C.
Collapse
Affiliation(s)
- Kamini Shivhare
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Neeraj Kumar Satija
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Salwa A, Ferraresi A, Vallino L, Maheshwari C, Moia R, Gaidano G, Isidoro C. High Mitophagy and Low Glycolysis Predict Better Clinical Outcomes in Acute Myeloid Leukemias. Int J Mol Sci 2024; 25:11527. [PMID: 39519080 PMCID: PMC11546612 DOI: 10.3390/ijms252111527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Acute myeloid leukemia (AML) emerges as one of the most common and fatal leukemias. Treatment of the disease remains highly challenging owing to profound metabolic rewiring mechanisms that confer plasticity to AML cells, ultimately resulting in therapy resistance. Autophagy, a highly conserved lysosomal-driven catabolic process devoted to macromolecular turnover, displays a dichotomous role in AML by suppressing or promoting disease development and progression. Glycolytic metabolism represents a pivotal strategy for AML cells to sustain increasing energy needs related to uncontrolled growth during disease progression. In this study, we tested the hypothesis that a high glycolytic rate and low autophagy flux could represent an advantage for AML cell proliferation and thus be detrimental for patient's prognosis, and vice versa. TCGA in silico analysis of the AML cohort shows that the high expression of MAP1LC3B (along with that of BECN1 and with low expression of p62/SQSTM1) and the high expression of BNIP3 (along with that of PRKN and of MAP1LC3B), which together are indicative of increased autophagy and mitophagy, correlate with better prognosis. On the other hand, the high expression of glycolytic markers HK2, PFKM, and PKM correlates with poor prognosis. Most importantly, the association of a low expression of glycolytic markers with a high expression of autophagy-mitophagy markers conferred the longest overall survival for AML patients. Transcriptomic analysis showed that this combined signature correlates with the downregulation of a subset of genes required for the differentiation of myeloid cells, lactate/pyruvate transporters, and cell cycle progression, in parallel with the upregulation of genes involved in autophagy/lysosomal trafficking and proteolysis, anti-tumor responses like beta-interferon production, and positive regulation of programmed cell death. Taken together, our data support the view that enhanced autophagy-mitophagy flux together with low glycolytic rate predisposes AML patients to a better clinical outcome, suggesting that autophagy inducers and glucose restrictors may hold potential as adjuvant therapeutics for improving AML management.
Collapse
Affiliation(s)
- Amreen Salwa
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Chinmay Maheshwari
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy;
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy;
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| |
Collapse
|
4
|
Walweel N, Aydin O. Enhancing Therapeutic Efficacy in Cancer Treatment: Integrating Nanomedicine with Autophagy Inhibition Strategies. ACS OMEGA 2024; 9:27832-27852. [PMID: 38973850 PMCID: PMC11223161 DOI: 10.1021/acsomega.4c02234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/01/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024]
Abstract
The complicated stepwise lysosomal degradation process known as autophagy is in charge of destroying and eliminating damaged organelles and defective cytoplasmic components. This mechanism promotes metabolic adaptability and nutrition recycling. Autophagy functions as a quality control mechanism in cells that support homeostasis and redox balance under normal circumstances. However, the role of autophagy in cancer is controversial because, mostly depending on the stage of the tumor, it may either suppress or support the disease. While autophagy delays the onset of tumors and slows the dissemination of cancer in the early stages of tumorigenesis, numerous studies demonstrate that autophagy promotes the development and spread of tumors as well as the evolution and development of resistance to several anticancer drugs in advanced cancer stages. In this Review, we primarily emphasize the therapeutic role of autophagy inhibition in improving the treatment of multiple cancers and give a broad overview of how its inhibition modulates cancer responses. There have been various attempts to inhibit autophagy, including the use of autophagy inhibitor drugs, gene silencing therapy (RNA interference), and nanoparticles. In this Review, all these topics are thoroughly covered and illustrated by recent studies and field investigations.
Collapse
Affiliation(s)
- Nada Walweel
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera
Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
| | - Omer Aydin
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera
Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
- ERNAM-Nanotechnology
Research and Application Center, Erciyes
University, Kayseri 38039, Turkey
- ERKAM-Clinical-Engineering
Research and Implementation Center, Erciyes
University, Kayseri 38030, Turkey
| |
Collapse
|
5
|
Li M, Li J, Zhang S, Zhou L, Zhu Y, Li S, Li Q, Wang J, Song R. Progress in the study of autophagy-related proteins affecting resistance to chemotherapeutic drugs in leukemia. Front Cell Dev Biol 2024; 12:1394140. [PMID: 38887520 PMCID: PMC11180896 DOI: 10.3389/fcell.2024.1394140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Leukemia is a life-threatening malignant tumor of the hematopoietic system. Currently, the main treatment modalities are chemotherapy and hematopoietic stem cell transplantation. However, increased drug resistance due to decreased sensitivity of leukemia cells to chemotherapeutic drugs presents a major challenge in current treatments. Autophagy-associated proteins involved in autophagy initiation have now been shown to be involved in the development of various types of leukemia cells and are associated with drug resistance. Therefore, this review will explore the roles of autophagy-related proteins involved in four key autophagic processes: induction of autophagy and phagophore formation, phagophore extension, and autophagosome formation, on the development of various types of leukemias as well as drug resistance. Autophagy may become a promising therapeutic target for treating leukemia.
Collapse
Affiliation(s)
- Meng Li
- Nursing Department, The Third People’s Hospital of Henan Province, Zhengzhou, China
| | - Jing Li
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shiming Zhang
- Clinical College, Xiamen Medical University, Xiamen, Fujian, China
| | - Linghan Zhou
- Nursing Department, The Third People’s Hospital of Henan Province, Zhengzhou, China
| | - Yuanyuan Zhu
- Nursing Department, The Third People’s Hospital of Henan Province, Zhengzhou, China
| | - Shen Li
- Rehabilitation Department, Henan Institute of Massage, Luoyang, Henan, China
| | - Qiong Li
- Nursing Department, Xinxiang Medical University, Xinxiang, China
| | - Junjie Wang
- Plastic Surgery, The Third People’s Hospital of Henan Province, Zhengzhou, China
| | - Ruipeng Song
- Endocrinology Department, The Third People’s Hospital of Henan Province, Zhengzhou, China
| |
Collapse
|
6
|
Kubota Y, Hoshiko T, Higashi T, Motoyama K, Okada S, Kimura S. Folate-Appended Hydroxypropyl-β-Cyclodextrin Induces Autophagic Cell Death in Acute Myeloid Leukemia Cells. Int J Mol Sci 2023; 24:16720. [PMID: 38069042 PMCID: PMC10706821 DOI: 10.3390/ijms242316720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous myeloid neoplasm that remains challenging to treat. Because intensive conventional chemotherapy reduces survival rates in elderly patients, drugs with lower toxicity and fewer side effects are needed urgently. 2-Hydroxypropyl-β-cyclodextrin (HP-β-CyD) is used clinically as a pharmaceutical excipient for poorly water-soluble drugs. Previously, we showed that HP-β-CyD exerts antitumor activity by disrupting cholesterol homeostasis. Recently, we developed folate-conjugated HP-β-CyD (FA-HP-β-CyD) and demonstrated its potential as a new antitumor agent that induces not only apoptosis, but also autophagic cell death; however, we do not know whether FA-HP-β-CyD exerts these effects against AML. Here, we investigated the effects of FA-HP-β-CyD on folate receptor (FR)-expressing AML cells. We found that the cytotoxic activity of FA-HP-β-CyD against AML cells was stronger than that of HP-β-CyD. Also, FA-HP-CyD induced the formation of autophagosomes in AML cell lines. FA-HP-β-CyD increased the inhibitory effects of cytarabine and a BCL-2-selective inhibitor, Venetoclax, which are commonly used treat elderly AML patients. Notably, FA-HP-β-CyD suppressed the proliferation of AML cells in BALB/c nude recombinase-activating gene-2 (Rag-2)/Janus kinase 3 (Jak3) double-deficient mice with AML. These results suggest that FA-HP-β-CyD acts as a potent anticancer agent for AML chemotherapy by regulating autophagy.
Collapse
Affiliation(s)
- Yasushi Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan;
- Department of Transfusion Medicine and Cell Therapy, Saitama Medical Center, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Toshimi Hoshiko
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan;
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (T.H.); (K.M.)
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (T.H.); (K.M.)
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto 860-0811, Japan;
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan;
| |
Collapse
|
7
|
Yan Y, Li L, Wang Z, Pang J, Guan X, Yuan Y, Xia Z, Yi W. A comprehensive analysis of the role of QPRT in breast cancer. Sci Rep 2023; 13:15414. [PMID: 37723185 PMCID: PMC10507026 DOI: 10.1038/s41598-023-42566-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023] Open
Abstract
To explore the clinical role of QPRT in breast cancer. The gene expression, methylation levels and prognostic value of QPRT in breast cancer was analyzed using TCGA data. Validation was performed using the data from GEO dataset and TNMPLOT database. Meta analysis method was used to pool the survival data for QPRT. The predictive values of QPRT for different drugs were retrieved from the ROC plot. The expression differences of QPRT in acquired drug-resistant and sensitive cell lines were analyzed using GEO datasets. GO and KEGG enrichment analysis were conducted for those genes which were highly co-expressed with QPRT in tissue based on TCGA data and which changed after QPRT knockdown. Timer2.0 was utilized to explore the correlation between QPRT and immune cells infiltration, and the Human Protein Atlas was used to analyse QPRT's single-cell sequencing data across different human tissues. The expression of QPRT in different types of macrophages, and the expression of QPRT were analysed after coculturing HER2+ breast cancer cells with macrophages. Additionally, TargetScan, Comparative Toxicogenomics and the connectivity map were used to research miRNAs and drugs that could regulate QPRT expression. Cytoscape was used to map the interaction networks between QPRT and other proteins. QPRT was highly expressed in breast cancer tissue and highly expressed in HER2+ breast cancer patients (P < 0.01). High QPRT expression levels were associated with worse OS, DMFS, and RFS (P < 0.01). Two sites (cg02640602 and cg06453916) were found to be potential regulators of breast cancer (P < 0.01). QPRT might predict survival benefits in breast cancer patients who received taxane or anthracycline. QPRT was associated with tumour immunity, especially in macrophages. QPRT may influence the occurrence and progression of breast cancer through the PI3K-AKT signalling pathway, Wnt signalling pathway, and cell cycle-related molecules.
Collapse
Affiliation(s)
- Yiqing Yan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China
| | - Lun Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China
| | - Zixin Wang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China
| | - Jian Pang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China
| | - Xinyu Guan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China
| | - Yunchang Yuan
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China.
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha, 410011, China.
| |
Collapse
|
8
|
Choukrani G, Visser N, Ustyanovska Avtenyuk N, Olthuis M, Marsman G, Ammatuna E, Lourens HJ, Niki T, Huls G, Bremer E, Wiersma VR. Galectin-9 has non-apoptotic cytotoxic activity toward acute myeloid leukemia independent of cytarabine resistance. Cell Death Discov 2023; 9:228. [PMID: 37407572 DOI: 10.1038/s41420-023-01515-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is a malignancy still associated with poor survival rates, among others, due to frequent occurrence of therapy-resistant relapse after standard-of-care treatment with cytarabine (AraC). AraC triggers apoptotic cell death, a type of cell death to which AML cells often become resistant. Therefore, therapeutic options that trigger an alternate type of cell death are of particular interest. We previously identified that the glycan-binding protein Galectin-9 (Gal-9) has tumor-selective and non-apoptotic cytotoxicity towards various types of cancer, which depended on autophagy inhibition. Thus, Gal-9 could be of therapeutic interest for (AraC-resistant) AML. In the current study, treatment with Gal-9 was cytotoxic for AML cells, including for CD34+ patient-derived AML stem cells, but not for healthy cord blood-derived CD34+ stem cells. This Gal-9-mediated cytotoxicity did not rely on apoptosis but was negatively associated with autophagic flux. Importantly, both AraC-sensitive and -resistant AML cell lines, as well as AML patient samples, were sensitive to single-agent treatment with Gal-9. Additionally, Gal-9 potentiated the cytotoxic effect of DNA demethylase inhibitor Azacytidine (Aza), a drug that is clinically used for patients that are not eligible for intensive AraC treatment. Thus, Gal-9 is a potential therapeutic agent for the treatment of AML, including AraC-resistant AML, by inducing caspase-independent cell death.
