1
|
Kosałka-Węgiel J, Pacholczak-Madej R, Dziedzic R, Siwiec-Koźlik A, Spałkowska M, Milewski M, Zaręba L, Bazan-Socha S, Korkosz M. Malignancy in systemic lupus erythematosus: relation to disease characteristics in 92 patients - a single center retrospective study. Rheumatol Int 2024; 44:1701-1713. [PMID: 38850326 PMCID: PMC11343918 DOI: 10.1007/s00296-024-05623-3] [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: 05/07/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a variable clinical manifestation, potentially leading to death. Importantly, patients with SLE have an increased risk of neoplastic disorders. Thus, this study aimed to comprehensively evaluate the clinical and laboratory characteristics of patients with SLE and with or without malignancy. METHODS We conducted a retrospective analysis of medical records of 932 adult Caucasian patients with SLE treated at the University Hospital in Kraków, Poland, from 2012 to 2022. We collected demographic, clinical, and laboratory characteristics, but also treatment modalities with disease outcomes. RESULTS Among 932 patients with SLE, malignancy was documented in 92 (9.87%), with 7 (7.61%) patients experiencing more than one such complication. Non-hematologic malignancies were more prevalent (n = 77, 83.7%) than hematologic malignancies (n = 15, 16.3%). Patients with SLE and malignancy had a higher mean age of SLE onset and a longer mean disease duration than patients without malignancy (p < 0.001 and p = 0.027, respectively). The former group also presented more frequently with weight loss (odds ratio [OR] = 2.62, 95% confidence interval [CI] 1.61-4.23, p < 0.001), fatigue/weakness (OR = 2.10, 95% CI 1.22-3.77, p = 0.005), and fever (OR = 1.68, 95% CI 1.06-2.69, p = 0.024). In the malignancy-associated group, we noticed a higher prevalence of some clinical manifestations, such as pulmonary hypertension (OR = 3.47, 95% CI 1.30-8.42, p = 0.007), lung involvement (OR = 2.64, 95% CI 1.35-4.92, p = 0.003) with pleural effusion (OR = 2.39, 95% CI 1.43-3.94, p < 0.001), and anemia (OR = 2.24, 95% CI 1.29-4.38, p = 0.006). Moreover, the patients with SLE and malignancy more frequently had internal comorbidities, including peripheral arterial obliterans disease (OR = 3.89, 95% CI 1.86-7.75, p < 0.001), myocardial infarction (OR = 3.08, 95% CI 1.41-6.30, p = 0.003), heart failure (OR = 2.94, 95% CI 1.30-6.17, p = 0.005), diabetes mellitus (OR = 2.15, 95% CI 1.14-3.91, p = 0.011), hypothyroidism (OR = 2.08, 95% CI 1.29-3.34, p = 0.002), arterial hypertension (OR = 1.97, 95% CI 1.23-3.23, p = 0.003), and hypercholesterolemia (OR = 1.87, 95% CI 1.18-3.00, p = 0.006). Patients with SLE and malignancy were treated more often with aggressive immunosuppressive therapies, including cyclophosphamide (OR = 2.07, 95% CI 1.30-3.28, p = 0.002), however median cumulative cyclophosphamide dose in malignancy-associated SLE subgroup was 0 g (0-2 g). Interestingly, over a median follow-up period of 14 years (ranges: 8-22 years) a total of 47 patients with SLE died, with 16 cases (5.28%) in the malignancy-associated SLE group and 31 cases (5.73%) in the non-malignancy SLE group (p = 0.76). The most common causes of death were infections (21.28%) and SLE exacerbation (8.51%). CONCLUSION The study highlights the relatively frequent presence of malignancies in patients with SLE, a phenomenon that demands oncological vigilance, especially in patients with a severe clinical course and comorbidities, to improve long-term outcomes in these patients.
Collapse
Affiliation(s)
- Joanna Kosałka-Węgiel
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland.
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland.
| | - Renata Pacholczak-Madej
- Department of Gynaecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, Kraków, 31-115, Poland
- Department of Chemotherapy, The District Hospital, Szpitalna 22, Sucha Beskidzka, 34-200, Poland
- Department of Anatomy, Jagiellonian University Medical College, Kopernika 12, Kraków, 31-034, Poland
| | - Radosław Dziedzic
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Św. Łazarza 16, Kraków, 31-530, Poland
| | - Andżelika Siwiec-Koźlik
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Magdalena Spałkowska
- Department of Dermatology, Jagiellonian University Medical College, Botaniczna 3, Kraków, 31-501, Poland
| | - Mamert Milewski
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Lech Zaręba
- College of Natural Sciences, Institute of Computer Science, University of Rzeszów, Pigonia 1, Rzeszów, 35-310, Poland
| | - Stanisława Bazan-Socha
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
| |
Collapse
|
2
|
Wahyudianingsih R, Sanjaya A, Jonathan T, Pranggono EH, Achmad D, Hernowo BS. Chemotherapy's effects on autophagy in the treatment of Hodgkin's lymphoma: a scoping review. Discov Oncol 2024; 15:269. [PMID: 38976168 PMCID: PMC11231119 DOI: 10.1007/s12672-024-01142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Classical Hodgkin Lymphomas (HL) are a unique malignant growth with an excellent initial prognosis. However, 10-30% of patients will still relapse after remission. One primary cellular function that has been the focus of tumor progression is autophagy. This process can preserve cellular homeostasis under stressful conditions. Several studies have shown that autophagy may play a role in developing HL. Therefore, this review aimed to explore chemotherapy's effect on autophagy in HL, and the effects of autophagy on HL. METHODS A scoping review in line with the published PRISMA extension for scoping reviews (PRISMA-ScR) was conducted. A literature search was conducted on the MEDLINE database and the Cochrane Central Register of Controlled Trials (CENTRAL). All results were retrieved and screened, and the resulting articles were synthesized narratively. RESULTS The results showed that some cancer chemotherapy also induces autophagic flux. Although the data on HL is limited, since the mechanisms of action of these drugs are similar, we can infer a similar relationship. However, this increased autophagy activity may reflect a mechanism for increasing tumor growth or a cellular compensation to inhibit its growth. Although evidence supports both views, we argued that autophagy allowed cancer cells to resist cell death, mainly due to DNA damage caused by cytotoxic drugs. CONCLUSION Autophagy reflects the cell's adaptation to survive and explains why chemotherapy generally induces autophagy functions. However, further research on autophagy inhibition is needed as it presents a viable treatment strategy, especially against drug-resistant populations that may arise from HL chemotherapy regimens.
Collapse
Affiliation(s)
- Roro Wahyudianingsih
- Postgraduate Program of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
- Department of Anatomical Pathology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Ardo Sanjaya
- Department of Anatomy, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia.
| | - Timothy Jonathan
- Undergraduate Program in Medicine, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia
| | - Emmy Hermiyanti Pranggono
- Department of Internal Medicine, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| | - Dimyati Achmad
- Department of Oncological Surgery, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| | - Bethy Suryawathy Hernowo
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| |
Collapse
|
3
|
Gustafson DL, Viola LO, Towers CG, Das S, Duval DL, Van Eaton KM. Sensitivity of osteosarcoma cell lines to autophagy inhibition as determined by pharmacologic and genetic manipulation. Vet Comp Oncol 2023; 21:726-738. [PMID: 37724007 PMCID: PMC11470750 DOI: 10.1111/vco.12937] [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: 03/22/2023] [Revised: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023]
Abstract
Pharmacologic inhibition of autophagy can be achieved using lysosomotropic agents such as hydroxychloroquine (HCQ) that interfere with fusion of the autophagosome to the lysosome thus preventing completion of the recycling process. The goal of the present study is to determine the sensitivity of eight canine (cOSA) and four human (hOSA) osteosarcoma tumour cell lines to antiproliferative and cytotoxic effects of lysosomal autophagy inhibitors, and to compare these results to the autophagy-dependence measured using a CRISPR/Cas9 live-cell imaging assay in OSA and other tumour cell lines. Antiproliferative and cytotoxic response to HCQ and Lys05 was determined using live cell imaging and YOYO-1 staining. CRISPR/Cas9 live cell imaging screen was done using species specific guide RNA's and transfection of reagents into cells. Response to autophagy core genes was compared to response to an essential (PCNA) and non-essential (FOXO3A) gene. cOSA and hOSA cell lines showed similar antiproliferative and cytotoxic responses to HCQ and Lys05 with median lethal dose (Dm ) values ranging from 4.6-15.8 μM and 2.1-5.1 μM for measures of anti-proliferative response, respectively. A relationship was observed between antiproliferative responses to HCQ and Lys05 and VPS34 CRISPR score with Dm values correlating with VPS34 response (r = 0.968 and 0.887) in a species independent manner. The results show that a subset of cOSA and hOSA cell lines are autophagy-dependent and sensitive to HCQ at pharmacologically-relevant exposures.
Collapse
Affiliation(s)
- Daniel L. Gustafson
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Lindsey O. Viola
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Christina G. Towers
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Sunetra Das
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Dawn L. Duval
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Kristen M. Van Eaton
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
4
|
Baeva ME, Camara-Lemarroy C. The role of autophagy protein Atg5 in multiple sclerosis. Mult Scler Relat Disord 2023; 79:105029. [PMID: 37778158 DOI: 10.1016/j.msard.2023.105029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/08/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
Multiple sclerosis (MS) is a neurological disease which has a strong autoimmune component to its pathology. Although there are currently many approved immunomodulatory treatments that reduce the rate of relapse and slow down the progression of the disease, the cure is still elusive. This may be due to the underlying etiology still being unknown. Autophagy is the potential link between neurodegeneration and autoimmunity. Specifically, this review will focus on the autophagy protein Atg5 and examine the in vitro cell culture, animal and human studies that have examined its expression and effects in the context of MS. The findings of these investigations are summarized, and a model is proposed in which elevated Atg5 levels leads to dysfunctional autophagy, neurodegeneration, inflammation, and eventually clinical disability. While there are currently no drugs that specifically target Atg5, our review recommends that further investigations into the role that Atg5 plays in MS pathophysiology may eventually lead to the development of autophagy-specific treatments of MS.
Collapse
Affiliation(s)
- Maria-Elizabeth Baeva
- Department of Clinical Neurosciences, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada.
| | - Carlos Camara-Lemarroy
- Department of Clinical Neurosciences, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada
| |
Collapse
|
5
|
Ye W, Fan C, Fu K, Wang X, Lin J, Nian S, Liu C, Zhou W. The SAR and action mechanisms of autophagy inhibitors that eliminate drug resistance. Eur J Med Chem 2022; 244:114846. [DOI: 10.1016/j.ejmech.2022.114846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/02/2022] [Accepted: 10/10/2022] [Indexed: 11/03/2022]
|
6
|
Troumpoukis D, Papadimitropoulou A, Charalampous C, Kogionou P, Palamaris K, Sarantis P, Serafimidis I. Targeting autophagy in pancreatic cancer: The cancer stem cell perspective. Front Oncol 2022; 12:1049436. [PMID: 36505808 PMCID: PMC9730023 DOI: 10.3389/fonc.2022.1049436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Pancreatic cancer is currently the seventh leading cause of cancer-related deaths worldwide, with the estimated death toll approaching half a million annually. Pancreatic ductal adenocarcinoma (PDAC) is the most common (>90% of cases) and most aggressive form of pancreatic cancer, with extremely poor prognosis and very low survival rates. PDAC is initiated by genetic alterations, usually in the oncogene KRAS and tumor suppressors CDKN2A, TP53 and SMAD4, which in turn affect a number of downstream signaling pathways that regulate important cellular processes. One of the processes critically altered is autophagy, the mechanism by which cells clear away and recycle impaired or dysfunctional organelles, protein aggregates and other unwanted components, in order to achieve homeostasis. Autophagy plays conflicting roles in PDAC and has been shown to act both as a positive effector, promoting the survival of pancreatic tumor-initiating cells, and as a negative effector, increasing cytotoxicity in uncontrollably expanding cells. Recent findings have highlighted the importance of cancer stem cells in PDAC initiation, progression and metastasis. Pancreatic cancer stem cells (PaCSCs) comprise a small subpopulation of the pancreatic tumor, characterized by cellular plasticity and the ability to self-renew, and autophagy has been recognised as a key process in PaCSC maintenance and function, simultaneously suggesting new strategies to achieve their selective elimination. In this review we evaluate recent literature that links autophagy with PaCSCs and PDAC, focusing our discussion on the therapeutic implications of pharmacologically targeting autophagy in PaCSCs, as a means to treat PDAC.
