101
|
Singh SK, Abbas S, Saxena AK, Tiwari S, Sharma LK, Tiwari M. Critical role of three-dimensional tumorsphere size on experimental outcome. Biotechniques 2020; 69:333-338. [PMID: 33000639 DOI: 10.2144/btn-2020-0081] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/27/2020] [Indexed: 11/23/2022] Open
Abstract
Three-dimensional in vitro spheroids are a reliable model to study tumor biology and drug toxicity. However, inconsistencies exist in terms of seeding cell density that governs spheroid size and shape, influencing the experimental outcome. We investigated the effect of varying cell densities using glioblastoma cells on tumorsphere formation and their responsiveness to drug treatment. Our results demonstrated that in comparison with spheroids formed with lower cell density, spheroids formed with higher cell density were not only larger in size but also had a larger necrotic core surrounded by a higher number of quiescent cells and were irresponsive to drug treatment. Our study highlights the importance of predetermination of cell density to obtain desired/appropriate spheroid size to produce consistent and reliable data on drug toxicity studies in tumor cells.
Collapse
Affiliation(s)
- Suraj Kumar Singh
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences-Patna, Bihar, India
| | - Sabiya Abbas
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences-Lucknow, UP, India
| | - Ajit Kumar Saxena
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences-Patna, Bihar, India
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences-Lucknow, UP, India
| | - Lokendra Kumar Sharma
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences-Lucknow, UP, India
| | - Meenakshi Tiwari
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences-Patna, Bihar, India
| |
Collapse
|
102
|
Vargas JE, Puga R, Lenz G, Trindade C, Filippi-Chiela E. Cellular Mechanisms Triggered by the Cotreatment of Resveratrol and Doxorubicin in Breast Cancer: A Translational In Vitro-In Silico Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5432651. [PMID: 33204396 PMCID: PMC7654215 DOI: 10.1155/2020/5432651] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/22/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022]
Abstract
Doxorubicin (Doxo) is the most effective chemotherapeutic agent for the treatment of breast cancer. However, resistance to Doxo is common. Adjuvant compounds capable of modulating mechanisms involved in Doxo resistance may potentiate the effectiveness of the drug. Resveratrol (Rsv) has been tested as an adjuvant in mammary malignancies. However, the cellular and molecular mechanisms underlying the effects of cotreatment with Doxo and Rsv in breast cancer are poorly understood. Here, we combined in vitro and in silico analysis to characterize these mechanisms. In vitro, we employed a clinically relevant experimental design consisting of acute (24 h) treatment followed by 15 days of analysis. Acute Rsv potentiated the long-lasting effect of Doxo through the induction of apoptosis and senescence. Cells that survived to the cotreatment triggered high levels of autophagy. Autophagy inhibition during its peak of activation but not concomitant with Doxo+Rsv increased the long-term toxicity of the cotreatment. To uncover key proteins potentially associated with in vitro effects, an in silico multistep strategy was implemented. Chemical-protein networks were predicted based on constitutive gene expression of MCF7 cells and interatomic data from breast cancer. Topological analysis, KM survival analysis, and a quantitative model based on the connectivity between apoptosis, senescence, and autophagy were performed. We found seven putative genes predicted to be modulated by Rsv in the context of Doxo treatment: CCND1, CDH1, ESR1, HSP90AA1, MAPK3, PTPN11, and RPS6KB1. Six out of these seven genes have been experimentally proven to be modulated by Rsv in cancer cells, with 4 of the 6 genes in MCF7 cells. In conclusion, acute Rsv potentiated the long-term toxicity of Doxo in breast cancer potentially through the modulation of genes and mechanisms involved in Doxo resistance. Rational autophagy inhibition potentiated the effects of Rsv+Doxo, a strategy that should be further tested in animal models.
Collapse
Affiliation(s)
- José Eduardo Vargas
- Instituto de Ciências Biológicas, Universidade de Passo Fundo, Brazil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Renato Puga
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Guido Lenz
- Centro de Biotecnologia e Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Cristiano Trindade
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Eduardo Filippi-Chiela
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
103
|
Wang L, Li M, Sha B, Hu X, Sun Y, Zhu M, Xu Y, Li P, Wang Y, Guo Y, Li J, Shi J, Li P, Hu T, Chen P. Inhibition of deubiquitination by PR-619 induces apoptosis and autophagy via ubi-protein aggregation-activated ER stress in oesophageal squamous cell carcinoma. Cell Prolif 2020; 54:e12919. [PMID: 33129231 PMCID: PMC7791184 DOI: 10.1111/cpr.12919] [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: 06/15/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Targeting the deubiquitinases (DUBs) has become a promising avenue for anti-cancer drug development. However, the effect and mechanism of pan-DUB inhibitor, PR-619, on oesophageal squamous cell carcinoma (ESCC) cells remain to be investigated. MATERIALS AND METHODS The effect of PR-619 on ESCC cell growth and cell cycle was evaluated by CCK-8 and PI staining. Annexin V-FITC/PI double staining was performed to detect apoptosis. LC3 immunofluorescence and acridine orange staining were applied to examine autophagy. Intercellular Ca2+ concentration was monitored by Fluo-3AM fluorescence. The accumulation of ubi-proteins and the expression of the endoplasmic reticulum (ER) stress-related protein and CaMKKβ-AMPK signalling were determined by immunoblotting. RESULTS PR-619 could inhibit ESCC cell growth and induce G2/M cell cycle arrest by downregulating cyclin B1 and upregulating p21. Meanwhile, PR-619 led to the accumulation of ubiquitylated proteins, induced ER stress and triggered apoptosis by the ATF4-Noxa axis. Moreover, the ER stress increased cytoplasmic Ca2+ and then stimulated autophagy through Ca2+ -CaMKKβ-AMPK signalling pathway. Ubiquitin E1 inhibitor, PYR-41, could reduce the accumulation of ubi-proteins and alleviate ER stress, G2/M cell cycle arrest, apoptosis and autophagy in PR-619-treated ESCC cells. Furthermore, blocking autophagy by chloroquine or bafilomycin A1 enhanced the cell growth inhibition effect and apoptosis induced by PR-619. CONCLUSIONS Our findings reveal an unrecognized mechanism for the cytotoxic effects of general DUBs inhibitor (PR-619) and imply that targeting DUBs may be a potential anti-ESCC strategy.
Collapse
Affiliation(s)
- Longhao Wang
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Miaomiao Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Beibei Sha
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuanyu Hu
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaxin Sun
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mingda Zhu
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Xu
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pingping Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yating Wang
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanyan Guo
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiangfeng Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianxiang Shi
- Precision Medicine Center, Henan Institute of Medical and Pharmaceutical Sciences & BGI College, Zhengzhou University, Zhengzhou, China
| | - Pei Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tao Hu
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ping Chen
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
104
|
Petit PX, Ardilla-Osorio H, Penalvia L, Nathan E. R. Tafazzin Mutation Affecting Cardiolipin Leads to Increased Mitochondrial Superoxide Anions and Mitophagy Inhibition in Barth Syndrome. Cells 2020; 9:cells9102333. [PMID: 33096711 PMCID: PMC7589545 DOI: 10.3390/cells9102333] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 01/01/2023] Open
Abstract
Tafazzin is a phospholipid transacylase that catalyzes the remodeling of cardiolipin, a mitochondrial phospholipid required for oxidative phosphorylation. Mutations of the tafazzin gene cause Barth syndrome, which is characterized by mitochondrial dysfunction and dilated cardiomyopathy, leading to premature death. However, the molecular mechanisms underlying the cause of mitochondrial dysfunction in Barth syndrome remain poorly understood. We again highlight the fact that the tafazzin deficiency is also linked to defective oxidative phosphorylation associated with oxidative stress. All the mitochondrial events are positioned in a context where mitophagy is a key element in mitochondrial quality control. Here, we investigated the role of tafazzin in mitochondrial homeostasis dysregulation and mitophagy alteration. Using a HeLa cell model of tafazzin deficiency, we show that dysregulation of tafazzin in HeLa cells induces alteration of mitophagy. Our findings provide some additional insights into mitochondrial dysfunction associated with Barth syndrome, but also show that mitophagy inhibition is concomitant with apoptosis dysfunction through the inability of abnormal mitochondrial cardiolipin to assume its role in cytoplasmic signal transduction. Our work raises hope that pharmacological manipulation of the mitophagic pathway together with mitochondrially targeted antioxidants may provide new insights leading to promising treatment for these highly lethal conditions.
Collapse
Affiliation(s)
- Patrice X. Petit
- SSPIN Saints-Pères Paris Institut de Neurosciences, CNRS UMR 8003, “Mitochondria, Apoptosis and Autophagy Signalling” Université de Paris—Campus Saint-Germain, 45 rue des Saints-Pères, 75006 Paris, France; (L.P.); (R.N.E.)