Collapse
Affiliation(s)
- Ghizlane Choukrani
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nienke Visser
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Natasha Ustyanovska Avtenyuk
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Surflay Nanotec GmbH, Berlin, Germany
| | - Mirjam Olthuis
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Glenn Marsman
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Emanuele Ammatuna
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harm Jan Lourens
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Toshiro Niki
- Department of Immunology, Kagawa University, Takamatsu, Kagawa, Japan
| | - Gerwin Huls
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edwin Bremer
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Valerie R Wiersma
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
9
|
Chiou JT, Hsu CC, Hong YC, Lee YC, Chang LS. Cytarabine-induced destabilization of MCL1 mRNA and protein triggers apoptosis in leukemia cells. Biochem Pharmacol 2023; 211:115494. [PMID: 36924905 DOI: 10.1016/j.bcp.2023.115494] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/11/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023]
Abstract
Although cytarabine (Ara-C) is the mainstay of treatment for acute myeloid leukemia (AML), its cytotoxic mechanisms for inducing apoptosis are poorly understood. Therefore, we investigated the Ara-C-induced cell death pathway in human AML U937 cells. Ara-C-induced downregulation of MCL1 is associated with the induction of mitochondrial depolarization and apoptosis. Ara-C triggered NOX4-mediated ROS production, which in turn activated p38 MAPK but inactivated AKT. Ara-C-induced DNA damage modulates p38 MAPK activation without affecting AKT inactivation in U937 cells. Inactivated AKT promotes GSK3β-dependent CREB phosphorylation, which in turn increases NOXA transcription, thereby triggering the degradation of MCL1 protein. Activated p38 MAPK induces HuR downregulation, leading to accelerated MCL1 mRNA turnover. A similar pathway also explains the Ara-C-induced THP-1 cell death. Collectively, our data confirm that Ara-C-triggered apoptosis in the AML cell lines U937 and THP-1 is mediated through the destabilization of MCL1 mRNA and protein. Furthermore, Ara-C acts synergistically with the BCL2 inhibitor ABT-199 to induce cell death in ABT-199-resistant and parental U937 cells by inhibiting MCL1 expression.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Chia-Chi Hsu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Ying-Chung Hong
- Division of Hematology/Oncology, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
10
|
Ahmadi-Dehlaghi F, Mohammadi P, Valipour E, Pournaghi P, Kiani S, Mansouri K. Autophagy: A challengeable paradox in cancer treatment. Cancer Med 2023. [PMID: 36760166 DOI: 10.1002/cam4.5577] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/14/2022] [Accepted: 12/21/2022] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE Autophagy is an intracellular degradation pathway conserved in all eukaryotes from yeast to humans. This process plays a quality-control role by destroying harmful cellular components under normal conditions, maintaining cell survival, and establishing cellular adaptation under stressful conditions. Hence, there are various studies indicating dysfunctional autophagy as a factor involved in the development and progression of various human diseases, including cancer. In addition, the importance of autophagy in the development of cancer has been highlighted by paradoxical roles, as a cytoprotective and cytotoxic mechanism. Despite extensive research in the field of cancer, there are many questions and challenges about the roles and effects suggested for autophagy in cancer treatment. The aim of this study was to provide an overview of the paradoxical roles of autophagy in different tumors and related cancer treatment options. METHODS In this study, to find articles, a search was made in PubMed and Google scholar databases with the keywords Autophagy, Autophagy in Cancer Management, and Drug Design. RESULTS According to the investigation, some studies suggest that several advanced cancers are dependent on autophagy for cell survival, so when cancer cells are exposed to therapy, autophagy is induced and suppresses the anti-cancer effects of therapeutic agents and also results in cell resistance. However, enhanced autophagy from using anti-cancer drugs causes autophagy-mediated cell death in several cancers. Because autophagy also plays roles in both tumor suppression and promotion further research is needed to determine the precise mechanism of this process in cancer treatment. CONCLUSION We concluded in this article, autophagy manipulation may either promote or hinder the growth and development of cancer according to the origin of the cancer cells, the type of cancer, and the behavior of the cancer cells exposed to treatment. Thus, before starting treatment it is necessary to determine the basal levels of autophagy in various cancers.
Collapse
Affiliation(s)
- Farnaz Ahmadi-Dehlaghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Biology, Payame Noor University, Tehran, Iran
| | - Parisa Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elahe Valipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sarah Kiani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
11
|
Haghi A, Mohammadi Kian M, Salemi M, Eghdami MR, Nikbakht M. The Question of Survival or Death: What Is the Role of Autophagy in Acute Myeloid Leukemia (AML)? Int J Hematol Oncol Stem Cell Res 2022; 16:250-263. [PMID: 36883106 PMCID: PMC9985813 DOI: 10.18502/ijhoscr.v16i4.10883] [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: 02/13/2021] [Accepted: 01/11/2022] [Indexed: 03/09/2023] Open
Abstract
Autophagy plays a critical role in balancing sources of energy in response to harsh conditions and nutrient deprivation. Autophagy allows cells to survive in harsh condition and also serve as a death mechanism. Any dysregulation in autophagy signaling may lead to several disorders. Autophagy has been proposed to explain chemotherapy resistance in acute myeloid leukemia (AML). This signaling pathway can either act as a tumor suppressive function or chemo-resistance mechanism. Conventional chemotherapy drugs enhance apoptosis and indicate clinical benefit, but in some cases, relapse and chemotherapy resistance are observed. In leukemia, autophagy may promote cell survival in response to chemotherapy drugs. Therefore, new strategies by inhibiting or activating autophagy may find a broad application for treating leukemia and may significantly enhance clinical outcomes. In this review, we discussed the dimensional role of autophagy in leukemia.
Collapse
Affiliation(s)
- Atousa Haghi
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.,Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Mohammadi Kian
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Salemi
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.,Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Eghdami
- Department of Social Sciences, University of Guilan, Rasht, Iran.,Department of Biological Anthropology, Research Institute of Guilan Studies, University of Guilan, Rasht, Iran
| | - Mohsen Nikbakht
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.,Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Krunić M, Ristić B, Bošnjak M, Paunović V, Tovilović-Kovačević G, Zogović N, Mirčić A, Marković Z, Todorović-Marković B, Jovanović S, Kleut D, Mojović M, Nakarada Đ, Marković O, Vuković I, Harhaji-Trajković L, Trajković V. Graphene quantum dot antioxidant and proautophagic actions protect SH-SY5Y neuroblastoma cells from oxidative stress-mediated apoptotic death. Free Radic Biol Med 2021; 177:167-180. [PMID: 34678419 DOI: 10.1016/j.freeradbiomed.2021.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023]
Abstract
We investigated the ability of graphene quantum dot (GQD) nanoparticles to protect SH-SY5Y human neuroblastoma cells from oxidative/nitrosative stress induced by iron-nitrosyl complex sodium nitroprusside (SNP). GQD reduced SNP cytotoxicity by preventing mitochondrial depolarization, caspase-2 activation, and subsequent apoptotic death. Although GQD diminished the levels of nitric oxide (NO) in SNP-exposed cells, NO scavengers displayed only a slight protective effect, suggesting that NO quenching was not the main protective mechanism of GQD. GQD also reduced SNP-triggered increase in the intracellular levels of hydroxyl radical (•OH), superoxide anion (O2•-), and lipid peroxidation. Nonselective antioxidants, •OH scavenging, and iron chelators, but not superoxide dismutase, mimicked GQD cytoprotective activity, indicating that GQD protect cells by neutralizing •OH generated in the presence of SNP-released iron. Cellular internalization of GQD was required for optimal protection, since a removal of extracellular GQD by extensive washing only partly diminished their protective effect. Moreover, GQD cooperated with SNP to induce autophagy, as confirmed by the inhibition of autophagy-limiting Akt/PRAS40/mTOR signaling and increase in autophagy gene transcription, protein levels of proautophagic beclin-1 and LC3-II, formation of autophagic vesicles, and degradation of autophagic target p62. The antioxidant activity of GQD was not involved in autophagy induction, as antioxidants N-acetylcysteine and dimethyl sulfoxide failed to stimulate autophagy in SNP-exposed cells. Pharmacological inhibitors of early (wortmannin, 3-methyladenine) or late stages of autophagy (NH4Cl) efficiently reduced the protective effect of GQD. Therefore, the ability of GQD to prevent the in vitro neurotoxicity of SNP depends on both •OH/NO scavenging and induction of cytoprotective autophagy.
Collapse
Affiliation(s)
- Matija Krunić
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000, Belgrade, Serbia
| | - Biljana Ristić
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000, Belgrade, Serbia
| | - Mihajlo Bošnjak
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000, Belgrade, Serbia
| | - Verica Paunović
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000, Belgrade, Serbia
| | - Gordana Tovilović-Kovačević
- Department of Biochemistry, Institute for Biological Research, "Siniša Stanković"- National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000, Belgrade, Serbia
| | - Nevena Zogović
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000, Belgrade, Serbia
| | - Aleksandar Mirčić
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000, Belgrade, Serbia
| | - Zoran Marković
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade P.O. Box 522, 11000, Belgrade, Serbia
| | - Biljana Todorović-Marković
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade P.O. Box 522, 11000, Belgrade, Serbia
| | - Svetlana Jovanović
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade P.O. Box 522, 11000, Belgrade, Serbia
| | - Duška Kleut
- Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade P.O. Box 522, 11000, Belgrade, Serbia
| | - Miloš Mojović
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11000, Belgrade, Serbia
| | - Đura Nakarada
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11000, Belgrade, Serbia
| | - Olivera Marković
- Department of Chemistry, Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000, Belgrade, Serbia
| | - Irena Vuković
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000, Belgrade, Serbia
| | - Ljubica Harhaji-Trajković
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000, Belgrade, Serbia.
| | - Vladimir Trajković
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000, Belgrade, Serbia.
