1
|
Shi H, Zhang M, Zhang Y. Construction of a prognostic model for autophagy in Wilm's tumor. Pediatr Surg Int 2024; 40:122. [PMID: 38704513 DOI: 10.1007/s00383-024-05712-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Wilm's tumor (WT) is one of the most common childhood urological tumors, ranking second in the incidence of pediatric abdominal tumors. The development of WT is associated with various factors, and the correlation with autophagy is currently unclear. PURPOSE To develop a new prognostic model of autophagy-related genes (ATG) for WT. METHODS Using the Therapeutically applicable research to generate effective treatments (TARGET) database to screen for differentially expressed ATGs in WT and normal tissues. ATGs were screened for prognostic relevance to WT using one-way and multifactorial Cox regression analyses and prognostic models were constructed. The risk score was calculated according to the model, and the predictive ability of the constructed model was analyzed using the ROC (receiver operating characteristic) curve to verify the significance of the model for the prognosis of WT. RESULTS Sixty-eight differentially expressed ATGs were identified by univariate Cox regression analysis, and two critical prognostic ATGs (CXCR4 and ERBB2) were identified by multivariate Cox regression analysis. Patients were divided into high-risk and low-risk groups according to the differential expression of these two ATGs. Kaplan-Meier (KM) curves showed a significant difference in survival time between the two groups. The critical prognostic ATGs were combined with race, age, and stage in a multifactorial regression analysis, and the final prognostic model was produced as a line graph. CONCLUSION The prognostic model of autophagy-related genes composed of the CXCR4 gene and ERBB2 gene has a specific predictive value for the prognosis of WT, and the present study provides a clear basis for future research on biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Haoyu Shi
- Department of Pediatric Surgery, Affiliated Matern and Child Care Hospital of Nantong University, 399 Century Avenue, Chongchuan, Nantong, Jiangsu, China
| | - Min Zhang
- Department of Pediatric Surgery, Affiliated Matern and Child Care Hospital of Nantong University, 399 Century Avenue, Chongchuan, Nantong, Jiangsu, China.
| | - Youbo Zhang
- Department of Pediatric Surgery, Affiliated Matern and Child Care Hospital of Nantong University, 399 Century Avenue, Chongchuan, Nantong, Jiangsu, China
| |
Collapse
|
2
|
Wang Y, Xu Y, Zhu C. The Role of Autophagy in Childhood Central Nervous System Tumors. Curr Treat Options Oncol 2022; 23:1535-1547. [PMID: 36197606 PMCID: PMC9596594 DOI: 10.1007/s11864-022-01015-6] [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] [Accepted: 08/19/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT Autophagy is a physiological process that occurs in normal tissues. Under external environmental pressure or internal environmental changes, cells can digest part of their contents through autophagy in order to reduce metabolic pressure or remove damaged organelles. In cancer, autophagy plays a paradoxical role, acting as a tumor suppressor-by removing damaged organelles and inhibiting inflammation or by promoting genome stability and the tumor-adaptive responses-as a pro-survival mechanism to protect cells from stress. In this article, we review the autophagy-dependent mechanisms driving childhood central nervous system tumor cell death, malignancy invasion, chemosensitivity, and radiosensitivity. Autophagy inhibitors and inducers have been developed, and encouraging results have been achieved in autophagy modulation, suggesting that these might be potential therapeutic agents for the treatment of pediatric central nervous system (CNS) tumors.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Hematology and Oncology, Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital,Zhengzhou Children’s Hospital, Zhengzhou, 450018 China ,Henan Key Laboratory of Child Brain injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China ,Commission Key Laboratory of Birth Defects Prevention,Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China ,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
3
|
Huang Z, Jin G. Licochalcone B induced apoptosis and autophagy in osteosarcoma tumor cells via the inactivation of PI3K/AKT/mTOR pathway. Biol Pharm Bull 2022; 45:730-737. [DOI: 10.1248/bpb.b21-00991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Zhihui Huang
- Department of Orthopaedics, The 904th Hospital of Joint Logistic Support Force
| | - Genyang Jin
- Department of Orthopaedics, The 904th Hospital of Joint Logistic Support Force
| |
Collapse
|
4
|
The Dual Role of Autophagy in Crizotinib-Treated ALK + ALCL: From the Lymphoma Cells Drug Resistance to Their Demise. Cells 2021; 10:cells10102517. [PMID: 34685497 PMCID: PMC8533885 DOI: 10.3390/cells10102517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023] Open
Abstract
Autophagy has been described as harboring a dual role in cancer development and therapy. Depending on the context, it can exert either pro-survival or pro-death functions. Here, we review what is known about autophagy in crizotinib-treated ALK+ ALCL. We first present our main findings on the role and regulation of autophagy in these cells. Then, we provide literature-driven hypotheses that could explain mechanistically the pro-survival properties of autophagy in crizotinib-treated bulk and stem-like ALK+ ALCL cells. Finally, we discuss how the potentiation of autophagy, which occurs with combined therapies (ALK and BCL2 or ALK and RAF1 co-inhibition), could convert it from a survival mechanism to a pro-death process.