- Correspondence: or ; Tel.: +33(0)6-78-24-80-87
| | - Hector Ardilla-Osorio
- Laboratoire Cellules Souches et Prions, INSERM-S 1124, Université de Paris—Campus Saint-Germain, 45 rue des Saints Pères, 75006 Paris, France;
| | - Lucile Penalvia
- SSPIN Saints-Pères Paris Institut de Neurosciences, CNRS UMR 8003, “Mitochondria, Apoptosis and Autophagy Signalling” Université de Paris—Campus Saint-Germain, 45 rue des Saints-Pères, 75006 Paris, France; (L.P.); (R.N.E.)
| | - Rainey Nathan E.
- SSPIN Saints-Pères Paris Institut de Neurosciences, CNRS UMR 8003, “Mitochondria, Apoptosis and Autophagy Signalling” Université de Paris—Campus Saint-Germain, 45 rue des Saints-Pères, 75006 Paris, France; (L.P.); (R.N.E.)
| |
Collapse
|
105
|
How Reversible Are the Effects of Fumed Silica on Macrophages? A Proteomics-Informed View. NANOMATERIALS 2020; 10:nano10101939. [PMID: 33003391 PMCID: PMC7600894 DOI: 10.3390/nano10101939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/09/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Synthetic amorphous silica is one of the most used nanomaterials, and numerous toxicological studies have studied its effects. Most of these studies have used an acute exposure mode to investigate the effects immediately after exposure. However, this exposure modality does not allow the investigation of the persistence of the effects, which is a crucial aspect of silica toxicology, as exemplified by crystalline silica. In this paper, we extended the investigations by studying not only the responses immediately after exposure but also after a 72 h post-exposure recovery phase. We used a pyrolytic silica as the test nanomaterial, as this variant of synthetic amorphous silica has been shown to induce a more persistent inflammation in vivo than precipitated silica. To investigate macrophage responses to pyrolytic silica, we used a combination of proteomics and targeted experiments, which allowed us to show that most of the cellular functions that were altered immediately after exposure to pyrolytic silica at a subtoxic dose, such as energy metabolism and cell morphology, returned to normal at the end of the recovery period. However, some alterations, such as the inflammatory responses and some aldehyde detoxification proteins, were persistent. At the proteomic level, other alterations, such as proteins implicated in the endosomal/lysosomal pathway, were also persistent but resulted in normal function, thus suggesting cellular adaptation.
Collapse
|
106
|
Ranjan A, Kaushik I, Srivastava SK. Pimozide Suppresses the Growth of Brain Tumors by Targeting STAT3-Mediated Autophagy. Cells 2020; 9:2141. [PMID: 32971907 PMCID: PMC7563195 DOI: 10.3390/cells9092141] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Brain tumors are considered as one of the most aggressive and incurable forms of cancer. The majority of the patients with brain tumors have a median survival rate of 12%. Brain tumors are lethal despite the availability of advanced treatment options such as surgical removal, chemotherapy, and radiotherapy. In this study, we have evaluated the anti-cancer effects of pimozide, which is a neuroleptic drug used for the treatment of schizophrenia and chronic psychosis. Pimozide significantly reduced the proliferation of U-87MG, Daoy, GBM 28, and U-251MG brain cancer cell lines by inducing apoptosis with IC50 (Inhibitory concentration 50) ranging from 12 to 16 μM after 48 h of treatment. Our Western blotting analysis indicated that pimozide suppressed the phosphorylation of STAT3 at Tyr705 and Src at Tyr416, and it inhibited the expression of anti-apoptotic markers c-Myc, Mcl-1, and Bcl-2. Significant autophagy induction was observed with pimozide treatment. LC3B, Beclin-1, and ATG5 up-regulation along with autolysosome formation confirmed the induction of autophagy with pimozide treatment. Inhibiting autophagy using 3-methyladenine or LC3B siRNA significantly blocked the apoptosis-inducing effects of pimozide, suggesting that pimozide mediated its apoptotic effects by inducing autophagy. Oral administration of 25 mg/kg pimozide suppressed the intracranially implanted U-87MG tumor growth by 45% in athymic nude mice. The chronic administration of pimozide showed no general signs of toxicity, and the behavioral activity of the mice remained unchanged. Taken together, these results indicate that pimozide inhibits the growth of brain cancer by autophagy-mediated apoptosis.
Collapse
Affiliation(s)
- Alok Ranjan
- Department of Biomedical Science, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (A.R.); (I.K.)
| | - Itishree Kaushik
- Department of Biomedical Science, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (A.R.); (I.K.)
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Center for Tumor Immunology and Targeted Cancer Therapy, Abilene, TX 79601, USA
| | - Sanjay K. Srivastava
- Department of Biomedical Science, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (A.R.); (I.K.)
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Center for Tumor Immunology and Targeted Cancer Therapy, Abilene, TX 79601, USA
| |
Collapse
|
107
|
Csekes E, Vágvölgyi M, Hunyadi A, Račková L. Protoflavones in melanoma therapy: Prooxidant and pro-senescence effect of protoapigenone and its synthetic alkyl derivative in A375 cells. Life Sci 2020; 260:118419. [PMID: 32931795 DOI: 10.1016/j.lfs.2020.118419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
AIMS In our study, the anticancer effects of a semisynthetic p-quinol, protoapigenone 1'-O-butyl ether (PABut), were tested in human melanoma A375 cells also in comparison with natural congener, protoapigenone (PA). MAIN METHODS The cytotoxic effect of PABut and PA was determined using MTT assay. Flow cytometry analysis was used to evaluate the influence of the compounds tested on ROS generation and cell cycle distribution in A375 cells. Moreover, apoptosis was evaluated by AO/EB dual staining as well as by flow cytometry. Markers of senescence were quantified by spectrofluorimetry and by Western blot analysis. KEY FINDINGS Both PABut and PA showed significant cytotoxicity against melanoma A375 cells at sub-micromolar concentrations. Both protoflavones induced comparable cell cycle arrest in G2/M phase. However, a more profound upregulation of intracellular ROS levels was found following PABut treatment. An increased apoptosis in the cells following 48 h treatment with both protoflavones tested was also confirmed. Both compounds tested remarkably upregulated p21 protein levels in A375 cells. Unlike PA, PABut significantly decreased protein levels of NAD+-dependent deacetylase SirT1 and β-actin accompanied by mild significant upregulation of mitochondrial SOD2 and senescence markers, p16 protein and SA-β-Gal activity. However, a significant upregulation of p53 only following PA treatment was found. SIGNIFICANCE These results suggest that PABut and PA confer high chemotherapeutic potential in melanoma cells and are suitable for further testing. Furthermore, modification of protoapigenone with 1'-O-butyl ether moiety can be associated with improved senescence-inducing effect and, thus, enhanced chemotherapeutic potency of PABut compared to the unmodified natural protoflavone.
Collapse
Affiliation(s)
- Erika Csekes
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovak Republic
| | - Máté Vágvölgyi
- Institute of Pharmacognosy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
| | - Attila Hunyadi
- Institute of Pharmacognosy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
| | - Lucia Račková
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovak Republic.
| |
Collapse
|
108
|
Carlos JAEG, Lima K, Coelho-Silva JL, de Melo Alves-Paiva R, Moreno NC, Vicari HP, de Souza Santos FP, Hamerschlak N, Costa-Lotufo LV, Traina F, Machado-Neto JA. Reversine exerts cytotoxic effects through multiple cell death mechanisms in acute lymphoblastic leukemia. Cell Oncol (Dordr) 2020; 43:1191-1201. [PMID: 32857324 DOI: 10.1007/s13402-020-00551-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Acute lymphoblastic leukemia (ALL) is an aggressive hematological cancer with limited therapeutic options for adult patients. Aurora kinases have drawn attention as potential targets in hematological neoplasms due to their high expression and biological functions. Aurora kinase A (AURKA) and AURKB are essential for a successful mitosis, acting in spindle mitotic organization and cytokinesis. Reversine is a synthetic purine analog that acts as a multi-kinase inhibitor with anti-neoplastic activity by targeting AURKA and AURKB. METHODS ALL patient gene expression data were retrieved from the Amazonia! DATABASE For functional assays, Jurkat (T-ALL) and Namalwa (B-ALL) cells were exposed to increasing concentrations of reversine and submitted to various cellular and molecular assays. RESULTS We found that AURKB expression was higher in ALL patient samples compared to normal lymphocytes (p < 0.0001). The ALL cell lines tested displayed aberrant AURKA and AURKB expression. In Jurkat and Namalwa cells, reversine reduced cell viability in a dose- and time-dependent manner (p < 0.05). Reversine also significantly reduced the viability of primary ALL cells. Reversine induced apoptosis and autophagy, and reduced cell proliferation in both cell lines (p < 0.05). Mitotic catastrophe markers, including cell cycle arrest at G2/M, increased cell size and DNA damage, were observed upon reversine exposure. Short- and long-term treatment with reversine inhibited autonomous clonogenicity (p < 0.05). At the molecular level, reversine reduced AURKB activity, induced SQSTM1/p62 consumption, and increased LC3BII and γ-H2AX levels. In Namalwa cells, reversine modulated 25 out of 84 autophagy-related genes, including BCL2, BAD, ULK1, ATG10, IRGM and MAP1LC3B, which indicates that reversine acts by initiating and sustaining autophagy signals in ALL cells. CONCLUSIONS From our data we conclude that reversine reduces the viability of ALL cells by triggering multiple cell death mechanisms, including apoptosis, mitotic catastrophe, and autophagy. Our findings highlight reversine as a potential anticancer agent for ALL.