Collapse
Affiliation(s)
- Dimitrios Troumpoukis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Chrysanthi Charalampous
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Paraskevi Kogionou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Kostas Palamaris
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Serafimidis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece,*Correspondence: Ioannis Serafimidis,
| |
Collapse
|
7
|
Wu L, Chatla S, Lin Q, Chowdhury FA, Geldenhuys W, Du W. Quinacrine-CASIN combination overcomes chemoresistance in human acute lymphoid leukemia. Nat Commun 2021; 12:6936. [PMID: 34836965 PMCID: PMC8626516 DOI: 10.1038/s41467-021-27300-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/11/2021] [Indexed: 01/30/2023] Open
Abstract
Chemoresistance posts a major hurdle for treatment of acute leukemia. There is increasing evidence that prolonged and intensive chemotherapy often fails to eradicate leukemic stem cells, which are protected by the bone marrow niche and can induce relapse. Thus, new therapeutic approaches to overcome chemoresistance are urgently needed. By conducting an ex vivo small molecule screen, here we have identified Quinacrine (QC) as a sensitizer for Cytarabine (AraC) in treating acute lymphoblastic leukemia (ALL). We show that QC enhances AraC-mediated killing of ALL cells, and subsequently abrogates AraC resistance both in vitro and in an ALL-xenograft model. However, while combo AraC+QC treatment prolongs the survival of primary transplanted recipients, the combination exhibits limited efficacy in secondary transplanted recipients, consistent with the survival of niche-protected leukemia stem cells. Introduction of Cdc42 Activity Specific Inhibitor, CASIN, enhances the eradication of ALL leukemia stem cells by AraC+QC and prolongs the survival of both primary and secondary transplanted recipients without affecting normal long-term human hematopoiesis. Together, our findings identify a small-molecule regimen that sensitizes AraC-mediated leukemia eradication and provide a potential therapeutic approach for better ALL treatment.
Collapse
Affiliation(s)
- Limei Wu
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, USA
| | - Srinivas Chatla
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Qiqi Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, USA
| | - Fabliha Ahmed Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, USA
- Molecular Pharmacology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Werner Geldenhuys
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, USA
| | - Wei Du
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, USA.
- Molecular Pharmacology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
8
|
Guo J, Ren Z, Li J, Li T, Liu S, Yu Z. The relationship between cancer and medication exposure in patients with systemic lupus erythematosus: a nested case-control study. Arthritis Res Ther 2020; 22:159. [PMID: 32586407 PMCID: PMC7318532 DOI: 10.1186/s13075-020-02228-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/02/2020] [Indexed: 11/10/2022] Open
Abstract
Background Systemic lupus erythematosus (SLE) is associated with increased risk of cancer and the mechanism remains unclear. Here, we examined the level of auto-antibodies and disease activity index scores in SLE patients with cancers and analyzed whether medications for SLE management might contribute to the higher cancer risk in SLE patients. Methods In this retrospective study, we carried out a nested case-control study in a large cohort of SLE patients. We screened 5858 SLE patients to identify the newly diagnosed and yet to be treated cancers. The following clinical features were evaluated: auto-antibodies levels, SLE disease activity index scores, and previous medication used for SLE management. Systemic glucocorticoid, cyclophosphamide, hydroxychloroquine (HCQ), methotrexate, and azathioprine were considered the main medication indices. Results Our analyses identified 51 SLE patients who also had cancer and 204 matched control patients who had SLE but not cancer. Of the 51 SLE patients, thyroid cancer (14/51, 27.45%), cervical cancer (10/51, 19.61%), and lung cancer (7/51, 13.73%) were the most common types. Our analyses did not reveal any significant differences in the levels of auto-antibodies in SLE patients with cancers relative to the control group. Further, we observed that disease activity was significantly lower in SLE patients with cancers relative to the matched control SLE group. There was no statistically significant association between the cancer risk and the use of systemic glucocorticoid, cyclophosphamide, methotrexate, or azathioprine. Importantly, the administration of HCQ was significantly lower in SLE patients suffering cancers relative to the cancer-free matched control group. Conclusions Our analyses indicate that SLE patients with cancers might have a lower disease activity at the time of cancer diagnosis. HCQ was negatively associated with cancer risk in SLE patients. These findings highlight a potential and novel prevention strategy for SLE.
Collapse
Affiliation(s)
- Jinyan Guo
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Zhigang Ren
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Jianhao Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Tianfang Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, China
| | - Shengyun Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, China.
| | - Zujiang Yu
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan Province, China.
| |
Collapse
|
9
|
Li G, Zheng YH, Xu L, Feng J, Tang HL, Luo C, Song YP, Chen XQ. BRD4 inhibitor nitroxoline enhances the sensitivity of multiple myeloma cells to bortezomib in vitro and in vivo by promoting mitochondrial pathway-mediated cell apoptosis. Ther Adv Hematol 2020; 11:2040620720932686. [PMID: 32551032 PMCID: PMC7281877 DOI: 10.1177/2040620720932686] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022] Open
Abstract
Background Multiple myeloma (MM) is the second most common hematological neoplasm. Wide administration of bortezomib significantly improves the survival of MM patients compared with conventional chemotherapy. Bromodomain-containing protein 4 (BRD4) inhibitors also have been demonstrated to retard cell proliferation and induce cellular apoptosis in various cancers. However, it is unclear whether the BRD4 inhibitor nitroxoline plus bortezomib has a synergistic anti-tumor effect on MM. Methods Cell viability was determined via 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell cycle and cell apoptosis were assessed via flow cytometry. Protein expression levels were determined via western blotting. The expression of apoptosis-related proteins in xenograft tissue were detected by means of immunohistochemistry. Results Treatment with nitroxoline or bortezomib suppressed cell proliferation, and caused G0/G1 phase arrest and apoptosis in H929 and RPMI8226 cells. Furthermore, nitroxoline intensified the retardation of cell proliferation, as well as further enhanced the G0/G1 phase arrest and apoptosis induced by bortezomib in H929 and RPMI8226 cells. The western blot analysis revealed that nitroxoline or bortezomib treatment markedly diminished the levels of Bcl-2 and cyclin D1, and increased the levels of p21, Bax, cleaved PARP and cleaved caspase-3. Combination of these two agents was observed to result in further marked changes on these levels compared with nitroxoline or bortezomib treatment alone. What is more, in the xenograft tumor model, combinative treatment markedly inhibited tumor growth compared with the single drug treatment. Conclusion Combination of bortezomib with nitroxoline has a synergistic anti-tumor activity in MM cells and may be a novel treatment method for MM.
Collapse
Affiliation(s)
- Guang Li
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan-Hua Zheng
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Li Xu
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Juan Feng
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hai-Long Tang
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Cheng Luo
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Yan-Ping Song
- Institute of Hematology, Xi'an Central Hospital, 161 Xiwu Road, Xi'an, Shaanxi 710003, P.R. China
| | - Xie-Qun Chen
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
10
|
Baek JH, Yun HS, Kim JY, Lee J, Lee YJ, Lee CW, Song JY, Ahn J, Park JK, Kim JS, Lee KH, Kim EH, Hwang SG. Kinesin light chain 4 as a new target for lung cancer chemoresistance via targeted inhibition of checkpoint kinases in the DNA repair network. Cell Death Dis 2020; 11:398. [PMID: 32457423 PMCID: PMC7250887 DOI: 10.1038/s41419-020-2592-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
The poor therapeutic efficacy of non-small cell lung cancer (NSCLC) is partly attributed to the acquisition of chemoresistance. To investigate the mechanism underlying this resistance, we examined the potential link between kinesin light chain 4 (KLC4), which we have previously reported to be associated with radioresistance in NSCLC, and sensitivity to chemotherapy in human lung cancer cell lines. KLC4 protein levels in lung cancer cells correlated with the degree of chemoresistance to cisplatin treatment. Furthermore, KLC4 silencing enhanced the cytotoxic effect of cisplatin by promoting DNA double-strand breaks and apoptosis. These effects were mediated by interaction with the checkpoint kinase CHK2, as KLC4 knockdown increased CHK2 activation, which was further enhanced in combination with cisplatin treatment. In addition, KLC4 and CHEK2 expression levels showed negative correlation in lung tumor samples from patients, and KLC4 overexpression correlated negatively with survival. Our results indicate a novel link between the KLC4 and CHK2 pathways regulating DNA damage response in chemoresistance, and highlight KLC4 as a candidate for developing lung cancer-specific drugs and customized targeted molecular therapy.
Collapse
Affiliation(s)
- Jeong-Hwa Baek
- Radiation Biology Research Team, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, Republic of Korea
| | - Hong Shik Yun
- Radiation Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ju-Young Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 440-746, Korea
| | - Janet Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Yeon-Joo Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 440-746, Korea
| | - Jie-Young Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jiyeon Ahn
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jong Kuk Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Kee-Ho Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, 33, 17-gil, Duryugongwon-ro, Nam-gu, Daegu, Korea.
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea.
| |
Collapse
|
11
|
Chidamide epigenetically represses autophagy and exerts cooperative antimyeloma activity with bortezomib. Cell Death Dis 2020; 11:297. [PMID: 32341332 PMCID: PMC7186232 DOI: 10.1038/s41419-020-2414-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022]
Abstract
Autophagy and ubiquitin proteasome system are two distinct and cooperative proteolytic pathways. The dual-pathway suppression represents a promising therapeutic strategy for multiple myeloma. Chidamide is a novel benzamide inhibitor of histone deacetylase, and shows potent antimyeloma activity. Here, we revealed the autophagy-suppressive role of chidamide in myeloma cells. We then demonstrated that chidamide treatment markedly downregulated histone deacetylase SIRT1, and simultaneously resulted in dose-dependent upregulation of acetyltransferase hMOF and histone methyltransferase EZH2, which contributed to an increase in global levels of histone H4 lysine 16 acetylation (H4k16ac) and histone H3 lysine 27 trimethylation (H3k27me3). We next confirmed concomitant upregulation of H4k16ac and H3k27me3 in the same promoter regions of the autophagy-related gene LC3B, reinforcing the specific roles for H4k16ac and H3k27me3 in mediating chidamide-induced transcriptional repression of LC3B. Finally, we provided experimental evidence that co-treatment with chidamide and proteasome inhibitor bortezomib induced clear synergistic cytotoxicity against MM cells, which was associated with increased accumulation of ubiquitinated proteins and excessive endoplasmic reticulum stress or dysregulated unfolded protein response. Our results altogether suggest that chidamide cooperatively potentiates antimyeloma activity of bortezomib, at least in part, by epigenetically repressing autophagic degradation of ubiquitinated proteins.