| |
Collapse
|
13
|
Hoshiko T, Kubota Y, Onodera R, Higashi T, Yokoo M, Motoyama K, Kimura S. Folic Acid-Appended Hydroxypropyl-β-Cyclodextrin Exhibits Potent Antitumor Activity in Chronic Myeloid Leukemia Cells via Autophagic Cell Death. Cancers (Basel) 2021; 13:cancers13215413. [PMID: 34771576 PMCID: PMC8582559 DOI: 10.3390/cancers13215413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
Simple Summary 2-Hydroxypropyl-β-cyclodextrin (HP-β-CyD) is a cyclic oligosaccharide widely used as an excipient in pharmaceutical preparations, in addition to also being used as a cholesterol regulator. HP-β-CyD was used in clinical trials for patients with Niemann-Pick Type C disease to remove accumulated intracellular lipid. HP-β-CyD has anti-leukemia activity by inducing apoptosis and cell-cycle arrest; however, the antitumor activity of HP-β-CyD lacks tumor cell-selectivity. Taking advantage of the fact that folate receptors are highly expressed in many cancer cells, we synthesized folate-appended HP-β-CyD (FA-HP-β-CyD) to confer tumor cell-selectivity to HP-β-CyD. FA-HP-β-CyD inhibited the proliferation of chronic myeloid leukemia (CML) cells and the mechanism underlying the effect of FA-HP-β-CyD in inducing cell death may involve autophagy. The combination of FA-HP-β-CyD and ABL tyrosine kinase inhibitors (imatinib and ponatinib) had a synergistic inhibitory effect on CML cells. In a mouse model of BCR-ABL-induced leukemia, FA-HP-β-CyD had a stronger inhibitory effect on leukemia progression than HP-β-CyD or imatinib. Abstract 2-Hydroxypropyl-β-cyclodextrin (HP-β-CyD) is widely used as an enabling excipient in pharmaceutical formulations. We previously demonstrated that HP-β-CyD disrupted cholesterol homeostasis, and inhibited the proliferation of leukemia cells by inducing apoptosis and cell-cycle arrest. Recently developed drug delivery systems using folic acid (FA) and folic acid receptors (FR) are currently being used in cancer treatment. To confer tumor cell-selectivity to HP-β-CyD, we synthesized folate-appended HP-β-CyD (FA-HP-β-CyD) and evaluated the potential of FA-HP-β-CyD as an anticancer agent using chronic myeloid leukemia (CML) cells in vitro and in vivo. FA-HP-β-CyD inhibited the growth of FR-expressing cells but not that of FR-negative cells. FA-HP-β-CyD had stronger anti-leukemia and cell-binding activities than HP-β-CyD in CML cells. Unlike HP-β-CyD, FA-HP-β-CyD entered CML cells through endocytosis and induced both apoptosis and autophagy via mitophagy. FA-HP-β-CyD increased the inhibitory effects of the ABL tyrosine kinase inhibitors imatinib mesylate and ponatinib, which are commonly used in CML. In vivo experiments in a BCR-ABL leukemia mouse model showed that FA-HP-β-CyD was more effective than HP-β-CyD at a ten-fold lower dose. These results indicate that FA-HP-β-CyD may be a novel tumor-targeting agent for the treatment of leukemia.
Collapse
Affiliation(s)
- Toshimi Hoshiko
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan; (T.H.); (Y.K.)
| | - Yasushi Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan; (T.H.); (Y.K.)
- Saitama Medical Center, Department of Transfusion Medicine and Cell Therapy, Saitama Medical University, Kawagoe 350-8550, Japan
| | - Risako Onodera
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (R.O.); (T.H.); (K.M.)
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (R.O.); (T.H.); (K.M.)
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto 862-0973, Japan
| | - Masako Yokoo
- Saga Medical Center Koseikan, Department of Hematology, Saga 849-8571, Japan;
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (R.O.); (T.H.); (K.M.)
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan; (T.H.); (Y.K.)
- Correspondence: ; Tel.: +81-952-34-2353; Fax: +81-952-34-2017
| |
Collapse
|
14
|
Poillet-Perez L, Sarry JE, Joffre C. Autophagy is a major metabolic regulator involved in cancer therapy resistance. Cell Rep 2021; 36:109528. [PMID: 34407408 DOI: 10.1016/j.celrep.2021.109528] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/13/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy sustains cellular homeostasis and metabolism in numerous diseases. By regulating cancer metabolism, both tumor and microenvironmental autophagy promote tumor growth. However, autophagy can support cancer progression through other biological functions such as immune response regulation or cytokine/growth factor secretion. Moreover, autophagy is induced in numerous tumor types as a resistance mechanism following therapy, highlighting autophagy inhibition as a promising target for anti-cancer therapy. Thus, better understanding the mechanisms involved in tumor growth and resistance regulation through autophagy, which are not fully understood, will provide insights into patient treatment.
Collapse
Affiliation(s)
- Laura Poillet-Perez
- Cancer Research Centre of Toulouse, UMR1037 Inserm, UMR5077 CNRS, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, 31037 Toulouse, France.
| | - Jean-Emmanuel Sarry
- Cancer Research Centre of Toulouse, UMR1037 Inserm, UMR5077 CNRS, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, 31037 Toulouse, France
| | - Carine Joffre
- Cancer Research Centre of Toulouse, UMR1037 Inserm, UMR5077 CNRS, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, 31037 Toulouse, France.
| |
Collapse
|
15
|
Autophagy a Close Relative of AML Biology. BIOLOGY 2021; 10:biology10060552. [PMID: 34207482 PMCID: PMC8235674 DOI: 10.3390/biology10060552] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Despite a high rate of complete remission following conventional chemotherapy, the prognosis remains poor due to frequent relapses caused by relapse-initiating leukemic cells (RICs), which are resistant to chemotherapies. While the development of new targeted therapies holds great promise (e.g., molecules targeting IDH1/2, FLT3, BCL2), relapses still occur. Therefore, a paramount issue in the elimination of RICs is to decipher the AML resistance mechanisms. Thus, it has been recently shown that AML cells exhibit metabolic changes in response to chemotherapy or targeted therapies. Autophagy is a major regulator of cell metabolism, involved in maintaining cancer state, metastasis, and resistance to anticancer therapy. However, whether autophagy acts as a tumor suppressor or promoter in AML is still a matter of debate. Therefore, depending on molecular AML subtypes or treatments used, a better understanding of the role of autophagy is needed to determine whether its modulation could result in a clinical benefit. Abstract Autophagy, which literally means “eat yourself”, is more than just a lysosomal degradation pathway. It is a well-known regulator of cellular metabolism and a mechanism implicated in tumor initiation/progression and therapeutic resistance in many cancers. However, whether autophagy acts as a tumor suppressor or promoter is still a matter of debate. In acute myeloid leukemia (AML), it is now proven that autophagy supports cell proliferation in vitro and leukemic progression in vivo. Mitophagy, the specific degradation of mitochondria through autophagy, was recently shown to be required for leukemic stem cell functions and survival, highlighting the prominent role of this selective autophagy in leukemia initiation and progression. Moreover, autophagy in AML sustains fatty acid oxidation through lipophagy to support mitochondrial oxidative phosphorylation (OxPHOS), a hallmark of chemotherapy-resistant cells. Nevertheless, in the context of therapy, in AML, as well as in other cancers, autophagy could be either cytoprotective or cytotoxic, depending on the drugs used. This review summarizes the recent findings that mechanistically show how autophagy favors leukemic transformation of normal hematopoietic stem cells, as well as AML progression and also recapitulates its ambivalent role in resistance to chemotherapies and targeted therapies.
Collapse
|
16
|
Kim DY, Nam J, Chung JS, Kim SW, Shin HJ. Role of Roflumilast Combined with ESHAP Chemotherapy in Relapsed/Refractory Patients with Diffuse Large B-Cell Lymphoma. Cancer Res Treat 2021; 54:301-313. [PMID: 33940789 PMCID: PMC8756117 DOI: 10.4143/crt.2020.1371] [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: 12/28/2020] [Accepted: 04/26/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose There are unmet needs associated with the current treatment strategies for relapsed/refractory diffuse large B-cell lymphoma (DLBCL) due to the poor treatment outcomes of these strategies. Roflumilast, a selective phosphodiesterase-4 inhibitor used for treating chronic obstructive pulmonary disease, is effective against B-cell malignancy via phosphoinositide 3-kinase (PI3K)–activity suppression. We analyzed the effects of roflumilast combined with ESHAP (etoposide, cisplatin, methylprednisolone, and cytarabine) chemotherapy in experimental and clinical settings. Materials and Methods An in vitro study using lymphoma cell lines and a pilot study on relapsed/refractory DLBCL patients were conducted to investigate the effects and mechanism of the combination of roflumilast and chemotherapy. The complete response (CR), overall response rate (ORR), and 1-year progression-free survival (PFS) were analyzed. Results We found that roflumilast is efficient when combined with other chemotherapy drugs, especially cytarabine. Synergistic effects between these two drugs influence the translation of mammalian target of rapamycin and myeloid cell leukemia 1, resulting in apoptosis and inhibition of B-cell lymphoma proliferation. In clinical setting, the roflumilast group showed better rates of CR (46.2% vs. 34.6%), ORR (76.9% vs. 53.8%), and 1-year PFS (50.0% vs. 25.9%) compared with the control group, though not statistically significant. The roflumilast group showed a higher incidence of asthenia and gastrointestinal adverse events. However, grade 3 or 4 adverse events were similar in both groups. Conclusion We found that roflumilast, when combined with ESHAP chemotherapy, for relapsed/refractory DLBCL was clinically active and well tolerated. This combined treatment was able to suppress PI3K activity, which is correlated with the degree of clinical response.
Collapse
Affiliation(s)
- Do Young Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Biochemical Research Institution, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Jehyun Nam
- Department of Biological Sciences, Pusan National University, Busan, Korea
| | - Joo-Seop Chung
- Division of Hematology-Oncology, Department of Internal Medicine, Biochemical Research Institution, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Sang-Woo Kim
- Department of Biological Sciences, Pusan National University, Busan, Korea
| | - Ho-Jin Shin
- Division of Hematology-Oncology, Department of Internal Medicine, Biochemical Research Institution, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| |
Collapse
|
17
|
Panina SB, Pei J, Kirienko NV. Mitochondrial metabolism as a target for acute myeloid leukemia treatment. Cancer Metab 2021; 9:17. [PMID: 33883040 PMCID: PMC8058979 DOI: 10.1186/s40170-021-00253-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemias (AML) are a group of aggressive hematologic malignancies resulting from acquired genetic mutations in hematopoietic stem cells that affect patients of all ages. Despite decades of research, standard chemotherapy still remains ineffective for some AML subtypes and is often inappropriate for older patients or those with comorbidities. Recently, a number of studies have identified unique mitochondrial alterations that lead to metabolic vulnerabilities in AML cells that may present viable treatment targets. These include mtDNA, dependency on oxidative phosphorylation, mitochondrial metabolism, and pro-survival signaling, as well as reactive oxygen species generation and mitochondrial dynamics. Moreover, some mitochondria-targeting chemotherapeutics and their combinations with other compounds have been FDA-approved for AML treatment. Here, we review recent studies that illuminate the effects of drugs and synergistic drug combinations that target diverse biomolecules and metabolic pathways related to mitochondria and their promise in experimental studies, clinical trials, and existing chemotherapeutic regimens.