Collapse
|
5
|
Autophagy and the Wnt signaling pathway: A focus on Wnt/β-catenin signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118926. [PMID: 33316295 DOI: 10.1016/j.bbamcr.2020.118926] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/07/2020] [Accepted: 12/05/2020] [Indexed: 12/11/2022]
Abstract
Cellular homeostasis and adaptation to various environmental conditions are importantly regulated by the sophisticated mechanism of autophagy and its crosstalk with Wnt signaling and other developmental pathways. Both autophagy and Wnt signaling are involved in embryogenesis and differentiation. Autophagy is responsible for degradation and recycling of cytosolic materials by directing them to lysosomes through the phagophore compartment. A dual feedback mechanism regulates the interface between autophagy and Wnt signaling pathways. During nutrient deprivation, β-catenin and Dishevelled (essential Wnt signaling proteins) are targeted for autophagic degradation by LC3. When Wnt signaling is activated, β-catenin acts as a corepressor of one of the autophagy proteins, p62. In contrast, another key Wnt signaling protein, GSK3β, negatively regulates the Wnt pathway and has been shown to induce autophagy by phosphorylation of the TSC complex. This article reviews the interplay between autophagy and Wnt signaling, describing how β-catenin functions as a key cellular integration point coordinating proliferation with autophagy, and it discusses the clinical importance of the crosstalk between these mechanisms.
Collapse
|
6
|
Sorrentino D, Frentzel J, Mitou G, Blasco RB, Torossian A, Hoareau-Aveilla C, Pighi C, Farcé M, Meggetto F, Manenti S, Espinos E, Chiarle R, Giuriato S. High Levels of miR-7-5p Potentiate Crizotinib-Induced Cytokilling and Autophagic Flux by Targeting RAF1 in NPM-ALK Positive Lymphoma Cells. Cancers (Basel) 2020; 12:cancers12102951. [PMID: 33066037 PMCID: PMC7650725 DOI: 10.3390/cancers12102951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Anaplastic lymphoma kinase positive anaplastic large cell lymphomas are a pediatric disease, which still needs treatment improvement. Crizotinib was the first ALK-targeted inhibitor used in clinics, but relapses are now known to occur. Current research efforts indicate that combined therapies could represent a superior strategy to eradicate malignant cells and prevent tumor recurrence. Autophagy is a self-digestion cellular process, known to be induced upon diverse cancer therapies. Our present work demonstrates that the potentiation of the crizotinib-induced autophagy flux, through the serine/threonine kinase RAF1 downregulation, drives ALK+ ALCL cells to death. These results should encourage further investigations on the therapeutic modulation of autophagy in this particular cancer settings and other ALK-related malignancies. Abstract Anaplastic lymphoma kinase positive anaplastic large cell lymphomas (ALK+ ALCL) are an aggressive pediatric disease. The therapeutic options comprise chemotherapy, which is efficient in approximately 70% of patients, and targeted therapies, such as crizotinib (an ALK tyrosine kinase inhibitor (TKI)), used in refractory/relapsed cases. Research efforts have also converged toward the development of combined therapies to improve treatment. In this context, we studied whether autophagy could be modulated to improve crizotinib therapy. Autophagy is a vesicular recycling pathway, known to be associated with either cell survival or cell death depending on the cancer and therapy. We previously demonstrated that crizotinib induced cytoprotective autophagy in ALK+ lymphoma cells and that its further intensification was associated with cell death. In line with these results, we show here that combined ALK and Rapidly Accelerated Fibrosarcoma 1 (RAF1) inhibition, using pharmacological (vemurafenib) or molecular (small interfering RNA targeting RAF1 (siRAF1) or microRNA-7-5p (miR-7-5p) mimics) strategies, also triggered autophagy and potentiated the toxicity of TKI. Mechanistically, we found that this combined therapy resulted in the decrease of the inhibitory phosphorylation on Unc-51-like kinase-1 (ULK1) (a key protein in autophagy initiation), which may account for the enforced autophagy and cytokilling effect. Altogether, our results support the development of ALK and RAF1 combined inhibition as a new therapeutic approach in ALK+ ALCL.