Collapse
|
12
|
Vu M, Kassouf N, Ofili R, Lund T, Bell C, Appiah S. Doxorubicin selectively induces apoptosis through the inhibition of a novel isoform of Bcl‑2 in acute myeloid leukaemia MOLM‑13 cells with reduced Beclin 1 expression. Int J Oncol 2020; 57:113-121. [PMID: 32377726 PMCID: PMC7252449 DOI: 10.3892/ijo.2020.5052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
The overexpression of anti-apoptotic Bcl-2 in acute myeloid leukaemia (AML) may contribute to difficulties in eradicating these cells during chemotherapy. In the present study, doxorubicin (Dox) was evaluated for its potential to induce selective apoptotic cell death in AML MOLM-13 cells and to modulate autophagy through Bcl-2 and Beclin 1 protein expression. Annexin V/propidium iodide and 5(6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) flow cytometric analyses were conducted to determine the effects of Dox on cell death and cell proliferation, respectively, following 48 h of co-incubation with AML MOLM-13 or U-937 monocytic cells. The protein expression levels of Bcl-2 and Beclin 1 in untreated and treated cells were quantified by western blot analysis. Dox reduced the viability of MOLM-13 cells partly by inhibiting cell division and inducing cell apoptosis. Dox demonstrated a level of selectivity in its cytotoxicity against MOLM-13 compared to U-937 cells (P<0.05). Dox induced a significant decrease in Beclin 1 protein levels in MOLM-13 cells without significantly affecting the protein levels in U-937 monocytes. A novel Bcl-2 15-20 kDa (p15-20-Bcl-2) isoform was found to be selectively expressed in AML MOLM-13 cells (but absent in the leukaemic cell lines tested, OCI-AML2, CML K562 and U-937). Dox induced a highly significant inhibition of p15-20-Bcl-2 at concentrations of 0.5, 0.75 and 1 µM (P<0.01). However, the usual 26 kDa Bcl-2 (p26-Bcl-2-α) isoform protein expression was not affected by the drug in either the MOLM-13 or U-937 cells. It was thus postulated that Dox exhibited some selectivity by targeting the p15-20-Bcl-2 isoform in MOLM-13 cells and activating Beclin 1 to induce cell death.
Collapse
Affiliation(s)
- Milan Vu
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| | - Nick Kassouf
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| | - Rosemary Ofili
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| | - Torben Lund
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| | - Celia Bell
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| | - Sandra Appiah
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| |
Collapse
|
13
|
Gajek A, Poczta A, Łukawska M, Cecuda-Adamczewska V, Tobiasz J, Marczak A. Chemical modification of melphalan as a key to improving treatment of haematological malignancies. Sci Rep 2020; 10:4479. [PMID: 32161295 PMCID: PMC7066245 DOI: 10.1038/s41598-020-61436-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 02/24/2020] [Indexed: 01/18/2023] Open
Abstract
Chemical modification of known, effective drugs is one method to improve chemotherapy. Thus, the object of this study was to generate melphalan derivatives with improved cytotoxic activity in human cancer cells (RPMI8226, HL60 and THP1). Several melphalan derivatives were synthesised, modified in their two important functional groups. Nine analogues were tested, including melphalan compounds modified: only at the amino group, by replacing the amine with an amidine group containing a morpholine ring (MOR-MEL) or with an amidino group and dipropyl chain (DIPR-MEL); only at the carboxyl group to form methyl and ethyl esters of melphalan (EM-MEL, EE-MEL); and in a similar manner at both functional groups (EM-MOR-MEL, EE-MOR-MEL, EM-DIPR-MEL, EE-DIPR-MEL). Melphalan derivatives were evaluated for cytotoxicity (resazurin viability assay), genotoxicity (comet assay) and the ability to induce apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labelling, TUNEL, phosphatidylserine externalisation, chromatin condensation, activity of caspases 3/7, 8 and 9 and intracellular concentration of calcium ions) in comparison with the parent drug. Almost all derivatives, with the exception of MOR-MEL and DIPR-MEL, were found to be more toxic than melphalan in all cell lines evaluated. Treatment of cultures with the derivatives generated a significant higher level of DNA breaks compared to those treated with melphalan, especially after longer incubation times. In addition, all the melphalan derivatives demonstrated a high apoptosis-inducing ability in acute monocytic and promyelocytic leukemia cells. This study showed that the mechanism of action of the tested compounds differed depending on the cell line, and allowed the selection of the most active compounds for further, more detailed investigations.
Collapse
Affiliation(s)
- Arkadiusz Gajek
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| | - Anastazja Poczta
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Małgorzata Łukawska
- ŁUKASIEWICZ Research Network-Institute of Biotechnology and Antibiotics, 5 Staroscinska St., 02-516, Warsaw, Poland
| | - Violetta Cecuda-Adamczewska
- ŁUKASIEWICZ Research Network-Institute of Biotechnology and Antibiotics, 5 Staroscinska St., 02-516, Warsaw, Poland
| | - Joanna Tobiasz
- ŁUKASIEWICZ Research Network-Institute of Biotechnology and Antibiotics, 5 Staroscinska St., 02-516, Warsaw, Poland
| | - Agnieszka Marczak
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| |
Collapse
|
14
|
Gu XY, Jiang Y, Li MQ, Han P, Liu YL, Cui BB. Over-expression of EGFR regulated by RARA contributes to 5-FU resistance in colon cancer. Aging (Albany NY) 2020; 12:156-177. [PMID: 31896739 PMCID: PMC6977699 DOI: 10.18632/aging.102607] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
A promising new strategy for cancer therapy is to target the autophagic pathway. However, comprehensive characterization of autophagy genes and their clinical relevance in cancer is still lacking. Here, we systematically characterized alterations of autophagy genes in multiple cancer lines by analyzing data from The Cancer Genome Atlas and CellMiner database. Interactions between autophagy genes and clinically actionable genes (CAGs) were identified by analyzing co-expression, protein-protein interactions (PPIs) and transcription factor (TF) data. A key subnetwork was identified that included 18 autophagy genes and 22 CAGs linked by 28 PPI pairs and 1 TF-target pair, which was EGFR targeted by RARA. Alterations in the expression of autophagy genes were associated with patient survival in multiple cancer types. RARA and EGFR were associated with worse survival in colorectal cancer patients. The regulatory role of EGFR in 5-FU resistance was validated in colon cancer cells in vivo and in vitro. EGFR contributed to 5-FU resistance in colon cancer cells through autophagy induction, and EGFR overexpression in 5-FU resistant colon cancer was regulated by RARA. The present study provides a comprehensive analysis of autophagy in different cancer cell lines and highlights the potential clinical utility of targeting autophagy genes.
Collapse
Affiliation(s)
- Xin-Yue Gu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, People's Republic of China
| | - Yang Jiang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150040, People's Republic of China
| | - Ming-Qi Li
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, People's Republic of China
| | - Peng Han
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, People's Republic of China
| | - Yan-Long Liu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, People's Republic of China
| | - Bin-Bin Cui
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin 150040, People's Republic of China
| |
Collapse
|
15
|
Shang J, Chen WM, Liu S, Wang ZH, Wei TN, Chen ZZ, Wu WB. CircPAN3 contributes to drug resistance in acute myeloid leukemia through regulation of autophagy. Leuk Res 2019; 85:106198. [DOI: 10.1016/j.leukres.2019.106198] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 07/07/2019] [Accepted: 08/01/2019] [Indexed: 12/24/2022]
|
16
|
Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma. Cells 2019; 8:cells8020103. [PMID: 30704144 PMCID: PMC6406467 DOI: 10.3390/cells8020103] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Haematopoiesis is a tightly orchestrated process where a pool of hematopoietic stem and progenitor cells (HSPCs) with high self-renewal potential can give rise to both lymphoid and myeloid lineages. The HSPCs pool is reduced with ageing resulting in few HSPC clones maintaining haematopoiesis thereby reducing blood cell diversity, a phenomenon called clonal haematopoiesis. Clonal expansion of HSPCs carrying specific genetic mutations leads to increased risk for haematological malignancies. Therefore, it comes as no surprise that hematopoietic tumours develop in higher frequency in elderly people. Unfortunately, elderly patients with leukaemia or lymphoma still have an unsatisfactory prognosis compared to younger ones highlighting the need to develop more efficient therapies for this group of patients. Growing evidence indicates that macroautophagy (hereafter referred to as autophagy) is essential for health and longevity. This review is focusing on the role of autophagy in normal haematopoiesis as well as in leukaemia and lymphoma development. Attenuated autophagy may support early hematopoietic neoplasia whereas activation of autophagy in later stages of tumour development and in response to a variety of therapies rather triggers a pro-tumoral response. Novel insights into the role of autophagy in haematopoiesis will be discussed in light of designing new autophagy modulating therapies in hematopoietic cancers.
Collapse
|
17
|
Xu J, Su Y, Xu A, Fan F, Mu S, Chen L, Chu Z, Zhang B, Huang H, Zhang J, Deng J, Ai L, Sun C, Hu Y. miR-221/222-Mediated Inhibition of Autophagy Promotes Dexamethasone Resistance in Multiple Myeloma. Mol Ther 2019; 27:559-570. [PMID: 30765325 DOI: 10.1016/j.ymthe.2019.01.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/26/2022] Open
Abstract
Inherent or acquired resistance to chemotherapeutic drugs is still an obstacle for the treatment of multiple myeloma (MM). MicroRNA dysregulation is related to the development of chemoresistance in cancers. However, its role in chemoresistance of MM is largely unknown. Here we demonstrated that miR-221/222 were upregulated in plasma cells from patients with MM, especially those with relapsed or refractory disease. Moreover, expression levels of miR-221/222 were inversely correlated with dexamethasone (Dex) sensitivity of human MM cell lines. Importantly, we found that Dex induced pro-death autophagy in MM cells and the inhibition of autophagy significantly decreased Dex-induced cell death. Mechanistically, autophagy-related gene 12 (ATG12) was identified as a novel target gene of miR-221/222, and miR-221/222 overexpression inhibited autophagy by directly targeting ATG12 and the p27kip (p27)-mammalian target of rapamycin (mTOR) pathway. Indeed, Dex treatment decreased the expression of miR-221/222, thereby activating the ATG12/p27-mTOR autophagy-regulatory axis and inducing cell death in Dex-sensitive MM cells. Furthermore, both in vitro and in vivo results showed that the inhibitions of miR-221/222 increased the expression of ATG12 and p27 and functionally induced extended autophagy and cell death of MM cells. In conclusion, our findings demonstrated the crucial role of the miR-221/222-ATG12/p27-mTOR autophagy-regulatory axis in Dex resistance of MM, and they suggest potential prediction and treatment strategies for glucocorticoid resistance.