Collapse
Affiliation(s)
| | - Jingqi Pei
- Department of BioSciences, Rice University, Houston, TX, USA
| | | |
Collapse
|
18
|
Zhang H, Liu L, Chen L, Liu H, Ren S, Tao Y. Long noncoding RNA DANCR confers cytarabine resistance in acute myeloid leukemia by activating autophagy via the miR-874-3P/ATG16L1 axis. Mol Oncol 2021; 15:1203-1216. [PMID: 33638615 PMCID: PMC8024725 DOI: 10.1002/1878-0261.12661] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/01/2019] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an important mechanism involved in the regulation of acute myeloid leukemia (AML) chemoresistance. The long noncoding RNA (lncRNA) differentiation antagonizing non‐protein coding RNA (DANCR) exhibits oncogenic activity in several types of human cancers, including AML, but it remains unclear whether it regulates autophagy and chemoresistance in AML. We report here that cytarabine (Ara‐C) treatment elevates DANCR expression in human AML cells. In addition, DANCR overexpression confers and its knockdown diminishes Ara‐C resistance in human AML cells, suggesting that DANCR positively regulates AML chemoresistance to Ara‐C. Moreover, DANCR promotes autophagy in Ara‐C‐treated human AML cells and acts as a sponge to decrease miR‐20a‐5p expression, thereby upregulating the expression of ATG16L1, a critical component of the autophagy machinery. Importantly, ATG16L1 silencing abrogates DANCR‐promoted autophagy and markedly restores DANCR‐conferred Ara‐C resistance, suggesting that DANCR promotes MIR‐874‐3P/ATG16L1 axis‐regulated autophagy to confer Ara‐C resistance in human AML cells. Together, this study identifies DANCR as a positive regulator of Ara‐C resistance in human AML cells, suggesting this lncRNA as a potential target for overcoming Ara‐C resistance in AML chemotherapy.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, China
| | - Ling Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, China
| | - Lulu Chen
- Graduate School, Jining Medical University, China
| | - Haihui Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, China
| | - Saisai Ren
- Department of Hematology, Affiliated Hospital of Jining Medical University, China
| | - Yanling Tao
- Department of Pediatric Hematology, Affiliated Hospital of Jining Medical University, China
| |
Collapse
|
19
|
Visser N, Lourens HJ, Huls G, Bremer E, Wiersma VR. Inhibition of Autophagy Does Not Re-Sensitize Acute Myeloid Leukemia Cells Resistant to Cytarabine. Int J Mol Sci 2021; 22:ijms22052337. [PMID: 33652766 PMCID: PMC7956277 DOI: 10.3390/ijms22052337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Elevated activation of the autophagy pathway is currently thought to be one of the survival mechanisms allowing therapy-resistant cancer cells to escape elimination, including for cytarabine (AraC)-resistant acute myeloid leukemia (AML) patients. Consequently, the use of autophagy inhibitors such as chloroquine (CQ) is being explored for the re-sensitization of AraC-resistant cells. In our study, no difference in the activity of the autophagy pathway was detected when comparing AraC-Res AML cell lines to parental AraC-sensitive AML cell lines. Furthermore, treatment with autophagy inhibitors CQ, 3-Methyladenine (3-MA), and bafilomycin A1 (BafA1) did not re-sensitize AraC-Res AML cell lines to AraC treatment. However, in parental AraC-sensitive AML cells, treatment with AraC did activate autophagy and, correspondingly, combination of AraC with autophagy inhibitors strongly reduced cell viability. Notably, the combination of these drugs also yielded the highest level of cell death in a panel of patient-derived AML samples even though not being additive. Furthermore, there was no difference in the cytotoxic effect of autophagy inhibition during AraC treatment in matched de novo and relapse samples with differential sensitivity to AraC. Thus, inhibition of autophagy may improve AraC efficacy in AML patients, but does not seem warranted for the treatment of AML patients that have relapsed with AraC-resistant disease.
Collapse
|
20
|
Lu Y, Zhou J, Pei R, Li F, Jin J, Jiang L. Expression and clinical significance of phospholipase D1 in de novo acute myeloid leukemia. ACTA ACUST UNITED AC 2020; 25:270-275. [PMID: 32615908 DOI: 10.1080/16078454.2020.1786971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Objective: Phospholipase D (PLD) is known to participate in several aspects of cellular processes including cell mitosis, migration, phagocytosis, and membrane vesicle trafficking. The role of PLD has been investigated in multiple cancers except hematologic malignances. Methods: We enrolled 291 patients with de novo acute myeloid leukemia (AML) and detected PLD1 mRNA expression levels of their bone marrow samples by quantitative real-time PCR (qRT-PCR). Clinical relevance of PLD1 was assessed using Kaplan-Meier analysis, univariate analysis, and multivariate analysis. Results: Compared to healthy donors, AML patients had higher mRNA levels of PLD1, which was significantly associated with FAB classification. Importantly, patients with low levels of PLD1 had impaired overall survival and event-free survival. Moreover, univariate and multivariate analyses confirmed that PLD1 expression was an independent prognostic factor for AML. Conclusion: PLD1 represented a useful diagnostic marker and prognostic factor for AML.
Collapse
Affiliation(s)
- Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, People's Republic of China.,Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, People's Republic of China
| | - Jiasi Zhou
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, People's Republic of China.,Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, People's Republic of China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, People's Republic of China.,Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, People's Republic of China
| | - Fenglin Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang University, Hangzhou, People's Republic of China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Zhejiang University, Hangzhou, People's Republic of China
| | - Lei Jiang
- Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, People's Republic of China
| |
Collapse
|
21
|
Silva VR, Neves SP, Santos LDS, Dias RB, Bezerra DP. Challenges and Therapeutic Opportunities of Autophagy in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12113461. [PMID: 33233671 PMCID: PMC7699739 DOI: 10.3390/cancers12113461] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Autophagy is a physiological process characterized by the degradation of the cell components through lysosomes due to stimuli/stress. In this study, we review the challenges and therapeutic opportunities that autophagy presents in the treatment of cancer. We discussed the results of several studies that evaluated autophagy as a therapeutic strategy in cancer, both through the modulation of therapeutic resistance and the death of cancer cells. Moreover, we discussed the role of autophagy in the biology of cancer stem cells and the inhibition of this process as a strategy to overcome resistance and progression of cancer stem cells. Abstract Autophagy is a physiological cellular process that is crucial for development and can occurs in response to nutrient deprivation or metabolic disorders. Interestingly, autophagy plays a dual role in cancer cells—while in some situations, it has a cytoprotective effect that causes chemotherapy resistance, in others, it has a cytotoxic effect in which some compounds induce autophagy-mediated cell death. In this review, we summarize strategies aimed at autophagy for the treatment of cancer, including studies of drugs that can modulate autophagy-mediated resistance, and/or drugs that cause autophagy-mediated cancer cell death. In addition, the role of autophagy in the biology of cancer stem cells has also been discussed.
Collapse
|
22
|
Antileukemic activity of the VPS34-IN1 inhibitor in acute myeloid leukemia. Oncogenesis 2020; 9:94. [PMID: 33093450 PMCID: PMC7581748 DOI: 10.1038/s41389-020-00278-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive disease with a poor prognosis. Vacuolar protein sorting 34 (VPS34) is a member of the phosphatidylinositol-3-kinase lipid kinase family that controls the canonical autophagy pathway and vesicular trafficking. Using a recently developed specific inhibitor (VPS34-IN1), we found that VPS34 inhibition induces apoptosis in AML cells but not in normal CD34+ hematopoietic cells. Complete and acute inhibition of VPS34 was required for the antileukemic activity of VPS34-IN1. This inhibitor also has pleiotropic effects against various cellular functions related to class III PI3K in AML cells that may explain their survival impairment. VPS34-IN1 inhibits basal and L-asparaginase-induced autophagy in AML cells. A synergistic cell death activity of this drug was also demonstrated. VPS34-IN1 was additionally found to impair vesicular trafficking and mTORC1 signaling. From an unbiased approach based on phosphoproteomic analysis, we identified that VPS34-IN1 specifically inhibits STAT5 phosphorylation downstream of FLT3-ITD signaling in AML. The identification of the mechanisms controlling FLT3-ITD signaling by VPS34 represents an important insight into the oncogenesis of AML and could lead to new therapeutic strategies.
Collapse
|
23
|
Zhang H, Kang J, Liu L, Chen L, Ren S, Tao Y. MicroRNA-143 sensitizes acute myeloid leukemia cells to cytarabine via targeting ATG7- and ATG2B-dependent autophagy. Aging (Albany NY) 2020; 12:20111-20126. [PMID: 33077697 PMCID: PMC7655171 DOI: 10.18632/aging.103614] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/24/2020] [Indexed: 02/06/2023]
Abstract
Targeting autophagy holds promise to enhance chemosensitivity in acute myeloid leukemia (AML). MicroRNA-143 (miR-143) has been found to suppress autophagy, however, it is not clear whether miR-143 augments cytarabine cytotoxicity in AML. Here, we report that cytarabine treatment reduces miR-143 expression in AML cell lines and primary AML cells. Moreover, ectopic expression of miR-143 further decreases cell viability in cytarabine-treated AML cells. By contrast, miR-143 knockdown inhibits cytarabine-induced cytotoxicity, together indicating a role of miR-143 in enhancing cytarabine sensitivity in AML. Subsequently, we show that miR-143 inhibits autophagy in cytarabine-treated AML cells by directly targeting autophagy-related proteins (ATG), ATG7 and ATG2B, two critical known components of autophagic machinery. More importantly, autophagy reconstructed via co-expression of ATG7 and ATG2B substantially attenuates miR-143-enhanced cytotoxicity, which is associated with suppression of caspase-dependent apoptotic pathway. Overall, this study demonstrates that targeting ATG7 and ATG2B-dependent autophagy is a critical mechanism by which miR-143 sensitizes AML to cytarabine, implicating it as a potential therapeutic target in AML treatment.
Collapse
MESH Headings
- Antimetabolites, Antineoplastic/pharmacology
- Apoptosis/drug effects
- Autophagy/drug effects
- Autophagy-Related Protein 7/genetics
- Autophagy-Related Protein 7/metabolism
- Autophagy-Related Proteins/genetics
- Autophagy-Related Proteins/metabolism
- Caspase 3/metabolism
- Caspase 9/metabolism
- Cytarabine/pharmacology
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Leukemic
- HL-60 Cells
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Signal Transduction
- U937 Cells
- Vesicular Transport Proteins/genetics
- Vesicular Transport Proteins/metabolism
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Jianmin Kang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Ling Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Lulu Chen
- Graduate School, Jining Medical University, Jining 272000, Shandong Province, China
| | - Saisai Ren
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| | - Yanling Tao
- Department of Pediatric Hematology, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong Province, China
| |
Collapse
|
24
|
Liu T, Zhang J, Li K, Deng L, Wang H. Combination of an Autophagy Inducer and an Autophagy Inhibitor: A Smarter Strategy Emerging in Cancer Therapy. Front Pharmacol 2020; 11:408. [PMID: 32322202 PMCID: PMC7156970 DOI: 10.3389/fphar.2020.00408] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
Autophagy is considered a cytoprotective function in cancer therapy under certain conditions and is a drug resistance mechanism that represents a clinical obstacle to successful cancer treatment and leads to poor prognosis in cancer patients. Because certain clinical drugs and agents in development have cytoprotective autophagy effects, targeting autophagic pathways has emerged as a potential smarter strategy for cancer therapy. Multiple preclinical and clinical studies have demonstrated that autophagy inhibition augments the efficacy of anticancer agents in various cancers. Autophagy inhibitors, such as chloroquine and hydroxychloroquine, have already been clinically approved, promoting drug combination treatment by targeting autophagic pathways as a means of discovering and developing more novel and more effective cancer therapeutic approaches. We summarize current studies that focus on the antitumor efficiency of agents that induce cytoprotective autophagy combined with autophagy inhibitors. Furthermore, we discuss the challenge and development of targeting cytoprotective autophagy as a cancer therapeutic approach in clinical application. Thus, we need to facilitate the exploitation of appropriate autophagy inhibitors and coadministration delivery system to cooperate with anticancer drugs. This review aims to note optimal combination strategies by modulating autophagy for therapeutic advantage to overcome drug resistance and enhance the effect of antitumor therapies on cancer patients.