Collapse
Affiliation(s)
- Domenico Sorrentino
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- Ligue Nationale Contre le Cancer, équipe labellisée 2016, F-31037 Toulouse, France
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
| | - Julie Frentzel
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Merck Serono S.A., Department of Biotechnology Process Sciences, Route de Fenil 25, Z.I. B, 1804 Corsier-sur-Vevey, Switzerland
| | - Géraldine Mitou
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
| | - Rafael B. Blasco
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
| | - Avédis Torossian
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
| | - Coralie Hoareau-Aveilla
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
| | - Chiara Pighi
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Manon Farcé
- Pôle Technologique du CRCT—Plateau de Cytométrie et Tri cellulaire—INSERM U1037, F-31037 Toulouse, France;
| | - Fabienne Meggetto
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
| | - Stéphane Manenti
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Ligue Nationale Contre le Cancer, équipe labellisée 2016, F-31037 Toulouse, France
| | - Estelle Espinos
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
| | - Roberto Chiarle
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Sylvie Giuriato
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- Ligue Nationale Contre le Cancer, équipe labellisée 2016, F-31037 Toulouse, France
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain
- Correspondence: ; Tel.: +33-(5)-82-74-16-35
| |
Collapse
|
7
|
Zhou J, Jiang Y, Chen H, Wu Y, Zhang L. Tanshinone I attenuates the malignant biological properties of ovarian cancer by inducing apoptosis and autophagy via the inactivation of PI3K/AKT/mTOR pathway. Cell Prolif 2020; 53:e12739. [PMID: 31820522 PMCID: PMC7046305 DOI: 10.1111/cpr.12739] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES Tanshinone I (Tan-I) is one of the vital fatsoluble monomer components, which extracted from Chinese medicinal herb Salvia miltiorrhiza Bunge. It has been shown that Tan-I exhibited anti-tumour activities on different types of cancers. However, the underlying mechanisms by which Tan-Ⅰ regulates apoptosis and autophagy in ovarian cancer remain unclear. Thus, this study aimed to access the therapy effect of Tan-Ⅰ and the underlying mechanisms. METHODS Ovarian cancer cells A2780 and ID-8 were treated with different concentrations of Tan-Ⅰ (0, 1.2, 2.4, 4.8 and 9.6 μg/mL) for 24 hours. The cell proliferation was analysed by CCK8 assay, EdU staining and clone formation assay. Apoptosis was assessed by the TUNEL assay and flow cytometry. The protein levels of apoptosis protein (Caspase-3), autophagy protein (Beclin1, ATG7, p62 and LC3II/LC3I) and PI3K/AKT/mTOR pathway were determined by Western blot. Autophagic vacuoles in cells were observed with LC3 dyeing using confocal fluorescent microscopy. Anti-tumour activity of Tan-Ⅰ was accessed by subcutaneous xeno-transplanted tumour model of human ovarian cancer in nude mice. The Ki67, Caspase-3 level and apoptosis level were analysed by immunohistochemistry and TUNEL staining. RESULTS Tan-Ⅰ inhibited the proliferation of ovarian cancer cells A2780 and ID-8 in a dose-dependent manner, based on CCK8 assay, EdU staining and clone formation assay. In additional, Tan-Ⅰ induced cancer cell apoptosis and autophagy in a dose-dependent manner in ovarian cancer cells by TUNEL assay, flow cytometry and Western blot. Tan-Ⅰ significantly inhibited tumour growth by inducing cell apoptosis and autophagy. Mechanistically, Tan-Ⅰ activated apoptosis-associated protein Caspase-3 cleavage to promote cell apoptosis and inhibited PI3K/AKT/mTOR pathway to induce autophagy. CONCLUSIONS This is the first evidence that Tan-Ⅰ induced apoptosis and promoted autophagy via the inactivation of PI3K/AKT/mTOR pathway on ovarian cancer and further inhibited tumour growth, which might be considered as effective strategy.