Collapse
Affiliation(s)
- Jian Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Su
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fengjuan Fan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shidai Mu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangbo Chu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haifan Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiasi Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
18
|
Shang J, Chen ZZ, Wang ZH, Wei TN, Wu WB, Chen WM. [Association of miRNA-196b-5p and miRNA-99a-5p with autophagy and apoptosis in multiple myeloma cells]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:766-772. [PMID: 30369190 PMCID: PMC7342263 DOI: 10.3760/cma.j.issn.0253-2727.2018.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Indexed: 12/15/2022]
Abstract
Objective: To investigate the relationship between miRNA-196b-5p and miRNA-99a-5p expression and autophagy and apoptosis in multiple myeloma cells. Methods: Human myeloma cell line U266 and normal CD138+ plasma cells were selected as the research objects. The subjects were divided into 45 cases of multiple myeloma patients and 40 healthy controls. The expression of miRNA-196b-5p and miRNA-99a-5p was measured by real-time quantitative PCR, and Western blot was used to determine the expression of autophagy related protein LC3-Ⅱ, LC3-Ⅰ, P62, Beclin-1 expression, apoptosis related protein CL caspase3, CL caspase7, Bcl-2, Bax, and TGF-β/Smad pathway associated proteins TGF-β1, Smad2/3, p-Smad3 and Smad7. The cell apoptosis rate was determined by flow cytometry. The correlation between miRNA expression level and clinical characteristics of multiple myeloma patients was analyzed. Results: Compared with normal plasma cells, the expression of miRNA-196b-5p in myeloma cells increased significantly (0.43±0.15 vs 2.44±0.63 or 2.02±0.85, all P<0.001), the expression of miRNA-99a-5p was significantly decreased (1.87±0.61 vs 0.62±0.15 or 0.80±0.33, P<0.001), LC3-Ⅱ/LC3-Ⅰ increased significantly (P<0.05), Beclin-1 expression increased significantly (P<0.05), P62 expression decreased significantly (P<0.05). The expression of Bax, CL caspase3 and CL caspase7 decreased significantly (P<0.05), and the expression of Bcl-2 increased significantly (P<0.05) and apoptosis rate significantly decreased (P<0.05). After transfected with miRNA-196b-5p mimic or miRNA-99a-5p inhibitor, the LC3-Ⅱ/LC3-Ⅰ of CD138+ plasma cells increased significantly (P<0.05), the expression of Beclin-1 increased significantly (P<0.05), P62 expression decreased significantly (P<0.05), and the apoptosis rate significantly decreased (P<0.05). However, after autophagy inhibitor of 3-MA was administered, the apoptotic rate of the above reaction system did not change significantly (P>0.05). The expression of miRNA-196b-5p and miRNA-99a-5p was significantly correlated with DS and ISS stage in multiple myeloma patients (P<0.05). Conclusion: miRNA-196b-5p and miRNA-99a-5p are closely related to the clinical characteristics of patients with multiple myeloma. The overexpression of miRNA-196b-5p and down regulation of miRNA-99a-5p could inhibit the apoptosis of myeloma cells by up regulation of autophagy, and the mechanism is related to the activation of the TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- J Shang
- Fujian Medical University Shengli Clinical Medical College, Department of Hematology, Fujian Provincial Hospital, Fuzhou 350001, China
| | | | | | | | | | | |
Collapse
|
19
|
Onorati A, Dyczynski M, Ojha R, Amaravadi RK. Targeting autophagy in cancer. Cancer 2018; 124:3307-3318. [PMID: 29671878 PMCID: PMC6108917 DOI: 10.1002/cncr.31335] [Citation(s) in RCA: 502] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/15/2018] [Accepted: 02/16/2018] [Indexed: 12/16/2022]
Abstract
Autophagy is a conserved, self-degradation system that is critical for maintaining cellular homeostasis during stress conditions. Dysregulated autophagy has implications in health and disease. Specifically, in cancer, autophagy plays a dichotomous role by inhibiting tumor initiation but supporting tumor progression. Early results from clinical trials that repurposed hydroxychloroquine for cancer have suggested that autophagy inhibition may be a promising approach for advanced cancers. In this review of the literature, the authors present fundamental advances in the biology of autophagy, approaches to targeting autophagy, the preclinical rationale and clinical experience with hydroxychloroquine in cancer clinical trials, the potential role of autophagy in tumor immunity, and recent developments in next-generation autophagy inhibitors that have clinical potential. Autophagy is a promising target for drug development in cancer. Cancer 2018. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Angelique Onorati
- Abramson Cancer Center and Department of Medicine; University of Pennsylvania, Philadelphia, PA, 19104; U.S.A
| | - Matheus Dyczynski
- Department of Oncology & Pathology Karolinska Institute, Stockholm, Sweden
| | - Rani Ojha
- Abramson Cancer Center and Department of Medicine; University of Pennsylvania, Philadelphia, PA, 19104; U.S.A
| | - Ravi K. Amaravadi
- Abramson Cancer Center and Department of Medicine; University of Pennsylvania, Philadelphia, PA, 19104; U.S.A
| |
Collapse
|
20
|
Marcucci F, Rumio C. How Tumor Cells Choose Between Epithelial-Mesenchymal Transition and Autophagy to Resist Stress-Therapeutic Implications. Front Pharmacol 2018; 9:714. [PMID: 30013478 PMCID: PMC6036460 DOI: 10.3389/fphar.2018.00714] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/12/2018] [Indexed: 12/17/2022] Open
Abstract
Tumor cells undergo epithelial-mesenchymal transition (EMT) or macroautophagy (hereafter autophagy) in response to stressors from the microenvironment. EMT ensues when stressors act on tumor cells in the presence of nutrient sufficiency, and mechanistic target of rapamycin (mTOR) appears to be the crucial signaling node for EMT induction. Autophagy, on the other hand, is induced in the presence of nutrient deprivation and/or stressors from the microenvironment with 5' adenosine monophosphate-activated protein kinase (AMPK) playing an important, but not exclusive role, in autophagy induction. Importantly, mTOR and EMT on one hand, and AMPK and autophagy on the other hand, negatively regulate each other. Such regulation occurs at different levels and suggests that, in many instances, these two stress responses are mutually exclusive. Nevertheless, EMT and autophagy are able to interconvert and we suggest that this may depend on spatiotemporal changes in the tumor microenvironment and/or on duration/intensity of the stressor signal(s). Eventually, we propose a three-pronged therapeutic approach aimed at targeting these three major tumor cell populations. First, cytotoxic drugs that act on differentiated and proliferating tumor cells and which, per se, may promote induction of EMT or autophagy in surviving tumor cells. Second, inhibitors of mTOR in order to prevent EMT induction. Third inducers of autophagic cell death (autosis) in order to deplete tumor cells that are constitutively in an autophagic state or are induced to enter an autophagic state in response to antitumor therapy.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Stebbing J, Shah K, Lit LC, Gagliano T, Ditsiou A, Wang T, Wendler F, Simon T, Szabó KS, O'Hanlon T, Dean M, Roslani AC, Cheah SH, Lee SC, Giamas G. LMTK3 confers chemo-resistance in breast cancer. Oncogene 2018; 37:3113-3130. [PMID: 29540829 PMCID: PMC5992129 DOI: 10.1038/s41388-018-0197-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/31/2022]
Abstract
Lemur tyrosine kinase 3 (LMTK3) is an oncogenic kinase that is involved in different types of cancer (breast, lung, gastric, colorectal) and biological processes including proliferation, invasion, migration, chromatin remodeling as well as innate and acquired endocrine resistance. However, the role of LMTK3 in response to cytotoxic chemotherapy has not been investigated thus far. Using both 2D and 3D tissue culture models, we found that overexpression of LMTK3 decreased the sensitivity of breast cancer cell lines to cytotoxic (doxorubicin) treatment. In a mouse model we showed that ectopic overexpression of LMTK3 decreases the efficacy of doxorubicin in reducing tumor growth. Interestingly, breast cancer cells overexpressing LMTK3 delayed the generation of double strand breaks (DSBs) after exposure to doxorubicin, as measured by the formation of γH2AX foci. This effect was at least partly mediated by decreased activity of ataxia-telangiectasia mutated kinase (ATM) as indicated by its reduced phosphorylation levels. In addition, our RNA-seq analyses showed that doxorubicin differentially regulated the expression of over 700 genes depending on LMTK3 protein expression levels. Furthermore, these genes were found to promote DNA repair, cell viability and tumorigenesis processes / pathways in LMTK3-overexpressing MCF7 cells. In human cancers, immunohistochemistry staining of LMTK3 in pre- and post-chemotherapy breast tumor pairs from four separate clinical cohorts revealed a significant increase of LMTK3 following both doxorubicin and docetaxel based chemotherapy. In aggregate, our findings show for the first time a contribution of LMTK3 in cytotoxic drug resistance in breast cancer.
Collapse
Affiliation(s)
- Justin Stebbing
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Kalpit Shah
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Lei Cheng Lit
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Teresa Gagliano
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Angeliki Ditsiou
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Tingting Wang
- Cancer Science Institute of Singapore, Centre for Life Sciences, 28 Medical Drive, #02-15, Singapore, Singapore
| | - Franz Wendler
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Thomas Simon
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Krisztina Sára Szabó
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK
| | - Timothy O'Hanlon
- Cancer Genomics Research Laboratory, National Cancer Institute, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc., Bethesda, MD, 20892, USA
| | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - April Camilla Roslani
- Department of Surgery, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Swee Hung Cheah
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Soo-Chin Lee
- Cancer Science Institute of Singapore, Centre for Life Sciences, 28 Medical Drive, #02-15, Singapore, Singapore
| | - Georgios Giamas
- School of Life Sciences, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|
22
|
Abstract
Autophagy is a conserved, self-degradation system that is critical for maintaining cellular homeostasis during stress conditions. Dysregulated autophagy has implications in health and disease. Specifically, in cancer, autophagy plays a dichotomous role by inhibiting tumor initiation but supporting tumor progression. Early results from clinical trials that repurposed hydroxychloroquine for cancer have suggested that autophagy inhibition may be a promising approach for advanced cancers. In this review of the literature, the authors present fundamental advances in the biology of autophagy, approaches to targeting autophagy, the preclinical rationale and clinical experience with hydroxychloroquine in cancer clinical trials, the potential role of autophagy in tumor immunity, and recent developments in next-generation autophagy inhibitors that have clinical potential. Autophagy is a promising target for drug development in cancer. Cancer 2018. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Angelique V Onorati
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matheus Dyczynski
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Rani Ojha
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ravi K Amaravadi
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
Nrf2-p62 autophagy pathway and its response to oxidative stress in hepatocellular carcinoma. Transl Res 2018; 193:54-71. [PMID: 29274776 DOI: 10.1016/j.trsl.2017.11.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Deregulation of autophagy is proposed to play a key pathogenic role in hepatocellular carcinoma (HCC), the most common primary malignancy of the liver and the third leading cause of cancer death. Autophagy is an evolutionarily conserved catabolic process activated to degrade and recycle cell's components. Under stress conditions, such as oxidative stress and nutrient deprivation, autophagy is an essential survival pathway that operates in harmony with other stress response pathways. These include the redox-sensitive transcription complex Nrf2-Keap1 that controls groups of genes with roles in detoxification and antioxidant processes, intermediary metabolism, and cell cycle regulation. Recently, a functional association between a dysfunctional autophagy and Nrf2 pathway activation has been identified in HCC. This appears to occur through the physical interaction of the autophagy adaptor p62 with the Nrf2 inhibitor Keap1, thus leading to increased stabilization and transcriptional activity of Nrf2, a key event in reprogramming metabolic and stress response pathways of proliferating hepatocarcinoma cells. These emerging molecular mechanisms and the therapeutic perspective of targeting Nrf2-p62 interaction in HCC are discussed in this paper along with the prognostic value of autophagy in this type of cancer.
Collapse
|
24
|
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
|
25
|
Tang H, Shu M, Dai B, Xu L, Dong B, Gao G, Chen X. DNA damage response-initiated cytokine secretion in bone marrow stromal cells promotes chemoresistance of myeloma cells. Leuk Lymphoma 2017; 59:2220-2226. [PMID: 29249192 DOI: 10.1080/10428194.2017.1413188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acquisition of chemoresistance accounts for a major cause of chemotherapy failure for multiple myeloma (MM). Bone marrow stromal cells (BMSCs) are considered to play a pivotal role in modulating drug resistance of MM cells. However, the underlying mechanism whereby BMSCs, particularly damaged stromal cells, affects chemoresistance has not been identified yet. Here, we show exposure to doxorubicin doxorubicin (Dox) induced dramatic ATM (ataxia-telangiectasia-mutated)-dependent DNA damage response (DDR) and increased secretion of interleukin (IL)-6 in HS-5 cell line and primary BMSCs derived from healthy donors. Specifically, IL-6-containing conditioned media (CM) derived from Dox-pretreated stromal cells displayed significant protective effect on Dox-induced apoptosis of MM cells. Also, treatment of BMSCs with ATM kinase inhibitor markedly reduced IL-6 secretion and concurrently, partially reversed CM-mediated chemoresistance in myeloma cells. These data indicate that DNA-damaging drug triggers an ATM-dependent DDR in BMSCs, leading to increased cytokine secretion and resistance of myeloma cells to chemotherapy-induced apoptosis.