Collapse
Affiliation(s)
- Ting Liu
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhang
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kangdi Li
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Lingnan Deng
- Department of Digestion, The Second Affiliated Hospital of Jiangxi University TCM, Nanchang, China
| | - Hongxiang Wang
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Putyrski M, Vakhrusheva O, Bonn F, Guntur S, Vorobyov A, Brandts C, Dikic I, Ernst A. Disrupting the LC3 Interaction Region (LIR) Binding of Selective Autophagy Receptors Sensitizes AML Cell Lines to Cytarabine. Front Cell Dev Biol 2020; 8:208. [PMID: 32296703 PMCID: PMC7137635 DOI: 10.3389/fcell.2020.00208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
Short linear motifs (SLiMs) located in disordered regions of multidomain proteins are important for the organization of protein-protein interaction networks. By dynamic association with their binding partners, SLiMs enable assembly of multiprotein complexes, pivotal for the regulation of various aspects of cell biology in higher organisms. Despite their importance, there is a paucity of molecular tools to study SLiMs of endogenous proteins in live cells. LC3 interacting regions (LIRs), being quintessential for orchestrating diverse stages of autophagy, are a prominent example of SLiMs and mediate binding to the ubiquitin-like LC3/GABARAP family of proteins. The role of LIRs ranges from the posttranslational processing of their binding partners at early stages of autophagy to the binding of selective autophagy receptors (SARs) to the autophagosome. In order to generate tools to study LIRs in cells, we engineered high affinity binders of LIR motifs of three archetypical SARs: OPTN, p62, and NDP52. In an array of in vitro and cellular assays, the engineered binders were shown to have greatly improved affinity and specificity when compared with the endogenous LC3/GABARAP family of proteins, thus providing a unique possibility for modulating LIR interactions in living systems. We exploited these novel tools to study the impact of LIR inhibition on the fitness and the responsiveness to cytarabine treatment of THP-1 cells - a model for studying acute myeloid leukemia (AML). Our results demonstrate that inhibition of LIR of a single autophagy receptor is insufficient to sensitize the cells to cytarabine, while simultaneous inhibition of three LIR motifs in three distinct SARs reduces the IC50 of the chemotherapeutic.
Collapse
Affiliation(s)
- Mateusz Putyrski
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany
| | - Olesya Vakhrusheva
- Department of Medicine, Hematology/Oncology, Goethe-University, Frankfurt, Germany
| | - Florian Bonn
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Suchithra Guntur
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Andrew Vorobyov
- Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany
| | - Christian Brandts
- Department of Medicine, Hematology/Oncology, Goethe-University, Frankfurt, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,University Cancer Center Frankfurt, Goethe-University, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences, Frankfurt, Germany
| | - Andreas Ernst
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany
| |
Collapse
|
26
|
Humbert M, Morán M, de la Cruz-Ojeda P, Muntané J, Wiedmer T, Apostolova N, McKenna SL, Velasco G, Balduini W, Eckhart L, Janji B, Sampaio-Marques B, Ludovico P, Žerovnik E, Langer R, Perren A, Engedal N, Tschan MP. Assessing Autophagy in Archived Tissue or How to Capture Autophagic Flux from a Tissue Snapshot. BIOLOGY 2020; 9:E59. [PMID: 32245178 PMCID: PMC7150830 DOI: 10.3390/biology9030059] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/14/2022]
Abstract
Autophagy is a highly conserved degradation mechanism that is essential for maintaining cellular homeostasis. In human disease, autophagy pathways are frequently deregulated and there is immense interest in targeting autophagy for therapeutic approaches. Accordingly, there is a need to determine autophagic activity in human tissues, an endeavor that is hampered by the fact that autophagy is characterized by the flux of substrates whereas histology informs only about amounts and localization of substrates and regulators at a single timepoint. Despite this challenging task, considerable progress in establishing markers of autophagy has been made in recent years. The importance of establishing clear-cut autophagy markers that can be used for tissue analysis cannot be underestimated. In this review, we attempt to summarize known techniques to quantify autophagy in human tissue and their drawbacks. Furthermore, we provide some recommendations that should be taken into consideration to improve the reliability and the interpretation of autophagy biomarkers in human tissue samples.
Collapse
Grants
- none Bernese Cancer League
- none Stiftung für klinisch-experimentelle Tumorforschung
- none Werner and Hedy Berger-Janser Foundation for Cancer Research
- PI14/01085 and PI17/00093 FIS and FEDER funds from the EU
- CPII16/00023 ISCIII and FSE funds
- RTI2018-096748-B-100 the Spanish Minsitry of Science, Innovation and Universities
- none University Professor Training Fellowship, Ministry of Science, Innovation and University, Government of Spain
- PI18/00442 the State Plan for R & D + I2013-2016 and funded by the Instituto de Salud Carlos III
- none European Regional Development Fund
- C18/BM/12670304/COMBATIC Luxembourg National Research Fund
- NORTE-01-0145-FEDER-000013 Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, by the European Regional Development Fund (FEDER), through the Competitiveness Factors Operational Programme (COMPETE)
- POCI-01-0145-FEDER-028159 and POCI-01-0145-FEDER-030782 FEDER, through the COMPETE
- none National funds, through the Foundation for Science and Technology (FCT
- none ARRS - the Slovenian research agency, programme P1-0140: Proteolysis and its regulation
- KFS-3360-02-2014 the Swiss Cancer Research
- KFS-3409-02-2014 the Swiss Cancer Research
- 31003A_173219 Swiss National Science Foundation
Collapse
Affiliation(s)
- Magali Humbert
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - María Morán
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital ‘12 de Octubre’ (‘imas12’), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain;
- Department of Surgery, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Jordi Muntané
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain;
- Department of Surgery, School of Medicine, University of Seville, 41009 Seville, Spain
- Spanish Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Tabea Wiedmer
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - Nadezda Apostolova
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Spanish Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
| | - Sharon L. McKenna
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Cancer Research at UCC, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Guillermo Velasco
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, and Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Walter Balduini
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Leopold Eckhart
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Bassam Janji
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology—Luxembourg Institute of Health, 1526 Luxembourg City, Luxembourg
| | - Belém Sampaio-Marques
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paula Ludovico
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eva Žerovnik
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
| | - Rupert Langer
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - Aurel Perren
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - Nikolai Engedal
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway
| | - Mario P. Tschan
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| |
Collapse
|
27
|
Qiu L, Zhou G, Cao S. Targeted inhibition of ULK1 enhances daunorubicin sensitivity in acute myeloid leukemia. Life Sci 2019; 243:117234. [PMID: 31887299 DOI: 10.1016/j.lfs.2019.117234] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE In acute myeloid leukemia (AML), complete remission can be achieved in parts of patients using cytarabine/anthracycline combination-based chemotherapy, however, drug resistance-related recurrence is still a common cause of treatment failure, leading to high mortality among patients. In our research, we revealed the molecular mechanisms that were sufficient to improve sensitivity of AML cells to the anthracycline daunorubicin (DNR). METHODS We evaluated the effects of autophagy and apoptosis induced by DNR using two AML cell lines HL60 and U937.Western blot was preformed to analyze the apoptotic pathway protein expression and flow cytometric analysis was used to detect the level of apoptosis in AML cells. The levels of autophagy-related proteins were detected by western blotting and autophagic vesicles were observed by electron microscopy. RESULTS DNR effectively induced autophagy in two AML cell lines HL60 and U937 confirming by upregulation of LC3-II lipidation, formation of autophagosomes. Inhibition of autophagy by pharmacologic inhibitor HCQ promoted apoptosis induced by DNR, suggesting that autophagy played a vital role in pro-survival in AML. Furthermore, ULK1 inhibition by a highly selective kinase inhibitor SBI-0206965 and shRNA enhanced cytotoxicity of DNR against AML cells. Independent of mTOR -ULK1 signaling pathway, activation of autophagy of DNR was proved to be mediated by AMPK (pThr172)/ULK1 pathway. CONCLUSIONS These results revealed that pro-survival autophagy induced by ULK1 activation was one of the potential mechanisms of AML resistance to DNR. Targeting ULK1 selectively could be a promising therapeutic strategy to enhance sensitivity of DNR for AML therapy.
Collapse
Affiliation(s)
- Li Qiu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan 410078, China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan 410078, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiang Ya Road, Changsha, Hunan 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, China
| | - Shan Cao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan 410078, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiang Ya Road, Changsha, Hunan 410078, China.
| |
Collapse
|
28
|
Stamenkovic M, Janjetovic K, Paunovic V, Ciric D, Kravic-Stevovic T, Trajkovic V. Comparative analysis of cell death mechanisms induced by lysosomal autophagy inhibitors. Eur J Pharmacol 2019; 859:172540. [DOI: 10.1016/j.ejphar.2019.172540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/18/2019] [Accepted: 07/12/2019] [Indexed: 12/21/2022]
|
29
|
Castro I, Sampaio-Marques B, Ludovico P. Targeting Metabolic Reprogramming in Acute Myeloid Leukemia. Cells 2019; 8:cells8090967. [PMID: 31450562 PMCID: PMC6770240 DOI: 10.3390/cells8090967] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
The cancer metabolic reprogramming allows the maintenance of tumor proliferation, expansion and survival by altering key bioenergetics, biosynthetic and redox functions to meet the higher demands of tumor cells. In addition, several metabolites are also needed to perform signaling functions that further promote tumor growth and progression. These metabolic alterations have been exploited in different cancers, including acute myeloid leukemia, as novel therapeutic strategies both in preclinical models and clinical trials. Here, we review the complexity of acute myeloid leukemia (AML) metabolism and discuss how therapies targeting different aspects of cellular metabolism have demonstrated efficacy and how they provide a therapeutic window that should be explored to target the metabolic requirements of AML cells.
Collapse
Affiliation(s)
- Isabel Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal.
| |
Collapse
|
30
|
Castelli G, Pelosi E, Testa U. Emerging Therapies for Acute Myelogenus Leukemia Patients Targeting Apoptosis and Mitochondrial Metabolism. Cancers (Basel) 2019; 11:E260. [PMID: 30813354 PMCID: PMC6406361 DOI: 10.3390/cancers11020260] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023] Open
Abstract
Acute Myelogenous Leukemia (AML) is a malignant disease of the hematopoietic cells, characterized by impaired differentiation and uncontrolled clonal expansion of myeloid progenitors/precursors, resulting in bone marrow failure and impaired normal hematopoiesis. AML comprises a heterogeneous group of malignancies, characterized by a combination of different somatic genetic abnormalities, some of which act as events driving leukemic development. Studies carried out in the last years have shown that AML cells invariably have abnormalities in one or more apoptotic pathways and have identified some components of the apoptotic pathway that can be targeted by specific drugs. Clinical results deriving from studies using B-cell lymphoma 2 (BCL-2) inhibitors in combination with standard AML agents, such as azacytidine, decitabine, low-dose cytarabine, provided promising results and strongly support the use of these agents in the treatment of AML patients, particularly of elderly patients. TNF-related apoptosis-inducing ligand (TRAIL) and its receptors are frequently deregulated in AML patients and their targeting may represent a promising strategy for development of new treatments. Altered mitochondrial metabolism is a common feature of AML cells, as supported through the discovery of mutations in the isocitrate dehydrogenase gene and in mitochondrial electron transport chain and of numerous abnormalities of oxidative metabolism existing in AML subgroups. Overall, these observations strongly support the view that the targeting of mitochondrial apoptotic or metabolic machinery is an appealing new therapeutic perspective in AML.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
31
|
Brendle C, Stefan N, Stef I, Ripkens S, Soekler M, la Fougère C, Nikolaou K, Pfannenberg C. Impact of diverse chemotherapeutic agents and external factors on activation of brown adipose tissue in a large patient collective. Sci Rep 2019; 9:1901. [PMID: 30760750 PMCID: PMC6374459 DOI: 10.1038/s41598-018-37924-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/18/2018] [Indexed: 11/21/2022] Open
Abstract
Increased activity of brown adipose tissue (BAT) activity in adults is thought to prevent obesity. Therefore, regulators of BAT activity might serve as anti-obesity therapy in future, but are not investigated thoroughly up to now. In our study, we assessed retrospectively the association of BAT activity with several external factors and diverse chemotherapeutic and immunosuppressive agents in a collective of 702 patients. The patients underwent at least two clinically indicated PET/CT examinations in the course of different oncological and inflammatory diseases. BAT activity was identified according to predefined PET/CT criteria in all examinations. In multivariate analysis, the type of disease, the disease activity and the therapeutic regimen did not influence BAT activity. In contrast, sex and age were confirmed as independent factors for BAT activity. For the association of therapeutic agents with BAT activity, we examined 53 different disease-related agents, which were applied to patients without initial BAT activity between their PET/CT examinations. Out of these, cytarabine therapy was significantly associated with increased new onset of BAT activity. Cytarabine is a therapeutic agent for lymphoma patients. Further targeted studies might investigate the usefulness of Cytarabine serving as possible therapeutic approach against obesity via BAT regulation.