Collapse
Affiliation(s)
- Jin Zhou
- College of ScienceSichuan Agricultural UniversityYa'anChina
| | | | - Huan Chen
- College of ScienceSichuan Agricultural UniversityYa'anChina
| | - Yi‐chao Wu
- College of Life ScienceChina West Normal UniversityNanchongChina
| | - Li Zhang
- College of ScienceSichuan Agricultural UniversityYa'anChina
| |
Collapse
|
8
|
A novel ALK inhibitor ZYY inhibits Karpas299 cell growth in vitro and in a mouse xenograft model and induces protective autophagy. Toxicol Appl Pharmacol 2019; 383:114781. [DOI: 10.1016/j.taap.2019.114781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022]
|
9
|
Oncogenic KIT mutations induce STAT3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 2019; 8:39. [PMID: 31311917 PMCID: PMC6635375 DOI: 10.1038/s41389-019-0148-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 04/10/2019] [Accepted: 05/31/2019] [Indexed: 12/14/2022] Open
Abstract
Autophagy is associated with both survival and cell death in myeloid malignancies. Therefore, deciphering its role in different genetically defined subtypes of acute myeloid leukemia (AML) is critical. Activating mutations of the KIT receptor tyrosine kinase are frequently detected in core-binding factor AML and are associated with a greater risk of relapse. Herein, we report that basal autophagy was significantly increased by the KITD816V mutation in AML cells and contributed to support their cell proliferation and survival. Invalidation of the key autophagy protein Atg12 strongly reduced tumor burden and improved survival of immunocompromised NSG mice engrafted with KITD816V TF-1 cells. Downstream of KITD816V, STAT3, but not AKT or ERK pathways, was identified as a major regulator of autophagy. Accordingly, STAT3 pharmacological inhibition or downregulation inhibited autophagy and reduced tumor growth both in vitro and in vivo. Taken together, our results support the notion that targeting autophagy or STAT3 opens up an exploratory pathway for finding new therapeutic opportunities for patients with CBF-AML or others malignancies with KITD816V mutations.
Collapse
|
10
|
Poillet-Perez L. [Host autophagy is feeding tumor]. Med Sci (Paris) 2019; 35:512-514. [PMID: 31274079 DOI: 10.1051/medsci/2019103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Laura Poillet-Perez
- Institut Rutgers du cancer de l'État du New Jersey, 195 Little Albany street, NJ 08903 New Brunswick, États-Unis
| |
Collapse
|
11
|
Torossian A, Broin N, Frentzel J, Daugrois C, Gandarillas S, Saati TA, Lamant L, Brousset P, Giuriato S, Espinos E. Blockade of crizotinib-induced BCL2 elevation in ALK-positive anaplastic large cell lymphoma triggers autophagy associated with cell death. Haematologica 2019; 104:1428-1439. [PMID: 30679328 PMCID: PMC6601090 DOI: 10.3324/haematol.2017.181966] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphomas are tumors that carry translocations involving the ALK gene at the 2p23 locus, leading to the expression of ALK tyrosine kinase fusion oncoproteins. Amongst hematologic malignancies, these lymphomas are particular in that they express very low levels of B-cell lymphoma 2 (BCL2), a recognized inhibitor of apoptosis and autophagy, two processes that share complex interconnections. We have previously shown that treatment of ALK-positive anaplastic large cell lymphoma cells with the ALK tyrosine kinase inhibitor crizotinib induces autophagy as a pro-survival response. Here, we observed that crizotinib-mediated inactivation of ALK caused an increase in BCL2 levels that restrained the cytotoxic effects of the drug. BCL2 downregulation in combination with crizotinib treatment potentiated loss of cell viability through both an increase in autophagic flux and cell death, including apoptosis. More importantly, our data revealed that the blockade of autophagic flux completely reversed impaired cell viability, which demonstrates that excessive autophagy is associated with cell death. We propose that the downregulation of BCL2 protein, which plays a central role in the autophagic and apoptotic machinery, combined with crizotinib treatment may represent a promising therapeutic alternative to current ALK-positive anaplastic large cell lymphoma treatments.