Collapse
Affiliation(s)
- Jorge Antonio Elias Godoy Carlos
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - Juan Luiz Coelho-Silva
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, SP, Brazil
| | | | - Natália Cestari Moreno
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | | | - Nelson Hamerschlak
- Einstein's Teaching and Research Institute, Albert Einstein Hospital, São Paulo, SP, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - Fabiola Traina
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, SP, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil.
| |
Collapse
|
109
|
Serum- and glucocorticoid- inducible kinase 2, SGK2, is a novel autophagy regulator and modulates platinum drugs response in cancer cells. Oncogene 2020; 39:6370-6386. [PMID: 32848212 PMCID: PMC7529585 DOI: 10.1038/s41388-020-01433-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 12/16/2022]
Abstract
For many tumor types chemotherapy still represents the therapy of choice and many standard treatments are based on the use of platinum (PT) drugs. However, de novo or acquired resistance to platinum is frequent and leads to disease progression. In Epithelial Ovarian Cancer (EOC) patients, PT-resistant recurrences are very common and improving the response to treatment still represents an unmet clinical need. To identify new modulators of PT-sensitivity, we performed a loss-of-function screening targeting 680 genes potentially involved in the response of EOC cells to platinum. We found that SGK2 (Serum-and Glucocorticoid-inducible kinase 2) plays a key role in PT-response. We show here that EOC cells relay on the induction of autophagy to escape PT-induced death and that SGK2 inhibition increases PT sensitivity inducing a block in the autophagy cascade due to the impairment of lysosomal acidification. Mechanistically we demonstrate that SGK2 controls autophagy in a kinase-dependent manner by binding and inhibiting the V-ATPase proton pump. Accordingly, SGK2 phosphorylates the subunit V1H (ATP6V1H) of V-ATPase and silencing or chemical inhibition of SGK2, affects the normal autophagic flux and sensitizes EOC cells to platinum. Hence, we identified a new pathway that links autophagy to the survival of cancer cells under platinum treatment in which the druggable kinase SGK2 plays a central role. Our data suggest that blocking autophagy via SGK2 inhibition could represent a novel therapeutic strategy to improve patients' response to platinum.
Collapse
|
110
|
Bernard M, Cardin GB, Cahuzac M, Ayad T, Bissada E, Guertin L, Bahig H, Nguyen-Tan PF, Filion E, Ballivy O, Soulieres D, Rodier F, Christopoulos A. Dual Inhibition of Autophagy and PI3K/AKT/MTOR Pathway as a Therapeutic Strategy in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12092371. [PMID: 32825725 PMCID: PMC7563873 DOI: 10.3390/cancers12092371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/12/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
Genomic analyses of head and neck squamous cell carcinoma (HNSCC) have highlighted alterations in the phosphatidylinositol 3-kinase (PI3K) signaling pathway, presenting a therapeutic target for multiple ongoing clinical trials with PI3K or PI3K/MTOR inhibitors. However, these inhibitors can potentially increase autophagy in HNSCC and indirectly support cancer cell survival. Here, we sought to understand the relationship between the PI3K signaling pathway and autophagy during their dual inhibition in a panel of HNSCC cell lines. We used acridine orange staining, immunoblotting, and tandem sensor Red Fluorescent Protein- Green Fluorescent Protein-, microtubule-associated protein 1 light chain 3 beta (RFP-GFP-LC3B) expression analysis to show that PI3K inhibitors increase autophagosomes in HNSCC cells, but that chloroquine treatment effectively inhibits the autophagy that is induced by PI3K inhibitors. Using the Bliss independence model, we determined that the combination of chloroquine with PI3K inhibitors works in synergy to decrease cancer cell proliferation, independent of the PIK3CA status of the cell line. Our results indicate that a strategy focusing on autophagy inhibition enhances the efficacy of therapeutics already in clinical trials. Our results suggest a broader application for this combination therapy that can be promptly translated to in vivo studies.
Collapse
Affiliation(s)
- Monique Bernard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Institut du Cancer de Montréal (ICM), Montreal, QC H2X 0A9, Canada
| | - Guillaume B. Cardin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Institut du Cancer de Montréal (ICM), Montreal, QC H2X 0A9, Canada
| | - Maxime Cahuzac
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Institut du Cancer de Montréal (ICM), Montreal, QC H2X 0A9, Canada
| | - Tareck Ayad
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (E.B.); (L.G.)
| | - Eric Bissada
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (E.B.); (L.G.)
| | - Louis Guertin
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (E.B.); (L.G.)
| | - Houda Bahig
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (P.F.N.-T.); (E.F.); (O.B.)
| | - Phuc Felix Nguyen-Tan
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (P.F.N.-T.); (E.F.); (O.B.)
| | - Edith Filion
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (P.F.N.-T.); (E.F.); (O.B.)
| | - Olivier Ballivy
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (P.F.N.-T.); (E.F.); (O.B.)
| | - Denis Soulieres
- Department of Medicine, Service of Hemato-Oncology, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada;
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Institut du Cancer de Montréal (ICM), Montreal, QC H2X 0A9, Canada
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Apostolos Christopoulos
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (M.B.); (G.B.C.); (M.C.); (T.A.); (H.B.); (F.R.)
- Institut du Cancer de Montréal (ICM), Montreal, QC H2X 0A9, Canada
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0A9, Canada; (E.B.); (L.G.)
- Correspondence: ; Tel.: +514-890-8000 (ext. 31292)
| |
Collapse
|
111
|
Ramesh G, Das S, Bola Sadashiva SR. Berberine, a natural alkaloid sensitizes human hepatocarcinoma to ionizing radiation by blocking autophagy and cell cycle arrest resulting in senescence. J Pharm Pharmacol 2020; 72:1893-1908. [PMID: 32815562 DOI: 10.1111/jphp.13354] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To study the radiosensitizing potential of Berberine and the underlying mechanism in human hepatocarcinoma (HepG2) cells. METHODS HepG2 cells were challenged with X-rays in combination with Berberine treatment and several in vitro assays were performed. Alteration in cell viability was determined by MTT assay. Changes in intracellular ROS levels, mitochondrial membrane potential/mass, intracellular acidic vesicular organelles as well as cell cycle arrest and apoptotic cell death were analysed by flow cytometry. Induction of autophagy was assessed by staining the cells with Monodansylcadaverine/Lysotracker red dyes and immunoblotting for LC3I/II and p62 proteins. Phase-contrast/fluorescence microscopy was employed to study mitotic catastrophe and senescence. Cellular senescence was confirmed by immunoblotting for p21 levels and ELISA for Interleukin-6. KEY FINDINGS X-rays + Berberine had a synergistic effect in reducing cell proliferation accompanied by a robust G2/M arrest. Berberine-mediated radiosensitization was associated with elevated levels of LC3II and p62 suggesting blocked autophagy that was followed by mitotic catastrophe and senescence. Treatment of cells with X-rays + Berberine resulted in increased oxidative stress, hyperpolarized mitochondria with increased mitochondrial mass and reduced ATP levels. CONCLUSIONS The study expands the understanding of the pharmacological properties of Berberine and its applicability as a radiosensitizer towards treating liver cancer.
Collapse
Affiliation(s)
- Gautham Ramesh
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shubhankar Das
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Satish Rao Bola Sadashiva
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
112
|
Zipporah E B, Patra B, Govarthanan K, Yadav R, Mohan S, Shyamsunder P, Verma RS. Defective cell proliferation is an attribute of overexpressed Notch1 receptor and impaired autophagy in Fanconi Anemia. Genomics 2020; 112:4628-4639. [PMID: 32800766 DOI: 10.1016/j.ygeno.2020.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/01/2020] [Accepted: 08/07/2020] [Indexed: 01/22/2023]
Abstract
Fanconi Anemia (FA) is an inherited bone marrow failure syndrome caused by mutation in FA pathway proteins, involved in Interstrand Cross Link (ICL) repair. FA cells exhibit in vitro proliferation arrest due to accumulated DNA damage, hence understanding the rescue mechanism that renders proliferation advantage is required. Gene expression profiling performed in FA patients Peripheral Blood Mononuclear Cells (PBMCs) revealed a wide array of dysregulated biological processes. Functional enrichment and gene clustering analysis showed crippled autophagy process and escalated Notch signalling pathway in FA clinical samples and cell lines. Notch pathway mediators overexpression were reverted in FANCA mutant cells when treated with Rapamycin, an autophagy inducer. Additionally, Rapamycin stabilized cell viability after treatment with the DNA damaging agent, MitomycinC (MMC) and enhanced cell proliferation genes expression in FANCA mutant cells. Inherently FANCA mutant cells express impaired autophagy; thus activation of autophagy channelizes Notch signalling cascade and sustains cell viability.