Collapse
Affiliation(s)
- Cornelia Brendle
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany. .,Diagnostic and Interventional Neuroradiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany.
| | - Norbert Stefan
- Endocrinology and Diabetology, Department of Internal Medicine, Eberhard Karls University, Otfried-Mueller-Straße 10, 72076, Tuebingen, Germany
| | - Irina Stef
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| | - Sabine Ripkens
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| | - Martin Soekler
- Oncology, Hematology, Clinical Immunology, Rheumatology and Pulmology, Department of Internal Medicine, Eberhard Karls University, Otfried-Mueller-Straße 10, 72076, Tuebingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University, Otfried-Mueller-Straße 14, 72076, Tuebingen, Germany
| | - Konstantin Nikolaou
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| | - Christina Pfannenberg
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany
| |
Collapse
|
32
|
Current Outlook on Autophagy in Human Leukemia: Foe in Cancer Stem Cells and Drug Resistance, Friend in New Therapeutic Interventions. Int J Mol Sci 2019; 20:ijms20030461. [PMID: 30678185 PMCID: PMC6387281 DOI: 10.3390/ijms20030461] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 01/07/2023] Open
Abstract
Autophagy is an evolutionarily conserved cellular recycling process in cell homeostasis and stress adaptation. It confers protection and promotes survival in response to metabolic/environmental stress, and is upregulated in response to nutrient deprivation, hypoxia, and chemotherapies. Autophagy is also known to sustain malignant cell growth and contributes to cancer stem cell survival when challenged by cytotoxic and/or targeted therapies, a potential mechanism of disease persistence and drug resistance that has gathered momentum. However, different types of human leukemia utilize autophagy in complex, context-specific manners, and the molecular and cellular mechanisms underlying this process involve multiple protein networks that will be discussed in this review. There is mounting preclinical evidence that targeting autophagy can enhance the efficacy of cancer therapies. Chloroquine and other lysosomal inhibitors have spurred initiation of clinical trials and demonstrated that inhibition of autophagy restores chemosensitivity of anticancer drugs, but with limited autophagy-dependent effects. Intriguingly, several autophagy-specific inhibitors, with better therapeutic indexes and lower toxicity, have been developed. Promising preclinical studies with novel combination approaches as well as potential challenges to effectively eradicate drug-resistant cells, particularly cancer stem cells, in human leukemia are also detailed in this review.
Collapse
|
33
|
Kazemi A, Sadri M, Houshmand M, Yazdi N, Zarif MN, Anjam-Najmedini A, Tavakoli R, Ojaghi M, Ajami M, Ajami M, Atashi A. The anticancer effects of pharmacological inhibition of autophagy in acute erythroid leukemia cells. Anticancer Drugs 2018; 29:944-955. [DOI: 10.1097/cad.0000000000000668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
In vitro and in vivo antimelanoma effect of ethyl ester cyclohexyl analog of ethylenediamine dipropanoic acid. Melanoma Res 2018; 28:8-20. [PMID: 29135861 DOI: 10.1097/cmr.0000000000000409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melanoma, an aggressive skin tumor with high metastatic potential, is associated with high mortality and increasing morbidity. Multiple available chemotherapeutic and immunotherapeutic modalities failed to improve survival in advanced disease, and the search for new agents is ongoing. The aim of this study was to investigate antimelanoma effects of O,O-diethyl-(S,S)-ethylenediamine-N,N'di-2-(3-cyclohexyl) propanoate dihydrochloride (EE), a previously synthesized and characterized organic compound. Mouse melanoma B16 cell viability was assessed using acid phosphatase, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, sulforhodamine B, and lactate dehydrogenase assays. Apoptosis and autophagy were investigated using flow cytometry, fluorescence and electron microscopy, and western blotting. In vivo antitumor potential was assessed in subcutaneous mouse melanoma model after 14 days of treatment with EE. Tumor mass and volume were measured, and RT-PCR was used for investigating the expression of autophagy-related, proapoptotic, and antiapoptotic molecules in tumor tissue. Investigated organic compound exerts significant cytotoxic effect against B16 cells. EE induced apoptosis, as confirmed by phosphatidyl serine externalisation, caspase activation, and ultrastructural features typical for apoptosis seen on fluorescence and electron microscopes. The apoptotic mechanism included prompt disruption of mitochondrial membrane potential and oxidative stress. No autophagy was observed. Antimelanoma action and apoptosis induction were confirmed in vivo, as EE decreased mass and volume of tumors, and increased expression of several proapoptotic genes. EE possesses significant antimelanoma action and causes caspase-dependent apoptosis mediated by mitochondrial damage and reactive oxygen species production. Decrease in tumor growth and increase in expression of proapoptotic genes in tumor tissue suggest that EE warrants further investigation as a candidate agent in treating melanoma.
Collapse
|
35
|
Delou JMA, Biasoli D, Borges HL. The Complex Link between Apoptosis and Autophagy: a Promising New Role for RB. AN ACAD BRAS CIENC 2018; 88:2257-2275. [PMID: 27991962 DOI: 10.1590/0001-3765201620160127] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
Physiological processes, as autophagy, proliferation and apoptosis are affected during carcinogenesis. Restoring cellular sensitivity to apoptotic stimuli, such as the antineoplastic cocktails, has been explored as a strategy to eliminate cancer cells. Autophagy, a physiological process of recycling organelles and macromolecules can be deviated from homeostasis to support cancer cells survival, proliferation, escape from apoptosis, and therapy resistance. The relationship between autophagy and apoptosis is complex and many stimuli can induce both processes. Most chemotherapeutic agents induce autophagy and it is not clear whether and how this chemotherapy-induced autophagy might contribute to resistance to apoptosis. Here, we review current strategies to sensitize cancer cells by interfering with autophagy. Moreover, we discuss a new link between autophagy and apoptosis: the tumor suppressor retinoblastoma protein (RB). Inactivation of RB is one of the earliest and more frequent hallmarks of cancer transformation, known to control cell cycle progression and apoptosis. Therefore, understanding RB functions in controlling cell fate is essential for an effective translation of RB status in cancer samples to the clinical outcome.
Collapse
Affiliation(s)
- João M A Delou
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Ilha do Fundão, 21949-590 Rio de Janeiro, RJ, Brazil
| | - Deborah Biasoli
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Ilha do Fundão, 21949-590 Rio de Janeiro, RJ, Brazil
| | - Helena L Borges
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Ilha do Fundão, 21949-590 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
36
|
Ultimo S, Simioni C, Martelli AM, Zauli G, Evangelisti C, Celeghini C, McCubrey JA, Marisi G, Ulivi P, Capitani S, Neri LM. PI3K isoform inhibition associated with anti Bcr-Abl drugs shows in vitro increased anti-leukemic activity in Philadelphia chromosome-positive B-acute lymphoblastic leukemia cell lines. Oncotarget 2018; 8:23213-23227. [PMID: 28390196 PMCID: PMC5410298 DOI: 10.18632/oncotarget.15542] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/12/2017] [Indexed: 12/30/2022] Open
Abstract
B-acute lymphoblastic leukemia (B-ALL) is a malignant disorder characterized by the abnormal proliferation of B-cell progenitors. Philadelphia chromosome-positive (Ph+) B-ALL is a subtype that expresses the Bcr-Abl fusion protein which represents a negative prognostic factor. Constitutive activation of the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) network is a common feature of B-ALL, influencing cell growth and survival. In the present study, we aimed to investigate the efficacy of PI3K isoform inhibition in B-ALL cell lines harboring the Bcr-Abl fusion protein.We studied the effects of anti Bcr-Abl drugs Imatinib, Nilotinib and GZD824 associated with PI3K isoform inhibitors. We used a panel of six compounds which specifically target PI3K isoforms including the pan-PI3K inhibitor ZSTK474, p110α BYL719 inhibitor and the dual p110γ/p110δ inhibitor IPI145. The effects of single drugs and of several drug combinations were analyzed to assess cytotoxicity by MTS assays, apoptosis and autophagy by flow cytometry and Western blot, as well as the phosphorylation status of the pathway.ZSTK474, BYL719 and IPI145 administered in combination with imatinib, nilotinib and GZD824 for 48 h, decreased cell viability, induced apoptosis and autophagy in a marked synergistic manner.These findings suggest that selected PI3K isoform inhibitors used in combination with anti Bcr-Abl drugs may be an attractive novel therapeutic intervention in Ph+ B-ALL.
Collapse
Affiliation(s)
- Simona Ultimo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, Rizzoli Orthopedic Institute, National Research Council, Bologna, Italy
| | | | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,LTTA Center, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
37
|
Oncogenic FLT3-ITD supports autophagy via ATF4 in acute myeloid leukemia. Oncogene 2017; 37:787-797. [PMID: 29059168 PMCID: PMC5808073 DOI: 10.1038/onc.2017.376] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 08/08/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022]
Abstract
In acute myeloid leukemia (AML), internal tandem duplication mutations in the FLT3 tyrosine kinase receptor (FLT3-ITD) account for up to 25% of cases and are associated with a poor outcome. In order to better target this AML subtype, a comprehensive view of how FLT3-ITD impacts AML cell biology is required. Here, we found that FLT3-ITD expression increased basal autophagy in AML cells, and that both pharmacological and genetic inhibition of the receptor reduced autophagy in primary AML samples and cell lines. Conditional expression of shRNAs against key autophagy proteins demonstrated that autophagy is required for AML cell proliferation in vitro and for leukemic cells survival in a mouse model of xenograft. Importantly, autophagy inhibition also overcame FLT3 inhibitor resistance both in vitro and in vivo. The transcription factor ATF4 was identified as an essential actor of FLT3-ITD-induced autophagy. Cellular levels of ATF4 were highly dependent on FLT3-ITD activity, and downregulation of ATF4 inhibited autophagy-dependent AML cell proliferation and improved overall mouse survival similarly to autophagy inhibition. These results suggest that targeting autophagy or ATF4 in patients expressing FLT3 mutations may represent a novel promising and innovative therapeutic strategy for AML.