Collapse
Affiliation(s)
- Avedis Torossian
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France
| | - Nicolas Broin
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France
| | - Julie Frentzel
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France
| | - Camille Daugrois
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Laboratoire d'Excellence Toulouse-Cancer-TOUCAN, F-31024 Toulouse, France
| | | | - Talal Al Saati
- Inserm/UPS, US006/CREFRE, Service d'Histopathologie, F-31000 Toulouse, France
| | - Laurence Lamant
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Laboratoire d'Excellence Toulouse-Cancer-TOUCAN, F-31024 Toulouse, France.,Département de Pathologie, IUCT, F-31000 Toulouse, France.,European Research Initiative on ALK-related Malignancies (ERIA), Cambridge, UK
| | - Pierre Brousset
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Laboratoire d'Excellence Toulouse-Cancer-TOUCAN, F-31024 Toulouse, France.,Département de Pathologie, IUCT, F-31000 Toulouse, France.,European Research Initiative on ALK-related Malignancies (ERIA), Cambridge, UK
| | - Sylvie Giuriato
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France .,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,European Research Initiative on ALK-related Malignancies (ERIA), Cambridge, UK.,Transautophagy: European network for multidisciplinary research and translation of autophagy knowledge, COST Action CA15138, Brussel, Belgium
| | - Estelle Espinos
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France .,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 UMR1037 CRCT, F-31000, Toulouse, France.,Laboratoire d'Excellence Toulouse-Cancer-TOUCAN, F-31024 Toulouse, France.,European Research Initiative on ALK-related Malignancies (ERIA), Cambridge, UK
| |
Collapse
|
12
|
Frentzel J, Sorrentino D, Giuriato S. Targeting Autophagy in ALK-Associated Cancers. Cancers (Basel) 2017; 9:E161. [PMID: 29186933 PMCID: PMC5742809 DOI: 10.3390/cancers9120161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/17/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022] Open
Abstract
Autophagy is an evolutionarily conserved catabolic process, which is used by the cells for cytoplasmic quality control. This process is induced following different kinds of stresses e.g., metabolic, environmental, or therapeutic, and acts, in this framework, as a cell survival mechanism. However, under certain circumstances, autophagy has been associated with cell death. This duality has been extensively reported in solid and hematological cancers, and has been observed during both tumor development and cancer therapy. As autophagy plays a critical role at the crossroads between cell survival and cell death, its involvement and therapeutic modulation (either activation or inhibition) are currently intensively studied in cancer biology, to improve treatments and patient outcomes. Over the last few years, studies have demonstrated the occurrence of autophagy in different Anaplastic Lymphoma Kinase (ALK)-associated cancers, notably ALK-positive anaplastic large cell lymphoma (ALCL), non-small cell lung carcinoma (NSCLC), Neuroblastoma (NB), and Rhabdomyosarcoma (RMS). In this review, we will first briefly describe the autophagic process and how it can lead to opposite outcomes in anti-cancer therapies, and we will then focus on what is currently known regarding autophagy in ALK-associated cancers.
Collapse
Affiliation(s)
- Julie Frentzel
- Merck Serono S.A., Route de Fenil 25, Z.I. B, 1804 Corsier-sur-Vevey, Switzerland.
| | - Domenico Sorrentino
- Inserm, UMR1037, CNRS, ERL5294, Université Toulouse III-Paul Sabatier, CRCT, F-31000 Toulouse, France.
| | - Sylvie Giuriato
- Inserm, UMR1037, CNRS, ERL5294, Université Toulouse III-Paul Sabatier, CRCT, F-31000 Toulouse, France.
- European Research Initiative on ALK-related malignancies (ERIA).
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138.
| |
Collapse
|
13
|
Jacquel A, Luciano F, Puissant A, Robert G, Auberger P. [Autophagy, a key player in leukemogenesis and a therapeutic target in hematopoietic malignancies]. Med Sci (Paris) 2017; 33:226-229. [PMID: 28367807 DOI: 10.1051/medsci/20173303007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Arnaud Jacquel
- Inserm U1065 (C3M), Université Nice Côte d'Azur, Hôpital de l'Archet, bâtiment Archimed, 151 route de St-Antoine de Ginèstière, 06204 Nice, France
| | - Frédéric Luciano
- Inserm U1065 (C3M), Université Nice Côte d'Azur, Hôpital de l'Archet, bâtiment Archimed, 151 route de St-Antoine de Ginèstière, 06204 Nice, France
| | - Alexandre Puissant
- Inserm U1065 (C3M), Université Nice Côte d'Azur, Hôpital de l'Archet, bâtiment Archimed, 151 route de St-Antoine de Ginèstière, 06204 Nice, France
| | - Guillaume Robert
- Inserm U1065 (C3M), Université Nice Côte d'Azur, Hôpital de l'Archet, bâtiment Archimed, 151 route de St-Antoine de Ginèstière, 06204 Nice, France
| | - Patrick Auberger
- Inserm U1065 (C3M), Université Nice Côte d'Azur, Hôpital de l'Archet, bâtiment Archimed, 151 route de St-Antoine de Ginèstière, 06204 Nice, France
| |
Collapse
|