Collapse
Affiliation(s)
- Hailong Tang
- a Department of Hematology , Xijing Hospital, Fourth Military Medical University , Xi'an , Shaanxi , China
| | - Mimi Shu
- a Department of Hematology , Xijing Hospital, Fourth Military Medical University , Xi'an , Shaanxi , China
| | - Bo Dai
- b Shaanxi Center for Stem Cell Application Engineering Research , Xi'an , Shaanxi , China
| | - Li Xu
- a Department of Hematology , Xijing Hospital, Fourth Military Medical University , Xi'an , Shaanxi , China
| | - Baoxia Dong
- a Department of Hematology , Xijing Hospital, Fourth Military Medical University , Xi'an , Shaanxi , China
| | - Guangxun Gao
- a Department of Hematology , Xijing Hospital, Fourth Military Medical University , Xi'an , Shaanxi , China
| | - Xiequn Chen
- a Department of Hematology , Xijing Hospital, Fourth Military Medical University , Xi'an , Shaanxi , China
| |
Collapse
|
26
|
Hu F, Zhao Y, Yu Y, Fang JM, Cui R, Liu ZQ, Guo XL, Xu Q. Docetaxel-mediated autophagy promotes chemoresistance in castration-resistant prostate cancer cells by inhibiting STAT3. Cancer Lett 2017; 416:24-30. [PMID: 29246644 DOI: 10.1016/j.canlet.2017.12.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022]
Abstract
Signal transducer and activator of transcription (STAT)3 expression is correlated with neoplasm growth, metastasis, and prognosis; it has also been implicated in the regulation of autophagy, which may in turn contribute to tumor chemoresistance. However, it is unknown whether STAT3 is involved in cancer cell survival in response to chemotherapy. In this study, we show that autophagy is triggered during chemotherapy and that inhibiting autophagy increased chemosensitivity of castration-resistant prostate cancer (CRPC) cells. Meanwhile, docetaxel induced autophagy was inhibited by STAT3 activation, which increased mitochondrial damage and decreased CRPC cell viability. These results suggest that STAT3 contributes to CRPC cell survival and chemoresistance by modulating autophagy.
Collapse
Affiliation(s)
- Fei Hu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Yu Zhao
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Yi Yu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Jue-Min Fang
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Ran Cui
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Zhu-Qing Liu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China
| | - Xian-Ling Guo
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China.
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, PR China; Tongji University Cancer Center, Shanghai, 200072, PR China; Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, 200443, PR China.
| |
Collapse
|
27
|
Yuan H, He M, Cheng F, Bai R, da Silva SR, Aguiar RCT, Gao SJ. Tenovin-6 inhibits proliferation and survival of diffuse large B-cell lymphoma cells by blocking autophagy. Oncotarget 2017; 8:14912-14924. [PMID: 28118604 PMCID: PMC5362454 DOI: 10.18632/oncotarget.14741] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is one of the most aggressive non-Hodgkin lymphomas. It is curable but one-third of cases are refractory to therapy or relapse after initial response highlighting the urgent need for developing novel therapeutic approaches. Targeting sirtuins, particularly SIRT1 by genetic approaches or using pharmaceutical inhibitor tenovin-6, has shown promising therapeutic potential in various hematopoietic malignancies. However, it remains unknown whether these approaches are effective for DLBCL. In this study, we have found that tenovin-6 potently inhibits the proliferation and survival of DLBCL cells. Surprisingly, specific knockdown of SIRT1/2/3 has no effect on DLBCL. Mechanistically, tenovin-6 increases the level of microtubule-associated protein 1 light chain 3B (LC3B)-II in a SIRT1/2/3- and p53-independent manner in DLBCL cell lines. Tenovin-6-mediated increase of LC3B-II is through inhibition of classical autophagy pathway. Furthermore, inhibition of the autophagy pathway by using other inhibitors or by knocking down key genes in the pathway impairs cell proliferation and survival of DLBCL cells. These results indicate that targeting the autophagic pathway could be a novel therapeutic strategy for DLBCL and that precaution should be taken to interpret data where tenovin-6 was used as an inhibitor of sirtuins.
Collapse
Affiliation(s)
- Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Meilan He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rosemary Bai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ricardo C T Aguiar
- Department of Medicine and Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,South Texas Veterans Health Care System, Audie Murphy VA Hospital, San Antonio, TX, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
28
|
Circu M, Cardelli J, Barr M, O’Byrne K, Mills G, El-Osta H. Modulating lysosomal function through lysosome membrane permeabilization or autophagy suppression restores sensitivity to cisplatin in refractory non-small-cell lung cancer cells. PLoS One 2017; 12:e0184922. [PMID: 28945807 PMCID: PMC5612465 DOI: 10.1371/journal.pone.0184922] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 09/02/2017] [Indexed: 11/29/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths. Most patients develop resistance to platinum within several months of treatment. We investigated whether triggering lysosomal membrane permeabilization (LMP) or suppressing autophagy can restore cisplatin susceptibility in lung cancer with acquired chemoresistance. Cisplatin IC50 in A549Pt (parental) and A549cisR (cisplatin resistant) cells was 13 μM and 47 μM, respectively. Following cisplatin exposure, A549cisR cells failed to elicit an apoptotic response. This was manifested by diminished Annexin–V staining, caspase 3 and 9, BAX and BAK activation in resistant but not in parental cells. Chloroquine preferentially promoted LMP in A549cisR cells, revealed by leakage of FITC-dextran into the cytosol as detected by immunofluorescence microscopy. This was confirmed by increased cytosolic cathepsin D signal on Immunoblot. Cell viability of cisplatin-treated A549cisR cells was decreased when co-treated with chloroquine, corresponding to a combination index below 0.8, suggesting synergism between the two drugs. Notably, chloroquine activated the mitochondrial cell death pathway as indicated by increase in caspase 9 activity. Interestingly, inhibition of lysosomal proteases using E64 conferred cytoprotection against cisplatin and chloroquine co-treatment, suggesting that chloroquine-induced cell death occurred in a cathepsin-mediated mechanism. Likewise, blockage of caspases partially rescued A549cisR cells against the cytotoxicity of cisplatin and chloroquine combination. Cisplatin promoted a dose-dependent autophagic flux induction preferentially in A549cisR cells, as evidenced by a surge in LC3-II/α-tubulin following pre-treatment with E64 and increase in p62 degradation. Compared to untreated cells, cisplatin induced an increase in cyto-ID-loaded autophagosomes in A549cisR cells that was further amplified by chloroquine, pointing toward autophagic flux activation by cisplatin. Interestingly, this effect was less pronounced in A549Pt cells. Blocking autophagy by ATG5 depletion using siRNA markedly enhances susceptibility to cisplatin in A549cisR cells. Taken together, our results underscore the utility of targeting lysosomal function in overcoming acquired cisplatin refractoriness in lung cancer.
Collapse
Affiliation(s)
- Magdalena Circu
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - James Cardelli
- Segue Therapeutics, LLC, Shreveport, Louisiana, United States of America
| | - Martin Barr
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James’s Hospital & Trinity College Dublin, Dublin, Ireland
| | - Kenneth O’Byrne
- Cancer & Ageing Research Program, Queensland University of Technology, Brisbane, Australia
| | - Glenn Mills
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Hazem El-Osta
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
- * E-mail: ,
| |
Collapse
|
29
|
Transcription factor EB is involved in autophagy-mediated chemoresistance to doxorubicin in human cancer cells. Acta Pharmacol Sin 2017; 38:1305-1316. [PMID: 28603284 DOI: 10.1038/aps.2017.25] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/07/2017] [Indexed: 12/22/2022] Open
Abstract
Transcription factor EB (TFEB) is a master regulator of autophagy activity and lysosomal biogenesis, but its role in autophagy-mediated cell survival and chemotherapy resistance is not completely understood. In this study, we explored whether TFEB played an important role in autophagy-mediated chemotherapy resistance in human cancer LoVo and HeLa cells in vitro. Treatment of human colon cancer LoVo cells with doxorubicin (0.5 μmol/L) induced autophagy activation and nuclear translocation of TFEB, which resulted from inactivation of the mTOR pathway. In both LoVo and HeLa cells, overexpression of TFEB enhanced doxorubicin-induced autophagy activation and significantly decreased doxorubicin-induced cell death, whereas knockdown of TFEB with small interfering RNA blocked doxorubicin-induced autophagy and significantly enhanced the cytotoxicity of doxorubicin. In LoVo cells, autophagy inhibition by 3-methyladenine (3-MA) or knockdown of autophagy-related gene Atg5 increased cell death in response to doxorubicin, and abolished TFEB overexpression-induced chemotherapy resistance, suggesting that the inhibition of autophagy made cancer cells more sensitive to doxorubicin. The results demonstrate that TFEB-mediated autophagy activation decreases the sensitivity of cancer cells to doxorubicin.
Collapse
|
30
|
Bingel C, Koeneke E, Ridinger J, Bittmann A, Sill M, Peterziel H, Wrobel JK, Rettig I, Milde T, Fernekorn U, Weise F, Schober A, Witt O, Oehme I. Three-dimensional tumor cell growth stimulates autophagic flux and recapitulates chemotherapy resistance. Cell Death Dis 2017; 8:e3013. [PMID: 28837150 PMCID: PMC5596581 DOI: 10.1038/cddis.2017.398] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 12/18/2022]
Abstract
Current preclinical models in tumor biology are limited in their ability to recapitulate relevant (patho-) physiological processes, including autophagy. Three-dimensional (3D) growth cultures have frequently been proposed to overcome the lack of correlation between two-dimensional (2D) monolayer cell cultures and human tumors in preclinical drug testing. Besides 3D growth, it is also advantageous to simulate shear stress, compound flux and removal of metabolites, e.g., via bioreactor systems, through which culture medium is constantly pumped at a flow rate reflecting physiological conditions. Here we show that both static 3D growth and 3D growth within a bioreactor system modulate key hallmarks of cancer cells, including proliferation and cell death as well as macroautophagy, a recycling pathway often activated by highly proliferative tumors to cope with metabolic stress. The autophagy-related gene expression profiles of 2D-grown cells are substantially different from those of 3D-grown cells and tumor tissue. Autophagy-controlling transcription factors, such as TFEB and FOXO3, are upregulated in tumors, and 3D-grown cells have increased expression compared with cells grown in 2D conditions. Three-dimensional cultures depleted of the autophagy mediators BECN1, ATG5 or ATG7 or the transcription factor FOXO3, are more sensitive to cytotoxic treatment. Accordingly, combining cytotoxic treatment with compounds affecting late autophagic flux, such as chloroquine, renders the 3D-grown cells more susceptible to therapy. Altogether, 3D cultures are a valuable tool to study drug response of tumor cells, as these models more closely mimic tumor (patho-)physiology, including the upregulation of tumor relevant pathways, such as autophagy.