Collapse
Affiliation(s)
- Binita Zipporah E
- Stem Cell and Molecular Biology Lab, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India
| | - Bamadeb Patra
- Stem Cell and Molecular Biology Lab, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India
| | - Kavitha Govarthanan
- Stem Cell and Molecular Biology Lab, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India
| | - Rajesh Yadav
- Stem Cell and Molecular Biology Lab, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India
| | - Sheila Mohan
- Apollo Speciality hospital, 320 Padma complex, Anna Salai, Chennai 600 035, India; Registry for Fanconi Anemia in India (REFAIN), India
| | - Pavithra Shyamsunder
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 119077, Singapore
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Lab, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, Tamilnadu, India.
| |
Collapse
|
113
|
Kim SM, Ha SE, Lee HJ, Rampogu S, Vetrivel P, Kim HH, Venkatarame Gowda Saralamma V, Lee KW, Kim GS. Sinensetin Induces Autophagic Cell Death through p53-Related AMPK/mTOR Signaling in Hepatocellular Carcinoma HepG2 Cells. Nutrients 2020; 12:nu12082462. [PMID: 32824273 PMCID: PMC7468969 DOI: 10.3390/nu12082462] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/19/2022] Open
Abstract
Sinensetin (SIN) has been reported to exhibit anti-inflammatory and anti-cancer activity. However, the cellular and molecular mechanism by which SIN promotes hepatocellular carcinoma (HCC) cell death remains unclear. In the present study, we investigated the induction of cell death by SIN and its underlying mechanism in HepG2 cells, an HCC cell line. We found that SIN significantly induced cell death in HepG2 cells, whereas the proliferation rate of Thle2, human liver epithelial cells, was unaffected by SIN. SIN-treated HepG2 cells were not affected by apoptotic cell death; instead, autophagic cell death was induced through the p53-mediated AMPK/mTOR signaling pathway. Inhibition of p53 degradation led to both autophagy and apoptosis in HepG2 cells. p53 translocation led to SIN-induced autophagy, whereas p53 translocation inhibited SIN-induced apoptosis. However, SIN showed apoptosis in the p53-mutant Hep3B cell line. Molecular docking simulation of the p53 core domain showed effective binding with SIN, which was found significant compared with the known p53 activator, RITA. Collectively, these data suggest that SIN may be a potential anti-cancer agent targeting autophagic cell death in human liver cancer.
Collapse
Affiliation(s)
- Seong Min Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | - Sang Eun Ha
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | - Ho Jeong Lee
- Biological Resources Research Group, Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju 52834, Korea;
| | - Shailima Rampogu
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (K.W.L.)
| | - Preethi Vetrivel
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | - Hun Hwan Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | | | - Keun Woo Lee
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (K.W.L.)
| | - Gon Sup Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
- Correspondence: ; Tel.: +82-55-772-2346
| |
Collapse
|
114
|
Guizzardi S, Picotto G, Rodriguez V, Welsh J, Narvaez C, Bohl L, Tolosa de Talamoni N. Combined treatment of menadione and calcitriol increases the antiproliferative effect by promoting oxidative/nitrosative stress, mitochondrial dysfunction, and autophagy in breast cancer MCF-7 cells. Can J Physiol Pharmacol 2020; 98:548-556. [PMID: 32762631 DOI: 10.1139/cjpp-2019-0585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The aim of this study was to determine new insights into the molecular mechanisms involved in the antiproliferative action of menadione + calcitriol (MEN+D) on MCF-7 cells. After 24 h, MEN+D inhibited the cell growth but was not observed with each single treatment. The combined drugs reduced the mitochondrial respiration at that time, as judged by an increase in the proton leak and a decrease in the ATP generation and coupling efficiency. At longer times, 48 or 96 h, either D or MEN reduced the proliferation, but the effect was higher when both drugs were used together. The combined treatment increased the superoxide anion ([Formula: see text]) and nitric oxide (NO•) contents as well as acidic vesicular organelles (AVOs) formation. The percentage of cells showing the lower mitochondrial membrane potential (ΔΨm) was highly increased by the combined therapy. LC3-II protein expression was enhanced by any treatment. In conclusion, the antiproliferative action of MEN+D involves oxidative/nitrosative stress, mitochondrial alteration, and autophagy. This combined therapy could be useful to treat breast cancer cells because it inhibits multiple oncogenic pathways more effectively than each single agent.
Collapse
Affiliation(s)
- Solange Guizzardi
- Laboratorio "Dr. Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-UNC), Córdoba, Argentina
| | - Gabriela Picotto
- Laboratorio "Dr. Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-UNC), Córdoba, Argentina
| | - Valeria Rodriguez
- Laboratorio "Dr. Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-UNC), Córdoba, Argentina
| | - JoEllen Welsh
- University at Albany Cancer Research Center, Rensselaer, NY, USA
| | - Carmen Narvaez
- University at Albany Cancer Research Center, Rensselaer, NY, USA
| | - Luciana Bohl
- Centro de Investigaciones y Transferencia Villa María (CITVM-CONICET), Universidad Nacional Villa María, Villa María, Argentina
| | - Nori Tolosa de Talamoni
- Laboratorio "Dr. Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-UNC), Córdoba, Argentina
| |
Collapse
|
115
|
The Effects of Alpha-Linolenic Acid on the Secretory Activity of Astrocytes and β Amyloid-Associated Neurodegeneration in Differentiated SH-SY5Y Cells: Alpha-Linolenic Acid Protects the SH-SY5Y cells against β Amyloid Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8908901. [PMID: 32832007 PMCID: PMC7428942 DOI: 10.1155/2020/8908901] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/16/2020] [Accepted: 07/06/2020] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. Amyloid β- (Aβ-) induced mitochondrial dysfunction may be a primary process triggering all the cascades of events that lead to AD. Therefore, identification of natural factors and endogenous mechanisms that protect neurons against Aβ toxicity is needed. In the current study, we investigated whether alpha-linolenic acid (ALA), as a natural product, would increase insulin and IGF-I (insulin-like growth factor I) release from astrocytes. Moreover, we explored the protective effect of astrocytes-derived insulin/IGF-I on Aβ-induced neurotoxicity, with special attention paid to their impact on mitochondrial function of differentiated SH-SY5Y cells. The results showed that ALA induced insulin and IGF-I secretion from astrocytes. Our findings demonstrated that astrocyte-derived insulin/insulin-like growth factor I protects differentiated SH-SY5Y cells against Aβ1-42-induced cell death. Moreover, pretreatment with conditioned medium (CM) and ALA-preactivated CM (ALA-CM) protected the SH-SY5Y cells against Aβ1-42-induced mitochondrial dysfunction by reducing the depolarization of the mitochondrial membrane, increasing mitochondrial biogenesis, restoring the balance between fusion and fission processes, and regulation of mitophagy and autophagy processes. Our study suggested that astrocyte-derived insulin/insulin-like growth factor I suppresses Aβ1-42-induced cytotoxicity in the SH-SY5Y cells by protecting against mitochondrial dysfunction. Moreover, the neuroprotective effects of CM were intensified by preactivation with ALA.
Collapse
|
116
|
Simon Serrano S, Sime W, Abassi Y, Daams R, Massoumi R, Jemaà M. Inhibition of mitotic kinase Mps1 promotes cell death in neuroblastoma. Sci Rep 2020; 10:11997. [PMID: 32686724 PMCID: PMC7371706 DOI: 10.1038/s41598-020-68829-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022] Open
Abstract
Neuroblastoma is the most common paediatric cancer type. Patients diagnosed with high-risk neuroblastoma have poor prognosis and occasionally tumours relapse. As a result, novel treatment strategies are needed for relapse and refractory neuroblastoma patients. Here, we found that high expression of Mps1 kinase (mitotic kinase Monopolar Spindle 1) was associated with relapse-free neuroblastoma patient outcomes and poor overall survival. Silencing and inhibition of Mps1 in neuroblastoma or PDX-derived cells promoted cell apoptosis via the caspase-dependent mitochondrial apoptotic pathway. The mechanism of cell death upon Mps1 inhibition was dependent on the polyploidization/aneuploidization of the cells before undergoing mitotic catastrophe. Furthermore, tumour growth retardation was confirmed in a xenograft mouse model after Mps1-inhibitor treatment. Altogether, these results suggest that Mps1 expression and inhibition can be considered as a novel prognostic marker as well as a therapeutic strategy for the treatment of high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Sonia Simon Serrano
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Yasmin Abassi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| | - Mohamed Jemaà
- Department of Laboratory Medicine, Translational Cancer Research, Faculty of Medicine, Lund University, 22381, Lund, Sweden.