Collapse
|
38
|
Zhao KH, Zhang C, Bai Y, Li Y, Kang X, Chen JX, Yao K, Jiang T, Zhong XS, Li WB. Antiglioma effects of cytarabine on leptomeningeal metastasis of high-grade glioma by targeting the PI3K/Akt/mTOR pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1905-1915. [PMID: 28721010 PMCID: PMC5500519 DOI: 10.2147/dddt.s135711] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leptomeningeal metastasis (LM) of high-grade glioma is a highly lethal disease requiring new effective therapeutic measures. For both de novo or relapsed glioma with LM, intrathecal cytarabine chemotherapy is not frequently used for first-line and relapse protocols. We encountered a clinical case demonstrating effective application of cytarabine in high-grade glioma with LM, prompting us to explore the effects of cytarabine on malignant glioma and molecular mechanisms of such effects through in vivo and in vitro experiments. The U87 cell line was selected to represent human glioma for studies. Cell viability was measured by MTT assay, plate colony formation assay, and trypan-blue dye exclusion test. Apoptosis was assessed by flow cytometry. Protein expression levels were detected by Western blot assay and immunohistochemistry. mRNA expression was examined by quantitative real-time reverse transcription polymerase chain reaction. Cytarabine inhibited tumor growth during the in vivo experiment. The present study confirmed that cytarabine inhibits proliferation and promotes apoptosis of U87 cells, and molecular analysis of this effect showed that cytarabine significantly reduces expression of phosphatidylinositol 3-kinase/serine/threonine kinase also known as the protein kinase B/mechanistic target of rapamycin (PI3K/Akt/mTOR) pathway, Ki-67, BCL2, and 4-1BB, and upregulates Bax and cleaved caspase-3. Our findings indicated that intrathecal administration of cytarabine manifests potential in prophylaxis and treatment of malignant glioma with LM. Effective medications for high-grade glioma with LM should contain cytarabine.
Collapse
Affiliation(s)
| | - Can Zhang
- Clinical Center of Gene And Cell Engineering, Beijing Shijitan Hospital
| | | | | | | | | | - Kun Yao
- Department of Neurosurgery, Beijing Tiantan Hospital
| | - Tao Jiang
- Department of Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiao-Song Zhong
- Clinical Center of Gene And Cell Engineering, Beijing Shijitan Hospital
| | | |
Collapse
|
39
|
Tabe Y, Tafuri A, Sekihara K, Yang H, Konopleva M. Inhibition of mTOR kinase as a therapeutic target for acute myeloid leukemia. Expert Opin Ther Targets 2017; 21:705-714. [PMID: 28537457 DOI: 10.1080/14728222.2017.1333600] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML), the most common acute leukemia in adults, remains a therapeutic challenge. The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway is one of the key aberrant intracellular axes involved in AML. Areas covered: mTOR plays a critical role in sensing and responding to environmental determinants such as nutrient availability, stress, and growth factor concentrations; and in modulating key cellular functions such as proliferation, metabolism, and survival. Although abnormalities of mTOR signaling are strongly associated with neoplastic leukemic proliferation, the role of pharmacologic inhibitors of mTOR in the treatment of AML has not been established. Expert opinion: Inhibition of mTOR signaling has in general modest growth-inhibitory effects in preclinical AML models and clinical trials. Yet, combination of allosteric mTOR inhibitors with standard chemotherapy or targeted agents has a greater anti-leukemia efficacy. In turn, dual mTORC1/2 inhibitors, and dual PI3K/mTOR inhibitors show greater activity in pre-clinical AML models. Further, understanding the role of mTOR signaling in stemness of leukemias is important because AML stem cells may become chemoresistant by displaying aberrant signaling molecules, modifying epigenetic mechanisms, and altering the components of the bone marrow microenvironment.
Collapse
Affiliation(s)
- Yoko Tabe
- a Department of Next Generation Hematology Laboratory Medicine , Juntendo University School of Medicine , Tokyo , Japan.,b Section of Molecular Hematology and Therapy, Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Agostino Tafuri
- c Dipartimento di Medicina Clinica e Molecolare , "Sapienza" University of Rome , Rome , Italy
| | - Kazumasa Sekihara
- d Leading Center for the Development and Research of Cancer Medicine , Juntendo University School of Medicine , Tokyo , Japan
| | - Haeun Yang
- d Leading Center for the Development and Research of Cancer Medicine , Juntendo University School of Medicine , Tokyo , Japan
| | - Marina Konopleva
- b Section of Molecular Hematology and Therapy, Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
40
|
Tiong IS, Tan P, McManus J, Cummings N, Sadawarte S, Catalano J, Hills R, Wei A. Phase Ib study of the mTOR inhibitor everolimus with low dose cytarabine in elderly acute myeloid leukemia. Leuk Lymphoma 2017; 59:493-496. [DOI: 10.1080/10428194.2017.1334122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ing Soo Tiong
- Department of Clinical Haematology, Alfred Hospital, Melbourne, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Peter Tan
- Department of Clinical Haematology, Alfred Hospital, Melbourne, Australia
| | - Julie McManus
- Department of Pathology, Alfred Hospital, Melbourne, Australia
| | - Nik Cummings
- Department of Pathology, Alfred Hospital, Melbourne, Australia
| | - Sonali Sadawarte
- Department of Clinical Haematology, Alfred Hospital, Melbourne, Australia
| | - John Catalano
- Department of Haematology, Frankston Hospital, Frankston, Australia
| | - Robert Hills
- Department of Haematology, Cardiff University School of Medicine, Cardiff, UK
| | - Andrew Wei
- Department of Clinical Haematology, Alfred Hospital, Melbourne, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| |
Collapse
|
41
|
Siqueira RP, Barros MVDA, Barbosa ÉDAA, Onofre TS, Gonçalves VHS, Pereira HS, Silva Júnior A, de Oliveira LL, Almeida MR, Fietto JLR, Teixeira RR, Bressan GC. Trifluoromethyl arylamides with antileukemia effect and intracellular inhibitory activity over serine/arginine-rich protein kinases (SRPKs). Eur J Med Chem 2017; 134:97-109. [PMID: 28407594 DOI: 10.1016/j.ejmech.2017.03.078] [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: 12/20/2016] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 12/19/2022]
Abstract
The serine/arginine-rich protein kinases (SRPKs) have frequently been found with altered activity in a number of cancers, suggesting they could serve as potential therapeutic targets in oncology. Here we describe the synthesis of a series of twenty-two trifluoromethyl arylamides based on the known SRPKs inhibitor N-(2-(piperidin-1-yl)-5-(trifluoromethyl)phenyl)isonicotinamide (SRPIN340) and the evaluation of their antileukemia effects. Some derivatives presented superior cytotoxic effects against myeloid and lymphoid leukemia cell lines compared to SRPIN340. In particular, compounds 24, 30, and 36 presented IC50 values ranging between 6.0 and 35.7 μM. In addition, these three compounds were able to trigger apoptosis and autophagy, and to exhibit synergistic effects with the chemotherapeutic agent vincristine. Furthermore, compound 30 was more efficient than SRPIN340 in impairing the intracellular phosphorylation status of SR proteins as well as the expression of MAP2K1, MAP2K2, VEGF, and RON oncogenic isoforms. Therefore, novel compounds with increased intracellular effects against SRPK activity were obtained, contributing to medicinal chemistry efforts towards the development of new anticancer agents.
Collapse
Affiliation(s)
- Raoni Pais Siqueira
- Universidade Federal de Viçosa, Departamento de Bioquímica e Biologia Molecular, Viçosa, MG, Brazil
| | | | | | - Thiago Souza Onofre
- Universidade Federal de Viçosa, Departamento de Bioquímica e Biologia Molecular, Viçosa, MG, Brazil
| | | | - Higor Sette Pereira
- Universidade Federal de Viçosa, Departamento de Bioquímica e Biologia Molecular, Viçosa, MG, Brazil
| | | | | | - Márcia Rogéria Almeida
- Universidade Federal de Viçosa, Departamento de Bioquímica e Biologia Molecular, Viçosa, MG, Brazil
| | | | | | - Gustavo Costa Bressan
- Universidade Federal de Viçosa, Departamento de Bioquímica e Biologia Molecular, Viçosa, MG, Brazil.
| |
Collapse
|
42
|
Autophagy, a key mechanism of oncogenesis and resistance in leukemia. Blood 2016; 129:547-552. [PMID: 27956388 DOI: 10.1182/blood-2016-07-692707] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/07/2016] [Indexed: 01/01/2023] Open
Abstract
Autophagy is a lysosomal pathway involved in degradation of intracellular material. It appears as an adaptation mechanism that is essential for cellular homeostasis in response to various stress conditions. Over the past decade, many studies have linked alteration of autophagy with cancer initiation and progression, autoimmune, inflammatory, metabolic, and degenerative diseases. This review highlights recent findings on the impact of autophagy on leukemic transformation of normal hematopoietic stem cells and summarizes its role on leukemic cell response to chemotherapy.
Collapse
|
43
|
Thomé MP, Filippi-Chiela EC, Villodre ES, Migliavaca CB, Onzi GR, Felipe KB, Lenz G. Ratiometric analysis of Acridine Orange staining in the study of acidic organelles and autophagy. J Cell Sci 2016; 129:4622-4632. [PMID: 27875278 DOI: 10.1242/jcs.195057] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/07/2016] [Indexed: 01/06/2023] Open
Abstract
Acridine Orange is a cell-permeable green fluorophore that can be protonated and trapped in acidic vesicular organelles (AVOs). Its metachromatic shift to red fluorescence is concentration-dependent and, therefore, Acridine Orange fluoresces red in AVOs, such as autolysosomes. This makes Acridine Orange staining a quick, accessible and reliable method to assess the volume of AVOs, which increases upon autophagy induction. Here, we describe a ratiometric analysis of autophagy using Acridine Orange, considering the red-to-green fluorescence intensity ratio (R/GFIR) to quantify flow cytometry and fluorescence microscopy data of Acridine-Orange-stained cells. This method measured with accuracy the increase in autophagy induced by starvation or rapamycin, and the reduction in autophagy produced by bafilomycin A1 or the knockdown of Beclin1 or ATG7. Results obtained with Acridine Orange, considering R/GFIR, correlated with the conversion of the unlipidated form of LC3 (LC3-I) into the lipidated form (LC3-II), SQSTM1 degradation and GFP-LC3 puncta formation, thus validating this assay to be used as an initial and quantitative method for evaluating the late step of autophagy in individual cells, complementing other methods.
Collapse
Affiliation(s)
- Marcos P Thomé
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil
| | - Eduardo C Filippi-Chiela
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil.,Faculdade de Medicina, UFRGS, 91501970, Porto Alegre, Brazil
| | - Emilly S Villodre
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil
| | - Celina B Migliavaca
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil
| | - Giovana R Onzi
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil
| | - Karina B Felipe
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil.,Departamento de Análises Clínicas, Universidade Federal do Paraná (UFPR), 80060000, Curitiba, Brazil
| | - Guido Lenz
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), 91501970, Porto Alegre, Brazil .,Centro de Biotecnologia, UFRGS, 91501970, Porto Alegre, Brazil
| |
Collapse
|
44
|
Cytoprotective autophagy maintains leukemia-initiating cells in murine myeloid leukemia. Blood 2016; 128:1614-24. [DOI: 10.1182/blood-2015-12-684696] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 07/20/2016] [Indexed: 12/15/2022] Open
Abstract
Key Points
Autophagy is required for maintenance of AML-initiating cells and peripheral myeloblast survival. Loss of autophagy potentiates the therapeutic effects of AraC in vivo.
Collapse
|
45
|
Piya S, Kornblau SM, Ruvolo VR, Mu H, Ruvolo PP, McQueen T, Davis RE, Hail N, Kantarjian H, Andreeff M, Borthakur G. Atg7 suppression enhances chemotherapeutic agent sensitivity and overcomes stroma-mediated chemoresistance in acute myeloid leukemia. Blood 2016; 128:1260-9. [PMID: 27268264 PMCID: PMC5009514 DOI: 10.1182/blood-2016-01-692244] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/30/2016] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a cellular adaptive mechanism to stress, including that induced by chemotherapeutic agents. Reverse phase protein array suggested that high expression of the essential autophagy-related protein, Atg7, was associated with shorter remission in newly diagnosed acute myeloid leukemia (AML) patient samples, indicating a role in chemoresistance. Knockdown of Atg7 in AML cells using short hairpin RNA markedly increased apoptosis and DNA damage following treatment with cytarabine and idarubicin. Interestingly, coculture of AML cells with stromal cells increased autophagy and chemoresistance in the AML cells exposed to chemotherapeutic agents, and this was reversed following Atg7 knockdown. This effect was further enhanced by concomitant knockdown of Atg7 in both AML and stromal cells. These findings strongly suggest that Atg7, and likely microenvironment autophagy in general, plays an important role in AML chemoresistance. Mechanistic studies revealed that Atg7 knockdown induced a proapoptotic phenotype in AML cells, which was manifested by an increased NOXA expression at the transcriptional level. Finally, in a mouse model of human leukemia, Atg7 knockdown extended overall survival after chemotherapy. Thus, the inhibition of Atg7 appears to be a valid strategy to enhance chemosensitivity, and it could indeed improve outcomes in AML therapy.