Collapse
Affiliation(s)
- Corinna Bingel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany
| | - Emily Koeneke
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Johannes Ridinger
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Annika Bittmann
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Martin Sill
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heike Peterziel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Jagoda K Wrobel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Inga Rettig
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany
| | - Till Milde
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany.,Center for Individualized Pediatric Oncology (ZIPO) and Brain Tumors, Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Uta Fernekorn
- Department of Nano-Biosystem Technology, Technische Universität Ilmenau, Ilmenau, Germany
| | - Frank Weise
- Department of Nano-Biosystem Technology, Technische Universität Ilmenau, Ilmenau, Germany
| | - Andreas Schober
- Department of Nano-Biosystem Technology, Technische Universität Ilmenau, Ilmenau, Germany
| | - Olaf Witt
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Ina Oehme
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, D-69120 Heidelberg, Germany.,Translational Program, Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| |
Collapse
|
31
|
You L, Jin S, Zhu L, Qian W. Autophagy, autophagy-associated adaptive immune responses and its role in hematologic malignancies. Oncotarget 2017; 8:12374-12388. [PMID: 27902471 PMCID: PMC5355352 DOI: 10.18632/oncotarget.13583] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 11/11/2016] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a tightly regulated catabolic process that leads to the degradation of cytoplasmatic components such as aggregated/misfolded proteins and organelles through the lysosomal machinery. Recent studies suggest that autophagy plays such a role in the context of the anti-tumor immune response, make it an attractive target for cancer immunotherapy. Defective autophagy in hematopoietic stem cells may contribute to the development of hematologic malignancies, including leukemia, myelodysplastic syndrome, and lymphoproliferative disorder. In blood cancer cells, autophagy can either result in chemoresistance or induce autophagic cell death that may act as immunogenic. Based on the successful experimental findings in vitro and in vivo, clinical trials of autophagy inhibitor such as hydroxychloroquine in combination with chemotherapy in patients with blood cancers are currently underway. However, autophagy inactivation might impair autophagy-triggered anticancer immunity, whereas induction of autophagy might become an effective immunotherapy. These aspects are discussed in this review together with a brief introduction to the autophagic molecular machinery and its roles in hematologic malignancies.
Collapse
Affiliation(s)
- Liangshun You
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Shenhe Jin
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Li Zhu
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Wenbin Qian
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| |
Collapse
|
32
|
Hu F, Guo XL, Zhang SS, Zhao QD, Li R, Xu Q, Wei LX. Suppression of p53 potentiates chemosensitivity in nutrient-deprived cholangiocarcinoma cells via inhibition of autophagy. Oncol Lett 2017; 14:1959-1966. [PMID: 28789429 PMCID: PMC5530065 DOI: 10.3892/ol.2017.6449] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 01/13/2017] [Indexed: 12/21/2022] Open
Abstract
Tumor protein p53 has been intensively studied as a major tumor suppressor. The activation of p53 is associated with various anti-neoplastic functions, including cell senescence, cell cycle arrest, apoptosis and inhibition of angiogenesis. However, the role of p53 in cancer cell chemosensitivity remains unknown. Cholangiocarcinoma cell lines QBC939 and RBE were grown in full-nutrient and nutrient-deprived conditions. The cell lines were treated with 5-fluorouracil or cisplatin and the rate of cell death was determined in these and controls using Cell Counting Kit-8 and microscopy-based methods, including in the presence of autophagy inhibitor 3MA, p53 inhibitor PFT-α or siRNA against p53 or Beclin-1. The present study demonstrated that the inhibition of p53 enhanced the sensitivity to chemotherapeutic agents in nutrient-deprived cholangiocarcinoma cells. Nutrient deprivation-induced autophagy was revealed to be inhibited following inhibition of p53. These data indicate that p53 is important for the activation of autophagy in nutrient-deprived cholangiocarcinoma cells, and thus contributes to cell survival and chemoresistance.
Collapse
Affiliation(s)
- Fei Hu
- Department of Medical Oncology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, P.R. China.,Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Xian-Ling Guo
- Department of Medical Oncology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, P.R. China.,Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Shan-Shan Zhang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Qiu-Dong Zhao
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Rong Li
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Qing Xu
- Department of Medical Oncology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, P.R. China
| | - Li-Xin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
33
|
Scott EC, Maziarz RT, Spurgeon SE, Medvedova E, Gajewski J, Reasor-Heard S, Park B, Kratz A, Thomas GV, Loriaux M, Cascio M, Podolak J, Gordon M, Botelho J, Stadtmauer E, Amaravadi R, Vogl DT. Double autophagy stimulation using chemotherapy and mTOR inhibition combined with hydroxychloroquine for autophagy modulation in patients with relapsed or refractory multiple myeloma. Haematologica 2017; 102:e261-e265. [PMID: 28385778 DOI: 10.3324/haematol.2016.162321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- Emma C Scott
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Richard T Maziarz
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Stephen E Spurgeon
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Eva Medvedova
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - James Gajewski
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Shara Reasor-Heard
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Byung Park
- Public Health and Preventative Medicine, Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Anne Kratz
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - George V Thomas
- The Department of Pathology and Laboratory Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Marc Loriaux
- The Department of Pathology and Laboratory Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Michael Cascio
- The Department of Pathology and Laboratory Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Jennifer Podolak
- The Department of Pathology and Laboratory Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Miranda Gordon
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Jennifer Botelho
- The Department of Medicine and the Knight Cancer Institute at Oregon Health & Science University, the University of Pennsylvania, PN, USA
| | - Edward Stadtmauer
- Department of Medicine and the Abramson Cancer Center, the University of Pennsylvania, PN, USA
| | - Ravi Amaravadi
- Department of Medicine and the Abramson Cancer Center, the University of Pennsylvania, PN, USA
| | - Dan T Vogl
- Department of Medicine and the Abramson Cancer Center, the University of Pennsylvania, PN, USA
| |
Collapse
|
34
|
Wang M, Huang C, Su Y, Yang C, Xia Q, Xu DJ. Astragaloside II sensitizes human hepatocellular carcinoma cells to 5-fluorouracil via suppression of autophagy. ACTA ACUST UNITED AC 2017; 69:743-752. [PMID: 28266023 DOI: 10.1111/jphp.12706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/12/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Inhibition of autophagy has been increasingly recognized as a potential therapeutic approach against cancer. Our previous reports showed that Astragaloside II improves hepatic cancer cells resistance by downregulating MDR1 and P-gp .The purpose of this study was to further investigated the effect of autophagy on AS-II reversing multidrug resistance and its molecular mechanism in hepatocellular carcinoma cells in vitro. METHODS Bel-7402 and Bel-7402/FU cell lines were used in this study. Western blot was used to detect the expression of autophagy-related protein, p-mTOR and p-p79s6k, MTT was used to analyse cell viability, GFP-LC3 punctate dots distribution was observed by GFP-LC3 transient transfection under fluorescence microscopy and silencing of autophagy-related genes was detected by small interfering RNA transfection. KEY FINDINGS Astragaloside II was able to significantly decrease the expression of LC3-II and Beclin-1 in a dose-dependent manner, Astragaloside II (80 μm) further decreased LC3-II formation, Beclin-1 and GFP-LC3 puncta dots stimulated with 5-fluorouracil (0.2 mm) in Bel-7402/FU cells (P < 0.05). In addition, Astragaloside II is capable of sensitizing cells to 5-fluorouracil-induced cell death via inhibition of pro-survival autophagy involvement of MAPK-mTOR pathway. CONCLUSIONS These findings suggested that Astragaloside II could suppress autophagy by interfering with Beclin-1 and LC3 via MAPK-mTOR pathway, through which sensitized human cancer resistant cells to 5-FU-induced cell death.
Collapse
Affiliation(s)
- Meng Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Can Huang
- Department of Pharmacy, Affiliated Anqing Hospital of Anhui Medical University, Anqing, Anhui, China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Cui Yang
- Department of Pharmacy, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Du-Juan Xu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
35
|
Research progress of hydroxychloroquine and autophagy inhibitors on cancer. Cancer Chemother Pharmacol 2016; 79:287-294. [PMID: 27889812 DOI: 10.1007/s00280-016-3197-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/11/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Hydroxychloroquine (HCQ), the analog of chloroquine, augments the effect of chemotherapies and radiotherapy on various tumors identified in the current clinical trials. Meanwhile, the toxicity of HCQ retinopathy raises concern worldwide. Thus, the potent autophagy inhibitors are urgently needed. METHODS A systematic review was related to 'hydroxychloroquine' or 'chloroquine' with 'clinical trials,' 'retinopathy' and 'new autophagy inhibitors.' This led to many cross-references involving HCQ, and these data have been incorporated into the following study. RESULTS Many preclinical studies indicate that the combination of HCQ with chemotherapies or radiotherapies may enhance the effect of anticancer, providing base for launching cancer clinical trials involving HCQ. The new and more sensitive diagnostic techniques report a prevalence of HCQ retinopathy up to 7.5%. Lys05, SAR405, verteporfin, VATG-027, mefloquine and spautin-1 may be potent autophagy inhibitors. CONCLUSION Additional mechanistic studies of HCQ in preclinical models are still required in order to answer these questions whether HCQ actually inhibits autophagy in non-selective tumors and whether the extent of inhibition would be sufficient to alter chemotherapy or radiotherapy sensitivity.
Collapse
|
36
|
Eliopoulos AG, Havaki S, Gorgoulis VG. DNA Damage Response and Autophagy: A Meaningful Partnership. Front Genet 2016; 7:204. [PMID: 27917193 PMCID: PMC5116470 DOI: 10.3389/fgene.2016.00204] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/02/2016] [Indexed: 01/07/2023] Open
Abstract
Autophagy and the DNA damage response (DDR) are biological processes essential for cellular and organismal homeostasis. Herein, we summarize and discuss emerging evidence linking DDR to autophagy. We highlight published data suggesting that autophagy is activated by DNA damage and is required for several functional outcomes of DDR signaling, including repair of DNA lesions, senescence, cell death, and cytokine secretion. Uncovering the mechanisms by which autophagy and DDR are intertwined provides novel insight into the pathobiology of conditions associated with accumulation of DNA damage, including cancer and aging, and novel concepts for the development of improved therapeutic strategies against these pathologies.
Collapse
Affiliation(s)
- Aristides G Eliopoulos
- Molecular and Cellular Biology Laboratory, Division of Basic Sciences, Medical School, University of CreteHeraklion, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology HellasHeraklion, Greece
| | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of AthensAthens, Greece; Faculty Institute of Cancer Sciences, Manchester Academic Health Sciences Centre, University of ManchesterManchester, UK; Biomedical Research Foundation of the Academy of AthensAthens, Greece
| |
Collapse
|
37
|
Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy. Nat Rev Clin Oncol 2016; 14:247-258. [PMID: 27845767 DOI: 10.1038/nrclinonc.2016.183] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is fundamental to the maintenance of intracellular homeostasis in virtually all human cells. Accordingly, defective autophagy predisposes healthy cells to undergoing malignant transformation. By contrast, malignant cells are able to harness autophagy to thrive, despite adverse microenvironmental conditions, and to resist therapeutic challenges. Thus, inhibition of autophagy has been proposed as a strategy to kill cancer cells or sensitize them to therapy; however, autophagy is also critical for optimal immune function, and mediates cell-extrinsic homeostatic effects owing to its central role in danger signalling by neoplastic cells responding to immunogenic chemotherapy and/or radiation therapy. In this Perspective, we discuss accumulating preclinical and clinical evidence in support of the all-too-often dismissed possibility that activating autophagy might be a relevant clinical objective that enables an increase in the effectiveness of immunogenic chemotherapy and/or radiation therapy.