| |
Collapse
|
117
|
Sali VK, Mani S, Meenaloshani G, Velmurugan Ilavarasi A, Vasanthi HR. Type 5 17-hydroxysteroid dehydrogenase/prostaglandin F synthase (AKR1C3) inhibition and potential anti-proliferative activity of cholest-4-ene-3,6-dione in MCF-7 breast cancer cells. Steroids 2020; 159:108638. [PMID: 32209376 DOI: 10.1016/j.steroids.2020.108638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/10/2020] [Accepted: 03/18/2020] [Indexed: 12/28/2022]
Abstract
Cholest-4-ene-3,6-dione (KS) is a cholesterol oxidation product which exhibits anti-proliferative activity. However, its precise mechanism of action remains unknown. In this study, the effects of KS on AKR1C3 inhibition and anti-proliferative activities were investigated in the hormone-dependent MCF-7 breast cancer cells. We identified that KS arrested the enzymatic conversion of estrone to 17-β estradiol, by inhibiting AKR1C3 in intact MCF-7 cells. The anti-proliferative effects of KS were evaluated by MTT assay, acridine orange and ethidium bromide dual staining, cell cycle analysis and Western blotting. KS arrested the cell cycle progression in the G1 phase with a concomitant increase of the Sub-G0 population to increase in concentration and time. It also enhanced the p53 and NFkB expression and induced caspase-12, 9 and 3 processing and down-regulated the Bcl-2 expression. Molecular docking studies performed to understand the inhibition mechanism of KS on AKR1C3 revealed that KS occupied the binding region of AKR1C3 with almost similar orientation as indomethacin (IM), thereby acting as an antagonistic agent for AKR1C3. Based on the results it is identified that KS induces inhibition of AKR1C3 and cell death in MCF-7 cells. These results indicate that KS can be used as a molecular scaffold for further development of novel small-molecules with better specificity towards AKR1C3.
Collapse
Affiliation(s)
- Veeresh Kumar Sali
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - Sugumar Mani
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - G Meenaloshani
- National College (Autonomous), Tiruchirappalli, Tamil Nadu 620001, India
| | | | - Hannah R Vasanthi
- Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry 605014, India.
| |
Collapse
|
118
|
In Vivo Suppression of Autophagy via Lentiviral shRNA Targeting Atg5 Improves Lupus-Like Syndrome. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8959726. [PMID: 32462028 PMCID: PMC7212279 DOI: 10.1155/2020/8959726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 12/01/2022]
Abstract
In both mouse models and clinical patients with lupus, autophagy levels were significantly elevated and correlated with disease activity. Furthermore, autophagy can promote the survival of B and T cells, plasma cell differentiation, and antibody production. These results suggest that autophagy may promote the progression of lupus by regulating the survival of autoreactive immune cells. Therefore, we aimed at studying whether suppressing autophagy can modulate lupus progression in vivo. First, we found that the autophagy levels in splenocytes and lymphocytes of peripheral blood (PB) were elevated and positively correlated with disease severity in lupus-prone mice. The shAtg5-lentivirus, which effectively inhibits autophagy in vitro, was then injected into the lupus-prone mice. Autophagy levels in lymph node cells and PB lymphocytes were reduced following Atg5 suppression. We also found that lymphadenopathy and the numbers of plasma cells, CD4−CD8−, and CD4+ T cells decreased in mice treated with the shAtg5-lentivirus. The mice treated with shAtg5-lentivirus exhibited lower levels of proteinuria, serum anti-dsDNA antibody, B-cell activating factor (BAFF), and glomerular immune complex deposition. Therefore, targeting autophagy to moderate overactivated autophagy in immune cells seems to be a novel strategy for combination therapy of lupus.
Collapse
|
119
|
Solari JIG, Filippi-Chiela E, Pilar ES, Nunes V, Gonzalez EA, Figueiró F, Andrade CF, Klamt F. Damage-associated molecular patterns (DAMPs) related to immunogenic cell death are differentially triggered by clinically relevant chemotherapeutics in lung adenocarcinoma cells. BMC Cancer 2020; 20:474. [PMID: 32456685 PMCID: PMC7251700 DOI: 10.1186/s12885-020-06964-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
Background Chemotherapeutics can stimulate immune antitumor response by inducing immunogenic cell death (ICD), which is activated by Damage-Associated Molecular Patterns (DAMPs) like the exposure of calreticulin (CRT) on the cell surface, the release of ATP and the secretion of High Mobility Group Box 1 (HMGB1). Methods Here, we investigated the levels of ICD-associated DAMPs induced by chemotherapeutics commonly used in the clinical practice of non-small cell lung cancer (NSCLC) and the association of these DAMPs with apoptosis and autophagy. A549 human lung adenocarcinoma cells were treated with clinically relevant doses of cisplatin, carboplatin, etoposide, paclitaxel and gemcitabine. We assessed ICD-associated DAMPs, cell viability, apoptosis and autophagy in an integrated way. Results Cisplatin and its combination with etoposide induced the highest levels of apoptosis, while etoposide was the less pro-apoptotic treatment. Cisplatin also induced the highest levels of ICD-associated DAMPs, which was not incremented by co-treatments. Etoposide induced the lower levels of ICD and the highest levels of autophagy, suggesting that the cytoprotective role of autophagy is dominant in relation to its pro-ICD role. High levels of CRT were associated with better prognosis in TCGA databank. In an integrative analysis we found a strong positive correlation between DAMPs and apoptosis, and a negative correlation between cell number and ICD-associated DAMPs as well as between autophagy and apoptosis markers. We also purpose a mathematical integration of ICD-associated DAMPs in an index (IndImunnog) that may represent with greater biological relevance this process. Cisplatin-treated cells showed the highest IndImmunog, while etoposide was the less immunogenic and the more pro-autophagic treatment. Conclusions Cisplatin alone induced the highest levels of ICD-associated DAMPs, so that its combination with immunotherapy may be a promising therapeutic strategy in NSCLC.
Collapse
Affiliation(s)
- José Ignácio Gonzalez Solari
- Programa de Pós-Graduação em Ciências Pneumológicas, Laboratório de Pulmão e Vias Aéreas, FAMED/UFRGS, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eduardo Filippi-Chiela
- Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil. .,Hospital de Clínicas Porto Alegre, Unidade de Pesquisas Experimental (Laboratório de Células, Tecidos e Genes), Porto Alegre, Brazil.
| | - Emily Salles Pilar
- Hospital de Clínicas Porto Alegre, Unidade de Pesquisas Experimental (Laboratório de Células, Tecidos e Genes), Porto Alegre, Brazil
| | - Vitória Nunes
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Esteban Alberto Gonzalez
- Hospital de Clínicas Porto Alegre, Unidade de Pesquisas Experimental (Laboratório de Células, Tecidos e Genes), Porto Alegre, Brazil
| | - Fabrício Figueiró
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cristiano Feijó Andrade
- Programa de Pós-Graduação em Ciências Pneumológicas, Laboratório de Pulmão e Vias Aéreas, FAMED/UFRGS, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Fábio Klamt
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
120
|
Cheng L, Wang H, Wang Z, Huang H, Zhuo D, Lin J. Leflunomide Inhibits Proliferation and Induces Apoptosis via Suppressing Autophagy and PI3K/Akt Signaling Pathway in Human Bladder Cancer Cells. Drug Des Devel Ther 2020; 14:1897-1908. [PMID: 32546957 PMCID: PMC7244359 DOI: 10.2147/dddt.s252626] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Bladder cancer is a lethal human malignancy. Currently, treatment for bladder cancer is limited. The anti-tumor effects of leflunomide have attracted much more concern in multiple human cancers. MATERIALS AND METHODS This study evaluated the anti-tumor effects of leflunomide on cell viability, colony formation, apoptosis, and cell cycle in two human bladder carcinoma cell lines, 5637 and T24. Meanwhile, the underlying mechanism including PI3K/Akt signaling pathway and autophagy modulation was also identified. RESULTS Leflunomide markedly inhibited the growth of both bladder cancer cell lines and induced apoptosis and cell cycle arrest in S phase. The phosphorylation levels of Akt and P70S6K in both cell lines were significantly down-regulated with leflunomide treatment. Furthermore, the deceased formation of autophagosomes and the accumulation of LC3II and P62 suggested the blockade of autophagy by leflunomide. Modulation of autophagy with rapamycin and chloroquine markedly attenuated and enhanced the cytostatic effects of leflunomide, respectively. CONCLUSION Leflunomide significantly reduced the cell viability of bladder cancer cells via inducing apoptosis and cell cycle arrest and suppressing the PI3K/Akt signaling pathway. In addition, the blockade of autophagy was observed, and autophagy inhibition enhanced leflunomide-mediating anti-tumor effects. Our data presented here offer novel ideas for comprehensive therapeutic regimes on bladder cancer.