Collapse
MESH Headings
- Animals
- Autophagy/drug effects
- Autophagy/genetics
- Autophagy-Related Protein 7/genetics
- Autophagy-Related Protein 7/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Gene Knockdown Techniques
- HL-60 Cells
- Humans
- K562 Cells
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Proto-Oncogene Proteins c-bcl-2/biosynthesis
- Proto-Oncogene Proteins c-bcl-2/genetics
- Stromal Cells/metabolism
- Stromal Cells/pathology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Sujan Piya
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| | | | - Vivian R Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| | - Hong Mu
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| | - Peter P Ruvolo
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| | - Teresa McQueen
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| | - R Eric Davis
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Michael Andreeff
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| | - Gautam Borthakur
- Department of Leukemia, Section of Molecular Hematology and Therapy, and
| |
Collapse
|
46
|
Effects of IL-33/ST2 pathway in acute inflammation on tissue damage, antioxidative parameters, magnesium concentration and cytokines profile. Exp Mol Pathol 2016; 101:31-7. [PMID: 27222019 DOI: 10.1016/j.yexmp.2016.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/10/2016] [Accepted: 05/20/2016] [Indexed: 11/22/2022]
Abstract
AIM The aim of this study was to examine the role of IL-33/ST2 pathway in a pathogenesis of acute inflammation and its effects on tissue damage, antioxidative capacity, magnesium concentration and cytokine profile in acutely inflamed tissue. MATERIAL AND METHODS Male mice were randomly divided in four groups: wild-type control group (WT-C), ST2 knockout control group (KO-C), wild-type inflammatory group (WT-I), and ST2 knockout inflammatory group (KO-I). Acute inflammation was induced in WT-I and KO-I by intramuscular injection of turpentine oil, while mice in WT-C and KO-C were treated with saline. After 12h, animals were euthanized, and blood was collected for determination of creatine kinase (CK) and aspartate transaminase (AST) activity. The treated tissue was used for histopathological analysis, determination of volume density of inflammatory infiltrate (Vdii) and necrotic fiber (Vdnf), gene expression of interleukin (IL)-33, ST2, tumor necrosis factor alpha (TNF-alpha), IL-6, IL-12p35, and transforming growth factor beta (TGF-beta), concentration of magnesium (Mg), copper (Cu), selenium (Se), manganese (Mn) and reduced glutathione (GSH), and superoxide dismutase (SOD) and glutathione peroxidase (GPx) activity. RESULTS Presence of inflammatory infiltration and necrosis in the treated tissue was histopathologically confirmed in WT-I and KO-I. Vdii was significantly higher in WT-I when compared to KO-I, whereas Vdnf did not significantly differ between WT-I and KO-I. CK and AST significantly increased in both inflammatory groups when compared to corresponding control groups. However, the values of CK and AST were significantly higher in WT-I than in KO-I. Mg in the treated tissue was significantly lower in WT-I in comparison to WT-C and KO-I, while there was no significant difference between KO-C and KO-I. There was no significant difference in Cu, Se, and Mn in the treated tissue between WT-C, KO-C, WT-I and KO-I. Gene expression of IL-33 in the treated tissue increased in both inflammatory groups when compared to the corresponding control groups, but it was significantly higher in KO-I than in WT-I. Gene expression of ST2 in the treated tissue was significantly higher in WT-I than in WT-C. Gene expression of TNF-alpha, IL-6, and IL-12p35 in the treated tissue was significantly higher in WT-I and KO-I than in the corresponding control groups, and IL-6 was significantly higher in KO-C than in WT-C. TGF-beta gene expression in the treated tissue was significantly higher in KO-I when compared to WT-I, while there was no difference between WT-C and KO-C. SOD activity decreased at the site of acute inflammation in both inflammatory groups, while the GPx activity increased. GSH in the treated tissue was significantly higher in KO-I than in KO-C or WT-I. CONCLUSION The results of our study have indicated, to our knowledge for the first time, that IL-33/ST2 pathway plays a role in enhancing inflammation and tissue damage at the site of acute inflammation by affecting the concentration of magnesium and GSH, important for antioxidative capacity, as well as gene expression of anti-inflammatory cytokine TGF-beta.
Collapse
|
47
|
Nakanishi T, Song Y, He C, Wang D, Morita K, Tsukada J, Kanazawa T, Yoshida Y. Relationship between triterpenoid anticancer drug resistance, autophagy, and caspase-1 in adult T-cell leukemia. PeerJ 2016; 4:e2026. [PMID: 27190722 PMCID: PMC4868592 DOI: 10.7717/peerj.2026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/19/2016] [Indexed: 12/24/2022] Open
Abstract
We previously reported that the inflammasome inhibitor cucurbitacin D (CuD) induces apoptosis in human leukemia cell lines. Here, we investigated the effects of CuD and a B-cell lymphoma extra-large (Bcl-xL) inhibitor on autophagy in peripheral blood lymphocytes (PBL) isolated from adult T-cell leukemia (ATL) patients. CuD induced PBL cell death in patients but not in healthy donors. This effect was not significantly inhibited by treatment with rapamycin or 3-methyladenine (3-MA). The Bcl-xL inhibitor Z36 induced death in primary cells from ATL patients including that induced by CuD treatment, effects that were partly inhibited by 3-MA. Similarly, cell death induced by the steroid prednisolone was enhanced in the presence of Z36. A western blot analysis revealed that Z36 also promoted CuD-induced poly(ADP ribose) polymerase cleavage. Interestingly, the effects of CuD and Z36 were attenuated in primary ATL patient cells obtained upon recurrence after umbilical cord blood transplantation, as compared to those obtained before chemotherapy. Furthermore, cells from this patient expressed a high level of caspase-1, and treatment with caspase-1 inhibitor-enhanced CuD-induced cell death. Taken together, these results suggest that rescue from resistance to steroid drugs can enhance chemotherapy, and that caspase-1 is a good marker for drug resistance in ATL patients.
Collapse
Affiliation(s)
- Tsukasa Nakanishi
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.,Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuan Song
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.,Department of Clinical Laboratory, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Cuiying He
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Duo Wang
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kentaro Morita
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Junichi Tsukada
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tamotsu Kanazawa
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yasuhiro Yoshida
- Department of Immunology and Parasitology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
48
|
Nakanishi T, Song Y, He C, Wang D, Morita K, Tsukada J, Kanazawa T, Yoshida Y. Autophagy is associated with cucurbitacin D-induced apoptosis in human T cell leukemia cells. Med Oncol 2016; 33:30. [PMID: 26913856 DOI: 10.1007/s12032-016-0743-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/10/2016] [Indexed: 12/22/2022]
Abstract
We previously reported that the inflammasome inhibitor cucurbitacin D (CuD) induces apoptosis in human leukemia cell lines. In the present study, we investigated the effects of co-treatment with an additional Bcl-xL inhibitor, Z36. Treatment with Z36 induced cell death in leukemia cell lines, with MT-4 cells exhibiting the lowest sensitivity to Z36. Co-treatment of cells with Z36 and CuD resulted in a greater degree of cell death for Hut78 and Jurkat cells than treatment with CuD alone. In contrast, co-treatment of MT-4 cells with Z36 and CuD had a suppressive effect on cell death. The autophagy inhibitor 3-methyladenine (3-MA) suppressed the growth of leukemia cell lines HuT78, Jurkat, MT-1, and MT-4. CuD-induced cell death was enhanced by 3-MA in Jurkat cells, but inhibited in MT-4 cells. Western blotting results revealed cleavage of poly(ADP ribose) polymerase (PARP), supporting CuD-induced cell death; 3-MA enhanced CuD-Z36-induced PARP cleavage. Taken together, our results indicate that autophagy negatively regulates chemical-induced cell death of leukemia cells, and that controlling autophagy could be beneficial in the development of more effective chemotherapies against leukemia.
Collapse
Affiliation(s)
- Tsukasa Nakanishi
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Yuan Song
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.,Department of Clinical Laboratory, Fourth Hospital of Hebei Medical University, No. 169 Tian Shan Street, Shijiazhuang, 050035, China
| | - Cuiying He
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Duo Wang
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Kentaro Morita
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Junichi Tsukada
- Department of Hematology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Tamotsu Kanazawa
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Yasuhiro Yoshida
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
49
|
Carneiro BA, Kaplan JB, Altman JK, Giles FJ, Platanias LC. Targeting mTOR signaling pathways and related negative feedback loops for the treatment of acute myeloid leukemia. Cancer Biol Ther 2015; 16:648-56. [PMID: 25801978 PMCID: PMC4622839 DOI: 10.1080/15384047.2015.1026510] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/01/2015] [Indexed: 12/29/2022] Open
Abstract
An accumulating understanding of the complex pathogenesis of acute myeloid leukemia (AML) continues to lead to promising therapeutic approaches. Among the key aberrant intracellular signaling pathways involved in AML, the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) axis is of major interest. This axis modulates a wide array of critical cellular functions, including proliferation, metabolism, and survival. Pharmacologic inhibitors of components of this pathway have been developed over the past decade, but none has an established role in the treatment of AML. This review will discuss the preclinical data and clinical results driving ongoing attempts to exploit the PI3K/AKT/mTOR pathway in patients with AML and address issues related to negative feedback loops that account for leukemic cell survival. Targeting the PI3K/AKT/mTOR pathway is of high interest for the treatment of AML, but combination therapies with other targeted agents may be needed to block negative feedback loops in leukemia cells.
Collapse
Affiliation(s)
- Benedito A Carneiro
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Jason B Kaplan
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Jessica K Altman
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Francis J Giles
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Leonidas C Platanias
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
- Division of Hematology-Oncology; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL, USA
| |
Collapse
|
50
|
You eat what you are: autophagy inhibition as a therapeutic strategy in leukemia. Leukemia 2014; 29:517-25. [PMID: 25541151 DOI: 10.1038/leu.2014.349] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 11/25/2014] [Indexed: 12/13/2022]
Abstract
A deeper understanding of the role of autophagy, literally 'self-eating', in normal and cancer cell biology has emerged over the last few years. Autophagy serves as a vehicle for cells to respond to various stressors including genomic, hypoxic and nutrient stress, and to oppose mechanisms of 'programmed' cell death. Here, we review not only mechanisms of cell death and cell survival but also the early successes in applying autophagy inhibition strategies in solid tumors using the only currently available clinical inhibitor, oral hydroxychloroquine. In acute leukemia, currently available chemotherapy drugs promote cell death and demonstrate clinical benefit, but relapse and subsequent chemotherapy resistance is common. Increasing preclinical data suggest that autophagy is active in leukemia as a means of promoting cell survival in response to chemotherapy. We propose coupling autophagy inhibition strategies with current cytotoxic chemotherapy and discuss synergistic combinations of available anti-leukemic therapies with autophagy inhibition. Furthermore, novel autophagy inhibitors are in development and promise to provide new therapeutic opportunities for patients with leukemia.
Collapse
|