Collapse
|
38
|
Wu ST, Sun GH, Cha TL, Kao CC, Chang SY, Kuo SC, Way TD. CSC-3436 switched tamoxifen-induced autophagy to apoptosis through the inhibition of AMPK/mTOR pathway. J Biomed Sci 2016; 23:60. [PMID: 27526942 PMCID: PMC4986227 DOI: 10.1186/s12929-016-0275-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) lacks specific therapeutic target and limits to chemotherapy and is essential to develop novel therapeutic regimens. Increasing studies indicated that tamoxifen, a selective estrogen receptor modulators (SERMs), has anti-tumor therapeutic effect in estrogen receptor α (ERα)-negative tumor. Here, we determined whether autophagy was activated by tamoxifen in TNBC cells. Moreover, CSC-3436 displayed strong and selective growth inhibition on cancer cells. Next, we investigated the anti-proliferation effect of combination of CSC-3436 plus tamoxifen on cell death in TNBC cells. Results Our study found that tamoxifen induces autophagy in TNBC cells. Endoplasmic reticulum stress and AMPK/mTOR contributed tamoxifen-induced autophagy. Interestingly, in combination treatment with CSC-3436 enhanced the anti-proliferative effect of tamoxifen. We found that CSC-3436 switched tamoxifen-induced autophagy to apoptosis via cleavage of ATG-5. Moreover, AMPK/mTOR pathway may involve in CSC-3436 switched tamoxifen-induced autophagy to apoptosis. The combination of tamoxifen and CSC-3436 produced stronger tumor growth inhibition compared with CSC-3436 or tamoxifen alone treatments in vivo. Conclusion These data indicated that CSC-3436 combined with tamoxifen may be a potential approach for treatment TNBC.
Collapse
Affiliation(s)
- Sheng-Tang Wu
- Division of Urology, Department of Surgery, Tri-Service General Hospital and National Defense Medical Center, Taichung, Taiwan
| | - Guang-Huan Sun
- Division of Urology, Department of Surgery, Tri-Service General Hospital and National Defense Medical Center, Taichung, Taiwan
| | - Tai-Lung Cha
- Division of Urology, Department of Surgery, Tri-Service General Hospital and National Defense Medical Center, Taichung, Taiwan
| | - Chien-Chang Kao
- Division of Urology, Department of Surgery, Tri-Service General Hospital and National Defense Medical Center, Taichung, Taiwan
| | - Sun-Yran Chang
- Division of Urology, Department of Surgery, Tri-Service General Hospital and National Defense Medical Center, Taichung, Taiwan
| | - Sheng-Chu Kuo
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan. .,Graduate institute of Pharmaceutical Chemistry, China Medical University, Taichung, 40402, Taiwan R.O.C.
| | - Tzong-Der Way
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan R.O.C. .,Department of Health and Nutrition Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
39
|
Gomes LR, Vessoni AT, Menck CF. Microenvironment and autophagy cross-talk: Implications in cancer therapy. Pharmacol Res 2016; 107:300-307. [DOI: 10.1016/j.phrs.2016.03.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 03/25/2016] [Accepted: 03/27/2016] [Indexed: 02/07/2023]
|
40
|
Cheong JW, Kim Y, Eom JI, Jeung HK, Min YH. Enhanced autophagy in cytarabine arabinoside-resistant U937 leukemia cells and its potential as a target for overcoming resistance. Mol Med Rep 2016; 13:3433-40. [PMID: 26935591 PMCID: PMC4805098 DOI: 10.3892/mmr.2016.4949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 01/20/2016] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a lysosomal degradation mechanism that is essential for cell survival, differentiation, development, and homeostasis. Autophagy protects cells from various stresses, including protecting normal cells from harmful metabolic conditions, and cancer cells from chemotherapeutics. In the current study, a cytarabine arabinoside (Ara-C)-sensitive U937 leukemia cell line and an Ara-C-resistant U937 (U937/AR) cell line were assessed for baseline autophagy activity by investigating the LC3-I conversion to LC3-II, performing EGFP-LC3 puncta, an acidic autophagolysosome assay, and measuring the expression of various autophagy-related genes. The results demonstrated significantly higher autophagic activity in the U937/AR cells compared with the U937 cells, when the cells were cultured with or without serum. Furthermore, an increase in the autophagic activity in starved U937/AR cells was demonstrated, compared with that in the starved U937 cells. Administration of an autophagy inhibitor demonstrated no change in cell death in the two cell lines when cultured with serum, however, it induced cell death regardless of the Ara-C sensitivity when the cell lines were cultured without serum. In addition, the U937 cells demonstrated an Ara-C resistance when cultured without serum. Co-treatment with Ara-C and the autophagy inhibitor significantly induced cell death in the U937/AR and Ara-C-sensitive U937 cells. In conclusion, autophagy serves an important role in protecting U937 cells from Ara-C and in the development of Ara-C resistance. Inhibition of autophagy combined with the Ara-C treatment in the U937 cells augmented the anti-leukemic effect of Ara-C and overcame Ara-C resistance, suggesting that autophagy may be an important therapeutic target to further improve the treatment outcome in patients with acute myeloid leukemia.
Collapse
Affiliation(s)
- June-Won Cheong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Yundeok Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Ju In Eom
- Medical Research Center, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Hoi-Kyung Jeung
- Medical Research Center, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Yoo Hong Min
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| |
Collapse
|
41
|
Willenbacher W, Thangavadivel S, Greil R, Willenbacher E, Weger R, Manzl C, Jöhrer K, Brunner A. Low Beclin-1 expression predicts improved overall survival in patients treated with immunomodulatory drugs for multiple myeloma and identifies autophagy inhibition as a promising potentially druggable new therapeutic target: an analysis from The Austrian Myeloma Registry (AMR). Leuk Lymphoma 2016; 57:2330-41. [DOI: 10.3109/10428194.2016.1144880] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
42
|
Lefort S, Joffre C, Kieffer Y, Givel AM, Bourachot B, Zago G, Bieche I, Dubois T, Meseure D, Vincent-Salomon A, Camonis J, Mechta-Grigoriou F. Inhibition of autophagy as a new means of improving chemotherapy efficiency in high-LC3B triple-negative breast cancers. Autophagy 2015; 10:2122-42. [PMID: 25427136 DOI: 10.4161/15548627.2014.981788] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The triple-negative breast cancer (TN BC) subtype is the most aggressive form of invasive BC. Despite intensive efforts to improve BC treatments, patients with TN BC continue to exhibit poor survival, with half developing resistance to chemotherapy. Here we identify autophagy as a key mechanism in the progression and chemoresistance of a subset of TN tumors. We demonstrate that LC3B, a protein involved in autophagosome formation, is a reliable marker of poor prognosis in TN BC, validating this prognostic value at both the mRNA and protein levels in several independent cohorts. We also show that LC3B has no prognostic value for other BC subtypes (Luminal or HER2 BC), thus revealing a specific impact of autophagy on TN tumors. Autophagy is essential for the proliferative and invasive properties in 3D of TN BC cells characterized by high LC3B levels. Interestingly, the activity of the transcriptional co-activator YAP1 (Yes-associated protein 1) is regulated by the autophagy process and we identify YAP1 as a new actor in the autophagy-dependent proliferative and invasive properties of high-LC3B TN BC. Finally, inhibiting autophagy by silencing ATG5 or ATG7 significantly impaired high-LC3B TN tumor growth in vivo. Moreover, using a patient-derived TN tumor transplanted into mice, we show that an autophagy inhibitor, chloroquine, potentiates the effects of chemotherapeutic agents. Overall, our data identify LC3B as a new prognostic marker for TN BC and the inhibition of autophagy as a promising therapeutic strategy for TN BC patients.
Collapse
Key Words
- 3-dimensional culture
- 3D, 3-dimensions
- AC, adriamycin and cyclophosphamide
- ACTB, actin, β
- AP2A1/adaptin, adaptor-related protein complex 2, α 1 subunit
- ATG, autophagy-related
- BC, breast cancer
- BECN1, Beclin 1, autophagy related
- BafA1, bafilomycin A1
- Ctrl, control
- DFS, disease-free survival
- EBSS, Earle's balanced salt solution
- ERBB2/HER2, v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- HScore, histological scoring
- IHC, immunohistochemistry
- LC3B
- Lum, Luminal
- MAP1LC3B/LC3B, microtubule-associated protein one light chain 3 β
- OS, overall survival
- PDX, patient-derived xenografted tumor
- TCGA, The Cancer Genome Atlas
- TGI, tumor growth inhibition
- TN BC, triple-negative breast cancer
- YAP1
- YAP1, Yes-associated protein 1
- autophagy
- breast cancers
- i.p., intra-peritoneal
- prognosis
- response to treatment
- sem, standard error of mean
- three-MA, 3-methyladenine
Collapse
Affiliation(s)
- Sylvain Lefort
- a Laboratory of Stress and Cancer; Institut Curie ; Paris , France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kim DG, Jung KH, Lee DG, Yoon JH, Choi KS, Kwon SW, Shen HM, Morgan MJ, Hong SS, Kim YS. 20(S)-Ginsenoside Rg3 is a novel inhibitor of autophagy and sensitizes hepatocellular carcinoma to doxorubicin. Oncotarget 2015; 5:4438-51. [PMID: 24970805 PMCID: PMC4147336 DOI: 10.18632/oncotarget.2034] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. High mortality from HCC is mainly due to widespread prevalence and the lack of effective treatment, since systemic chemotherapy is ineffective, while the targeted agent Sorafenib extends median survival only briefly. The steroidal saponin 20(S)-ginsenoside Rg3 from Panax ginseng C.A. Meyer is proposed to chemosensitize to various therapeutic drugs through an unknown mechanism. Since autophagy often serves as cell survival mechanism in cancer cells exposed to chemotherapeutic agents, we examined the ability of Rg3 to inhibit autophagy and chemosensitize HCC cell lines to doxorubicin in vitro. We show that Rg3 inhibits late stage autophagy, possibly through changes in gene expression. Doxorubicin-induced autophagy plays a protective role in HCC cells, and therefore Rg3 treatment synergizes with doxorubicin to kill HCC cell lines, but the combination is relatively nontoxic in normal liver cells. In addition, Rg3 was well-tolerated in mice and synergized with doxorubicin to inhibit tumor growth in HCC xenografts in vivo. Since novel in vivo inhibitors of autophagy are desirable for clinical use, we propose that Rg3 is such a compound, and that combination therapy with classical chemotherapeutic drugs may represent an effective therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Dong-Gun Kim
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | | | - Da-Gyum Lee
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | - Jung-Ho Yoon
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | - Kyeong Sook Choi
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | - Sung Won Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Han-Ming Shen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Michael J Morgan
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| | | | - You-Sun Kim
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| |
Collapse
|
44
|
Tucci M, Stucci S, Savonarola A, Resta L, Cives M, Rossi R, Silvestris F. An imbalance between Beclin-1 and p62 expression promotes the proliferation of myeloma cells through autophagy regulation. Exp Hematol 2014; 42:897-908.e1. [DOI: 10.1016/j.exphem.2014.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/02/2014] [Accepted: 06/13/2014] [Indexed: 12/19/2022]
|
45
|
Dutta D, Xu J, Dirain MLS, Leeuwenburgh C. Calorie restriction combined with resveratrol induces autophagy and protects 26-month-old rat hearts from doxorubicin-induced toxicity. Free Radic Biol Med 2014; 74:252-62. [PMID: 24975655 PMCID: PMC4146714 DOI: 10.1016/j.freeradbiomed.2014.06.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 06/05/2014] [Accepted: 06/17/2014] [Indexed: 12/21/2022]
Abstract
The multiple beneficial effects of calorie restriction (CR) on several organs, including the heart, are widely known. Recently, the plant polyphenol resveratrol has been shown to possess several beneficial effects similar to those of CR. Among the host of effects on cardiac muscle, a cellular self-eating process called autophagy has been shown to be induced by both CR and resveratrol. Autophagy is vital in removing dysfunctional organelles and damaged proteins from the cell, thereby maintaining cellular quality control. In this study, we explored whether short-term moderate CR (20%), either alone or in combination with resveratrol, can induce autophagy in the hearts of 26-month-old Fischer 344 × Brown Norway rats. Autophagy stimulation was investigated by measuring the protein expression levels of the autophagy proteins beclin-1, Atg5, and p62 and the LC3-II/LC3-I ratio. We found that 20% CR or resveratrol alone for 6 weeks could not induce autophagy, but 20% CR in combination with 50 mg/kg/day resveratrol resulted in an induction of autophagy in the hearts of 26-month-old rats. Although oxidative stress has been proposed to be an inducer of autophagy, treatment with the chemotherapeutic drug doxorubicin was unable to stimulate autophagy. The enhanced autophagy due to CR + resveratrol was associated with protection from doxorubicin-induced damage, as measured by cardiac apoptotic levels and serum creatine kinase and lactate dehydrogenase activity. We propose that a combinatorial approach of low-dose CR and resveratrol has the potential to be used therapeutically to induce autophagy and provides protection against doxorubicin-mediated toxicity.