Collapse
Affiliation(s)
- Li Cheng
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Hao Wang
- Department of Geriatrics, Peking University First Hospital, Beijing, People’s Republic of China
| | - Zicheng Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Houbao Huang
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Dong Zhuo
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, People’s Republic of China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, People’s Republic of China
| |
Collapse
|
121
|
Vicencio E, Cordero EM, Cortés BI, Palominos S, Parra P, Mella T, Henrríquez C, Salazar N, Monasterio G, Cafferata EA, Murgas P, Vernal R, Cortez C. Aggregatibacter Actinomycetemcomitans Induces Autophagy in Human Junctional Epithelium Keratinocytes. Cells 2020; 9:E1221. [PMID: 32423042 PMCID: PMC7290389 DOI: 10.3390/cells9051221] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 12/23/2022] Open
Abstract
The adverse environmental conditions found in the periodontium during periodontitis pathogenesis stimulate local autophagy responses, mainly due to a continuous inflammatory response against the dysbiotic subgingival microbiome. The junctional epithelium represents the main site of the initial interaction between the host and the dysbiotic biofilm. Here, we investigated the role of autophagy in junctional epithelium keratinocytes (JEKs) in response to Aggregatibacter actinomycetemcomitans or its purified lipopolysaccharides (LPS). Immunofluorescence confocal analysis revealed an extensive nuclear translocation of transcription factor EB (TFEB) and consequently, an increase in autophagy markers and LC3-turnover assessed by immunoblotting and qRT-PCR. Correspondingly, challenged JEKs showed a punctuate cytosolic profile of LC3 protein contrasting with the diffuse distribution observed in untreated controls. Three-dimensional reconstructions of confocal images displayed a close association between intracellular bacteria and LC3-positive vesicles. Similarly, a close association between autophagic vesicles and the protein p62 was observed in challenged JEKs, indicating that p62 is the main adapter protein recruited during A. actinomycetemcomitans infection. Finally, the pharmacological inhibition of autophagy significantly increased the number of bacteria-infected cells as well as their death, similar to treatment with LPS. Our results indicate that A. actinomycetemcomitans infection induces autophagy in JEKs, and this homeostatic process has a cytoprotective effect on the host cells during the early stages of infection.
Collapse
Affiliation(s)
- Emiliano Vicencio
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (B.I.C.); (P.M.)
| | - Esteban M. Cordero
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- Parasitology Section, Instituto de Salud Pública de Chile, Avenida Marathon 1000, Ñuñoa 7780050, Chile
| | - Bastián I. Cortés
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (B.I.C.); (P.M.)
| | - Sebastián Palominos
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Dentistry, Faculty of Science, Universidad Mayor, Avenida Libertador Bernardo O’higgins 2013, Huechuraba 8580745, Chile
| | - Pedro Parra
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Dentistry, Faculty of Science, Universidad Mayor, Avenida Libertador Bernardo O’higgins 2013, Huechuraba 8580745, Chile
| | - Tania Mella
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| | - Constanza Henrríquez
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| | - Nelda Salazar
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| | - Gustavo Monasterio
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, Independencia 8380492, Chile; (G.M.); (E.A.C.)
| | - Emilio A. Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, Independencia 8380492, Chile; (G.M.); (E.A.C.)
- Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Av. Paseo de la República 5544, Lima 15074, Peru
| | - Paola Murgas
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (B.I.C.); (P.M.)
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, Independencia 8380492, Chile; (G.M.); (E.A.C.)
| | - Cristian Cortez
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Dentistry, Faculty of Science, Universidad Mayor, Avenida Libertador Bernardo O’higgins 2013, Huechuraba 8580745, Chile
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| |
Collapse
|
122
|
Chen C, Kuo YH, Lin CC, Chao CY, Pai MH, Chiang EPI, Tang FY. Decyl caffeic acid inhibits the proliferation of colorectal cancer cells in an autophagy-dependent manner in vitro and in vivo. PLoS One 2020; 15:e0232832. [PMID: 32401800 PMCID: PMC7219744 DOI: 10.1371/journal.pone.0232832] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/22/2020] [Indexed: 01/18/2023] Open
Abstract
The treatment of human colorectal cancer (CRC) cells through suppressing the abnormal survival signaling pathways has recently become a significant area of focus. In this study, our results demonstrated that decyl caffeic acid (DC), one of the novel caffeic acid derivatives, remarkedly suppressed the growth of CRC cells both in vitro and in vivo. The inhibitory effects of DC on CRC cells were investigated in an in vitro cell model and in vivo using a xenograft mouse model. CRC cells were treated with DC at various dosages (0, 10, 20 and 40 μM), and cell survival, the apoptotic index and the autophagy level were measured using an MTT assay and flow cytometry analysis, respectively. The signaling cascades in CRC were examined by Western blot assay. The anti-cancer effects of DC on tumor growth were examined by using CRC HCT-116 cells implanted in an animal model. Our results indicated that DC differentially suppressed the growth of CRC HT-29 and HCT-116 cells through an enhancement of cell-cycle arrest at the S phase. DC inhibited the expression of cell-cycle regulators, which include cyclin E and cyclin A proteins. The molecular mechanisms of action were correlated to the blockade of the STAT3 and Akt signaling cascades. Strikingly, a high dosage of DC prompted a self-protection action through inducing cell-dependent autophagy in HCT-116 cells. Suppression of autophagy induced cell death in the treatment of DC in HCT-116 cells. DC seemed to inhibit cell proliferation of CRC differentially, and the therapeutic advantage appeared to be autophagy dependent. Moreover, consumption of DC blocked the tumor growth of colorectal adenocarcinoma in an experimental animal model. In conclusion, our results suggested that DC could act as a therapeutic agent through the significant suppression of tumor growth of human CRC cells.
Collapse
Affiliation(s)
- Ching Chen
- Biomedical Science Laboratory, Department of Nutrition, China Medical University, Taichung, Taiwan, Republic of China
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Republic of China
- Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China
| | - Cheng-Chieh Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan, Republic of China
- Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan, Republic of China
- Department of Healthcare Administration, College of Health Science, Asia University, Taichung, Taiwan, Republic of China
| | - Che-Yi Chao
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan, Republic of China
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
| | - Man-Hui Pai
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - En-Pei Isabel Chiang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Innovation and Development Center of Sustainable Agriculture (IDCSA), National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Feng-Yao Tang
- Biomedical Science Laboratory, Department of Nutrition, China Medical University, Taichung, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
123
|
Azimee S, Rahmati M, Fahimi H, Moosavi MA. TiO 2 nanoparticles enhance the chemotherapeutic effects of 5-fluorouracil in human AGS gastric cancer cells via autophagy blockade. Life Sci 2020; 248:117466. [PMID: 32101760 DOI: 10.1016/j.lfs.2020.117466] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 02/15/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
Abstract
AIMS Nanoparticles (NPs)-based drugs have been recently introduced to improve the efficacy of current therapeutic strategies for the treatment of cancer; however, the molecular mechanisms by which a NP interacts with cellular systems still need to be delineated. Here, we utilize the autophagic potential of TiO2 NPs for improving chemotherapeutic effects of 5-fluorouracil (5-FU) in human AGS gastric cells. MATERIALS AND METHODS Cell growth and viability were determined by trypan blue exclusion test and MTT assay, respectively. Vesicular organelles formation was evaluated by acridine orange staining of cells. Cell cycle and apoptosis were monitored by flow cytometry. Reactive oxygen species (ROS) level were measured by DCHF-DA staining. Autophagy was examined by q-PCR and western blotting. Molecular docking was used for studying NP interaction with autophagic proteins. KEY FINDINGS TiO2 NPs increase ROS production, impair lysosomal function and subsequently block autophagy flux in AGS cells. In addition, the autophagy blockade induced by non-toxic concentrations of TiO2 NPs (1 μg/ml) can promote cytotoxic and apoptotic effects of 5-FU in AGS cells. SIGNIFICANCE These results confirm the beneficial effects of TiO2 NPs in combination with chemotherapy in in vitro model of gastric cancer, which may pave the way to develop a possible solution to circumvent chemoresistance in cancer.
Collapse
Affiliation(s)
- Shiva Azimee
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 14965/161, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Fahimi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 14965/161, Tehran, Iran.
| |
Collapse
|
124
|
Spinelli R, Barrero Guevara LA, López JA, Camargo CM, Groot de Restrepo H, Siano AS. Cytotoxic and antiproliferative activities of amphibian (anuran) skin extracts on human acute monocytic leukemia cells. Toxicon 2020; 177:25-34. [PMID: 31982457 DOI: 10.1016/j.toxicon.2020.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 11/25/2022]
Abstract
The use of preparations derived from frog skins for curative purposes antedates research history and is perpetuated in current medicine. The skins of anuran's (frogs and toads) are a rich source of compounds with a great importance in the search of antibiotics, analgesics, immunomodulators, enzymatic inhibitors and antitumoral agents applying to human health. Nowadays, cancer is the second most common cause of mortality with more than 8.2 million of deaths worldwide per year. Acute monocytic leukemia is the subtype M5 of acute myeloid leukemia (AML) a cancer type with reduced survival rates in patients. The monocyte to macrophage differentiation plays an essential role increasing the expansion of AML cell lines. Herein we studied the cytotoxic and antiproliferative activities of eleven amphibian species of three families belonging to Argentinean zones, against THP-1 monocytes and THP-1 macrophages acute monocytic leukemia cell lines. The evaluated species showed pronounced deleterious effects on acute monocytic leukemia THP-1 cell lines, reducing cell proliferation and inducing apoptosis, autophagy and in some cases cell aggregation. Being this work of great importance for the study of new natural anti-cancer compounds.
Collapse
Affiliation(s)
- Roque Spinelli
- Departmento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional Del Litoral, Paraje El pozo, 3000, Santa Fe, Argentina
| | - Laura Andrea Barrero Guevara
- Laboratorio de Genética Humana, Departamento de Ciencias Biológicas, Universidad de Los Andes, Cr. 1 Nº 18A-10 Building M1, 110321, Bogotá, Colombia
| | - Javier Alejandro López
- Instituto Nacional de Limnología, UNL-CONICET. Ciudad Universitaria, Paraje El Pozo, 3000, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Carolina Muñoz Camargo
- Departamento de Ingeniería Biomédica, Facultad de Ingeniería, Universidad de los Andes, Cr. 1 Este Nº 19A-40 Building ML, 110321, Bogotá, Colombia
| | - Helena Groot de Restrepo
- Laboratorio de Genética Humana, Departamento de Ciencias Biológicas, Universidad de Los Andes, Cr. 1 Nº 18A-10 Building M1, 110321, Bogotá, Colombia.
| | - Alvaro Sebastián Siano
- Departmento de Química Orgánica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional Del Litoral, Paraje El pozo, 3000, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
125
|
Zhou X, Bouitbir J, Liechti ME, Krähenbühl S, Mancuso RV. Hyperthermia Increases Neurotoxicity Associated with Novel Methcathinones. Cells 2020; 9:cells9040965. [PMID: 32295288 PMCID: PMC7227000 DOI: 10.3390/cells9040965] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/29/2020] [Accepted: 04/09/2020] [Indexed: 12/16/2022] Open
Abstract
Hyperthermia is one of the severe acute adverse effects that can be caused by the ingestion of recreational drugs, such as methcathinones. The effect of hyperthermia on neurotoxicity is currently not known. The primary aim of our study was therefore to investigate the effects of hyperthermia (40.5 °C) on the neurotoxicity of methcathinone (MC), 4-chloromethcathinone (4-CMC), and 4-methylmethcathinone (4-MMC) in SH-SY5Y cells. We found that 4-CMC and 4-MMC were cytotoxic (decrease in cellular ATP and plasma membrane damage) under both hyper- (40.5 °C) and normothermic conditions (37 °C), whereby cells were more sensitive to the toxicants at 40.5 °C. 4-CMC and 4-MMC impaired the function of the mitochondrial electron transport chain and increased mitochondrial formation of reactive oxygen species (ROS) in SH-SY5Y cells, which were accentuated under hyperthermic conditions. Hyperthermia was associated with a rapid expression of the 70 kilodalton heat shock protein (Hsp70), which partially prevented cell death after 6 h of exposure to the toxicants. After 24 h of exposure, autophagy was stimulated by the toxicants and by hyperthermia but could only partially prevent cell death. In conclusion, hyperthermic conditions increased the neurotoxic properties of methcathinones despite the stimulation of protective mechanisms. These findings may be important for the understanding of the mechanisms and clinical consequences of the neurotoxicity associated with these compounds.
Collapse
Affiliation(s)
- Xun Zhou
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Jamal Bouitbir
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Swiss Centre for Applied Human Toxicology, 4031 Basel, Switzerland
| | - Matthias E. Liechti
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Swiss Centre for Applied Human Toxicology, 4031 Basel, Switzerland
- Correspondence: ; Tel.: +41-61-265-4715
| | - Riccardo V. Mancuso
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, 4031 Basel, Switzerland; (X.Z.); (J.B.); (M.E.L.); (R.V.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
126
|
Tarnowski L, Collados Rodriguez M, Brzywczy J, Cysewski D, Wawrzynska A, Sirko A. Overexpression of the Selective Autophagy Cargo Receptor NBR1 Modifies Plant Response to Sulfur Deficit. Cells 2020; 9:E669. [PMID: 32164165 PMCID: PMC7140714 DOI: 10.3390/cells9030669] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
Plants exposed to sulfur deficit elevate the transcription of NBR1 what might reflect an increased demand for NBR1 in such conditions. Therefore, we investigated the role of this selective autophagy cargo receptor in plant response to sulfur deficit (-S). Transcriptome analysis of the wild type and NBR1 overexpressing plants pointed out differences in gene expression in response to -S. Our attention focused particularly on the genes upregulated by -S in roots of both lines because of significant overrepresentation of cytoplasmic ribosomal gene family. Moreover, we noticed overrepresentation of the same family in the set of proteins co-purifying with NBR1 in -S. One of these ribosomal proteins, RPS6 was chosen for verification of its direct interaction with NBR1 and proven to bind outside the NBR1 ubiquitin binding domains. The biological significance of this novel interaction and the postulated role of NBR1 in ribosomes remodeling in response to starvation remain to be further investigated. Interestingly, NBR1 overexpressing seedlings have significantly shorter roots than wild type when grown in nutrient deficient conditions in the presence of TOR kinase inhibitors. This phenotype probably results from excessive autophagy induction by the additive effect of NBR1 overexpression, starvation, and TOR inhibition.
Collapse
Affiliation(s)
- Leszek Tarnowski
- Department of Plant Biochemistry, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland
| | - Milagros Collados Rodriguez
- Department of Plant Biochemistry, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland
| | - Jerzy Brzywczy
- Department of Plant Biochemistry, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland
| | - Dominik Cysewski
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland
| | - Anna Wawrzynska
- Department of Plant Biochemistry, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland
| | - Agnieszka Sirko
- Department of Plant Biochemistry, Institute of Biochemistry and Biophysics Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland
| |
Collapse
|
127
|
Akhtar MJ, Ahamed M, Alrokayan SA, Ramamoorthy MM, Alaizeri ZM. High Surface Reactivity and Biocompatibility of Y 2O 3 NPs in Human MCF-7 Epithelial and HT-1080 FibroBlast Cells. Molecules 2020; 25:molecules25051137. [PMID: 32138335 PMCID: PMC7179248 DOI: 10.3390/molecules25051137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 01/16/2023] Open
Abstract
This study aimed to generate a comparative data on biological response of yttrium oxide nanoparticles (Y2O3 NPs) with the antioxidant CeO2 NPs and pro-oxidant ZnO NPs. Sizes of Y2O3 NPs were found to be in the range of 35±10 nm as measured by TEM and were larger from its hydrodynamic sizes in water (1004 ± 134 nm), PBS (3373 ± 249 nm), serum free culture media (1735 ± 305 nm) and complete culture media (542 ± 108 nm). Surface reactivity of Y2O3 NPs with bovine serum albumin (BSA) was found significantly higher than for CeO2 and ZnO NPs. The displacement studies clearly suggested that adsorption to either BSA, filtered serum or serum free media was quite stable, and was dependent on whichever component interacted first with the Y2O3 NPs. Enzyme mimetic activity, like that of CeO2 NPs, was not detected for the NPs of Y2O3 or ZnO. Cell viability measured by MTT and neutral red uptake (NRU) assays suggested Y2O3 NPs were not toxic in human breast carcinoma MCF-7 and fibroblast HT-1080 cells up to the concentration of 200 μg/mL for a 24 h treatment period. Oxidative stress markers suggested Y2O3 NPs to be tolerably non-oxidative and biocompatible. Moreover, mitochondrial potential determined by JC-1 as well as lysosomal activity determined by lysotracker (LTR) remained un-affected and intact due to Y2O3 and CeO2 NPs whereas, as expected, were significantly induced by ZnO NPs. Hoechst-PI dual staining clearly suggested apoptotic potential of only ZnO NPs. With high surface reactivity and biocompatibility, NPs of Y2O3 could be a promising agent in the field of nanomedicine.
Collapse
Affiliation(s)
- Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia (M.M.R.)
- Correspondence: ; Tel.: +966-146-96075
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia (M.M.R.)
| | - Salman A. Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | | | - ZabnAllah M. Alaizeri
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
128
|
Akimov Y, Bulanova D, Timonen S, Wennerberg K, Aittokallio T. Improved detection of differentially represented DNA barcodes for high-throughput clonal phenomics. Mol Syst Biol 2020; 16:e9195. [PMID: 32187448 PMCID: PMC7080434 DOI: 10.15252/msb.20199195] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular DNA barcoding has become a popular approach to study heterogeneity of cell populations and to identify clones with differential response to cellular stimuli. However, there is a lack of reliable methods for statistical inference of differentially responding clones. Here, we used mixtures of DNA-barcoded cell pools to generate a realistic benchmark read count dataset for modelling a range of outcomes of clone-tracing experiments. By accounting for the statistical properties intrinsic to the DNA barcode read count data, we implemented an improved algorithm that results in a significantly lower false-positive rate, compared to current RNA-seq data analysis algorithms, especially when detecting differentially responding clones in experiments with strong selection pressure. Building on the reliable statistical methodology, we illustrate how multidimensional phenotypic profiling enables one to deconvolute phenotypically distinct clonal subpopulations within a cancer cell line. The mixture control dataset and our analysis results provide a foundation for benchmarking and improving algorithms for clone-tracing experiments.