Collapse
Affiliation(s)
| | - Jinze Xu
- Department of Aging and Geriatric Research, Institute on Aging, and University of Florida, Gainesville, FL 32610, USA
| | - Marvin L S Dirain
- Department of Aging and Geriatric Research, Institute on Aging, and University of Florida, Gainesville, FL 32610, USA
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, Institute on Aging, and University of Florida, Gainesville, FL 32610, USA; Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
46
|
Clioquinol induces pro-death autophagy in leukemia and myeloma cells by disrupting the mTOR signaling pathway. Sci Rep 2014; 4:5749. [PMID: 25034786 PMCID: PMC4102920 DOI: 10.1038/srep05749] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/02/2014] [Indexed: 12/12/2022] Open
Abstract
Clioquinol is an anti-microbial drug, and it was recently found to induce cancer cell death. In the present study, clioquinol was found to trigger autophagy by inducing LC3 lipidation and autophagosome formation which was abolished by an autophagy inhibitor 3-methyladenine. Further study showed clioquinol displayed no effects on PI3KC3 or Beclin 1 expression but downregulated the expression and the enzymatic activity of mammalian target of Rapamycin (mTOR), a critical modulator of autophagy. Moreover, clioquinol inhibited the catalytic activity of the mTOR complex 1, thus suppressing phosphorylation of P70S6K and 4E-BP1, two major proteins associated with autophagy in the mTORC1 signaling pathway. Clioquinol induced leukemia and myeloma cell apoptosis, however, addition of autophagy inhibitor 3-methyladenine attenuated this kind of cell death. Therefore, this study demonstrated that clioquinol induces autophagy in associated with apoptosis in leukemia and myeloma cells by disrupting mTOR signaling pathway.
Collapse
|
47
|
Ristic B, Bosnjak M, Arsikin K, Mircic A, Suzin-Zivkovic V, Bogdanovic A, Perovic V, Martinovic T, Kravic-Stevovic T, Bumbasirevic V, Trajkovic V, Harhaji-Trajkovic L. Idarubicin induces mTOR-dependent cytotoxic autophagy in leukemic cells. Exp Cell Res 2014; 326:90-102. [PMID: 24907655 DOI: 10.1016/j.yexcr.2014.05.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/21/2014] [Accepted: 05/26/2014] [Indexed: 02/06/2023]
Abstract
We investigated if the antileukemic drug idarubicin induces autophagy, a process of programmed cellular self-digestion, in leukemic cell lines and primary leukemic cells. Transmission electron microscopy and acridine orange staining demonstrated the presence of autophagic vesicles and intracellular acidification, respectively, in idarubicin-treated REH leukemic cell line. Idarubicin increased punctuation/aggregation of microtubule-associated light chain 3B (LC3B), enhanced the conversion of LC3B-I to autophagosome-associated LC3B-II in the presence of proteolysis inhibitors, and promoted the degradation of the selective autophagic target p62, thus indicating the increase in autophagic flux. Idarubicin inhibited the phosphorylation of the main autophagy repressor mammalian target of rapamycin (mTOR) and its downstream target p70S6 kinase. The treatment with the mTOR activator leucine prevented idarubicin-mediated autophagy induction. Idarubicin-induced mTOR repression was associated with the activation of the mTOR inhibitor AMP-activated protein kinase and down-regulation of the mTOR activator Akt. The suppression of autophagy by pharmacological inhibitors or LC3B and beclin-1 genetic knockdown rescued REH cells from idarubicin-mediated oxidative stress, mitochondrial depolarization, caspase activation and apoptotic DNA fragmentation. Idarubicin also caused mTOR inhibition and cytotoxic autophagy in K562 leukemic cell line and leukocytes from chronic myeloid leukemia patients, but not healthy controls. By demonstrating mTOR-dependent cytotoxic autophagy in idarubicin-treated leukemic cells, our results warrant caution when considering combining idarubicin with autophagy inhibitors in leukemia therapy.
Collapse
Affiliation(s)
- Biljana Ristic
- Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia
| | - Mihajlo Bosnjak
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Katarina Arsikin
- Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia
| | - Aleksandar Mircic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Violeta Suzin-Zivkovic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Andrija Bogdanovic
- Clinic for Hematology, Clinical Centre of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Perovic
- Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia
| | - Tamara Martinovic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tamara Kravic-Stevovic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Bumbasirevic
- Institute of Histology and Embryology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia.
| | - Ljubica Harhaji-Trajkovic
- Institute for Biological Research, University of Belgrade, Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia.
| |
Collapse
|
48
|
Rangwala R, Leone R, Chang YC, Fecher LA, Schuchter LM, Kramer A, Tan KS, Heitjan DF, Rodgers G, Gallagher M, Piao S, Troxel AB, Evans TL, DeMichele AM, Nathanson KL, O'Dwyer PJ, Kaiser J, Pontiggia L, Davis LE, Amaravadi RK. Phase I trial of hydroxychloroquine with dose-intense temozolomide in patients with advanced solid tumors and melanoma. Autophagy 2014; 10:1369-79. [PMID: 24991839 PMCID: PMC4203514 DOI: 10.4161/auto.29118] [Citation(s) in RCA: 281] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Blocking autophagy with hydroxychloroquine (HCQ) augments cell death associated with alkylating chemotherapy in preclinical models. This phase I study evaluated the maximum tolerated dose (MTD), safety, preliminary activity, pharmacokinetics, and pharmacodynamics of HCQ in combination with dose-intense temozolomide (TMZ) in patients with advanced solid malignancies. Forty patients (73% metastatic melanoma) were treated with oral HCQ 200 to 1200 mg daily with dose-intense oral TMZ 150 mg/m2 daily for 7/14 d. This combination was well tolerated with no recurrent dose-limiting toxicities observed. An MTD was not reached for HCQ and the recommended phase II dose was HCQ 600 mg twice daily combined with dose-intense TMZ. Common toxicities included grade 2 fatigue (55%), anorexia (28%), nausea (48%), constipation (20%), and diarrhea (20%). Partial responses and stable disease were observed in 3/22 (14%) and 6/22 (27%) patients with metastatic melanoma. In the final dose cohort 2/6 patients with refractory BRAF wild-type melanoma had a near complete response, and prolonged stable disease, respectively. A significant accumulation in autophagic vacuoles (AV) in peripheral blood mononuclear cells was observed in response to combined therapy. Population pharmacokinetics (PK) modeling, individual PK simulations, and PK-pharmacodynamics (PD) analysis identified a threshold HCQ peak concentration that predicts therapy-associated AV accumulation. This study indicates that the combination of high-dose HCQ and dose-intense TMZ is safe and tolerable, and is associated with autophagy modulation in patients. Prolonged stable disease and responses suggest antitumor activity in melanoma patients, warranting further studies of this combination, or combinations of more potent autophagy inhibitors and chemotherapy in melanoma.
Collapse
Affiliation(s)
- Reshma Rangwala
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Robert Leone
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Yunyoung C Chang
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Leslie A Fecher
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Lynn M Schuchter
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Amy Kramer
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Kay-See Tan
- Center for Clinical Epidemiology and Biostatistics; University of Pennsylvania; Philadelphia, PA USA
| | - Daniel F Heitjan
- Center for Clinical Epidemiology and Biostatistics; University of Pennsylvania; Philadelphia, PA USA
| | - Glenda Rodgers
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Maryann Gallagher
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Shengfu Piao
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Andrea B Troxel
- Center for Clinical Epidemiology and Biostatistics; University of Pennsylvania; Philadelphia, PA USA
| | - Tracey L Evans
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Angela M DeMichele
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Katherine L Nathanson
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Peter J O'Dwyer
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Jonathon Kaiser
- Philadelphia College of Pharmacy; University of the Sciences; Philadelphia, PA USA
| | - Laura Pontiggia
- Department of Mathematics, Physics and Statistics; University of the Sciences; Philadelphia, PA USA
| | - Lisa E Davis
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA; Philadelphia College of Pharmacy; University of the Sciences; Philadelphia, PA USA
| | - Ravi K Amaravadi
- Department of Medicine and Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| |
Collapse
|
49
|
Sola B, Poirot M, de Medina P, Bustany S, Marsaud V, Silvente-Poirot S, Renoir JM. Antiestrogen-binding site ligands induce autophagy in myeloma cells that proceeds through alteration of cholesterol metabolism. Oncotarget 2014; 4:911-22. [PMID: 23978789 PMCID: PMC3757248 DOI: 10.18632/oncotarget.1066] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma (MM) is a malignancy characterized by the accumulation of clonal plasma cells in the bone marrow. Despite extensive efforts to design drugs targeting tumoral cells and their microenvironment, MM remains an incurable disease for which new therapeutic strategies are needed. We demonstrated here that antiestrogens (AEs) belonging to selective estrogen receptor modulators family induce a caspase-dependent apoptosis and trigger a protective autophagy. Autophagy was recognized by monodansylcadaverin staining, detection of autophagosomes by electronic microscopy, and detection of the cleaved form of the microtubule-associated protein light chain 3. Moreover, autophagy was inhibited by drugs such as bafilomycin A1 and 3-methyladenosine. Autophagy was mediated by the binding of AEs to a class of receptors called the antiestrogen binding site (AEBS) different from the classical estrogen nuclear receptors. The binding of specific ligands to the AEBS was accompanied by alteration of cholesterol metabolism and in particular accumulation of sterols: zymostenol or desmosterol depending on the ligand. This was due to the inhibition of the cholesterol-5,6-epoxide hydrolase activity borne by the AEBS. We further showed that the phosphoinositide 3-kinase/AKT/mammalian target of rapamycin pathway mediated autophagy signaling. Moreover, AEBS ligands restored sensitivity to dexamethasone in resistant MM cells. Since we showed previously that AEs arrest MM tumor growth in xenografted mice, we propose that AEBS ligands may have a potent antimyeloma activity alone or in combination with drugs used in clinic.
Collapse
|
50
|
Abstract
Autophagy is the process by which cellular material is delivered to lysosomes for degradation and recycling. There are three different types of autophagy, but macroautophagy, which involves the formation of double membrane vesicles that engulf proteins and organelles that fuse with lysosomes, is by far the most studied and is thought to have important context-dependent roles in cancer development, progression, and treatment. The roles of autophagy in cancer treatment are complicated by two important discoveries over the past few years. First, most (perhaps all) anticancer drugs, as well as ionizing radiation, affect autophagy. In most, but not all cases, these treatments increase autophagy in tumor cells. Second, autophagy affects the ability of tumor cells to die after drug treatment, but the effect of autophagy may be to promote or inhibit cell death, depending on context. Here we discuss recent research related to autophagy and cancer therapy with a focus on how these processes may be manipulated to improve cancer therapy.
Collapse
Affiliation(s)
- Andrew Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (A.T.); and Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado (D.H.T., D.L.G.)
| | | | | |
Collapse
|