Collapse
Affiliation(s)
- Yevhen Akimov
- Institute for Molecular Medicine Finland (FIMM)HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Daria Bulanova
- Institute for Molecular Medicine Finland (FIMM)HiLIFEUniversity of HelsinkiHelsinkiFinland
- Biotech Research and Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem)University of CopenhagenCopenhagenDenmark
| | - Sanna Timonen
- Institute for Molecular Medicine Finland (FIMM)HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM)HiLIFEUniversity of HelsinkiHelsinkiFinland
- Biotech Research and Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem)University of CopenhagenCopenhagenDenmark
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM)HiLIFEUniversity of HelsinkiHelsinkiFinland
- Department of Mathematics and StatisticsUniversity of TurkuTurkuFinland
- Department of Cancer GeneticsInstitute for Cancer ResearchOslo University HospitalOsloNorway
- Oslo Centre for Biostatistics and Epidemiology (OCBE)Faculty of MedicineUniversity of OsloOsloNorway
| |
Collapse
|
129
|
Highlighting Curcumin-Induced Crosstalk between Autophagy and Apoptosis as Supported by Its Specific Subcellular Localization. Cells 2020; 9:cells9020361. [PMID: 32033136 PMCID: PMC7072416 DOI: 10.3390/cells9020361] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 12/31/2022] Open
Abstract
Curcumin, a major active component of turmeric (Curcuma longa, L.), is known to have various effects on both healthy and cancerous tissues. In vitro studies suggest that curcumin inhibits cancer cell growth by activating apoptosis, but the mechanism underlying the anticancer effect of curcumin is still unclear. Since there is a recent consensus about endoplasmic reticulum (ER) stress being involved in the cytotoxicity of natural compounds, we have investigated using Image flow cytometry the mechanistic aspects of curcumin's destabilization of the ER, but also the status of the lysosomal compartment. Curcumin induces ER stress, thereby causing an unfolded protein response and calcium release, which destabilizes the mitochondrial compartment and induce apoptosis. These events are also associated with secondary lysosomal membrane permeabilization that occurs later together with an activation of caspase-8, mediated by cathepsins and calpains that ended in the disruption of mitochondrial homeostasis. These two pathways of different intensities and momentum converge towards an amplification of cell death. In the present study, curcumin-induced autophagy failed to rescue all cells that underwent type II cell death following initial autophagic processes. However, a small number of cells were rescued (successful autophagy) to give rise to a novel proliferation phase.
Collapse
|
130
|
Mohammadalipour Z, Rahmati M, Khataee A, Moosavi MA. Differential effects of N-TiO 2 nanoparticle and its photo-activated form on autophagy and necroptosis in human melanoma A375 cells. J Cell Physiol 2020; 235:8246-8259. [PMID: 31989650 DOI: 10.1002/jcp.29479] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022]
Abstract
The manipulation of autophagy provides a new opportunity for highly effective anticancer therapies. Recently, we showed that photodynamic therapy (PDT) with nitrogen-doped titanium dioxide (N-TiO2 ) nanoparticles (NPs) could promote the reactive oxygen species (ROS)-dependent autophagy in leukemia cells. However, the differential autophagic effects of N-TiO2 NPs in the dark and light conditions and the potential of N-TiO2- based PDT for the treatment of melanoma cells remain unknown. Here we show that depending on the visible-light condition, the autophagic response of human melanoma A375 cells to N-TiO2 NPs switches between two different statuses (ie., flux or blockade) with the opposite outcomes (ie., survival or death). Mechanistically, low doses of N-TiO2 NPs (1-100 µg/ml) stimulate a nontoxic autophagy flux response in A375 cells, whereas their photo-activation leads to the impairment of the autophagosome-lysosome fusion, the blockade of autophagy flux and consequently the induction of RIPK1-mediated necroptosis via ROS production. These results confirm that photo-controllable autophagic effects of N-TiO2 NPs can be utilized for the treatment of cancer, particularly melanoma.
Collapse
Affiliation(s)
- Zahra Mohammadalipour
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.,Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering, Near East University, Nicosia, North Cyprus, Turkey
| | - Mohammad A Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
131
|
Chung LY, Tang SJ, Wu YC, Yang KC, Huang HJ, Sun GH, Sun KH. Platinum-based combination chemotherapy triggers cancer cell death through induction of BNIP3 and ROS, but not autophagy. J Cell Mol Med 2019; 24:1993-2003. [PMID: 31856355 PMCID: PMC6991685 DOI: 10.1111/jcmm.14898] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/16/2019] [Accepted: 11/24/2019] [Indexed: 12/25/2022] Open
Abstract
These days, cancer can still not be effectively cured because cancer cells readily develop resistance to anticancer drugs. Therefore, an effective combination of drugs with different mechanisms to prevent drug resistance has become a very important issue. Furthermore, the BH3‐only protein BNIP3 is involved in both apoptotic and autophagic cell death. In this study, lung cancer cells were treated with a chemotherapy drug alone or in combination to identify the role of BNIP3 and autophagy in combination chemotherapy for treating cancer. Our data revealed that various combinational treatments of two drugs could increase cancer cell death and cisplatin in combination with rapamycin or LBH589, which triggered the cell cycle arrest at the S phase. Cells with autophagosome and pEGFP‐LC3 puncta increased when treated with drugs. To confirm the role of autophagy, cancer cells were pre‐treated with the autophagy inhibitor 3‐methyladenine (3‐MA). 3‐MA sensitized cancer cells to chemotherapy drug treatments. These results suggest that autophagy may be responsible for cell survival in combination chemotherapy for lung cancer. Moreover, BNIP3 was induced and localized in mitochondria when cells were treated with drugs. The transfection of a dominant negative transmembrane deletion construct of BNIP3 (BNIP3ΔTM) and treatment of a reactive oxygen species (ROS) inhibitor suppressed chemo drug‐induced cell death. These results indicate that BNIP3 and ROS may be involved in combination chemo drug‐induced cell death. However, chemo drug‐induced autophagy may protect cancer cells from drug cytotoxicity. As a result, inhibiting autophagy may improve the effects of combination chemotherapy when treating lung cancer.
Collapse
Affiliation(s)
- Ling-Yen Chung
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, and Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Shye-Jye Tang
- Institute of Marine Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Yi-Ching Wu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, and Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Kai-Chi Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, and Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Hui-Ju Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, and Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Guang-Huan Sun
- Division of Urology, Department of Surgery, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Kuang-Hui Sun
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, and Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| |
Collapse
|
132
|
Wang L, Huang W, Zhan J. Grape Seed Proanthocyanidins Induce Autophagy and Modulate Survivin in HepG2 Cells and Inhibit Xenograft Tumor Growth in Vivo. Nutrients 2019; 11:E2983. [PMID: 31817589 PMCID: PMC6950679 DOI: 10.3390/nu11122983] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 01/02/2023] Open
Abstract
Liver cancer is one of the leading causes of death worldwide. Although radiotherapy and chemotherapy are effective in general, they present various side effects, significantly limiting the curative effect. Increasing evidence has shown that the dietary intake of phytochemicals plays an essential role in the chemoprevention or chemotherapy of tumors. In this work, HepG2 cells and nude mice with HepG2-derived xenografts were treated with grape seed proanthocyanidins (GSPs). The results showed that GSPs induced autophagy, and inhibition of autophagy increased apoptosis in HepG2 cells. In addition, GSPs also reduced the expression of survivin. Moreover, survivin was involved in GSPs-induced apoptosis. GSPs at 100 mg/kg and 200 mg/kg significantly inhibited the growth of HepG2 cells in nude mice without causing observable toxicity and autophagy, while inducing the phosphorylation of mitogen-activated protein kinase (MAPK) pathway-associated proteins, p-JNK, p-ERK and p-p38 MAPK and reducing the expression of survivin. These results suggested that GSPs might be promising phytochemicals against liver cancer.
Collapse
Affiliation(s)
| | | | - Jicheng Zhan
- Beijing Key Laboratory of Viticulture and Enology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (L.W.); (W.H.)
| |
Collapse
|
133
|
Wang J, Rousseau J, Kim E, Ehresmann S, Cheng YT, Duraine L, Zuo Z, Park YJ, Li-Kroeger D, Bi W, Wong LJ, Rosenfeld J, Gleeson J, Faqeih E, Alkuraya FS, Wierenga KJ, Chen J, Afenjar A, Nava C, Doummar D, Keren B, Juusola J, Grompe M, Bellen HJ, Campeau PM. Loss of Oxidation Resistance 1, OXR1, Is Associated with an Autosomal-Recessive Neurological Disease with Cerebellar Atrophy and Lysosomal Dysfunction. Am J Hum Genet 2019; 105:1237-1253. [PMID: 31785787 PMCID: PMC6904826 DOI: 10.1016/j.ajhg.2019.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 11/01/2019] [Indexed: 12/28/2022] Open
Abstract
We report an early-onset autosomal-recessive neurological disease with cerebellar atrophy and lysosomal dysfunction. We identified bi-allelic loss-of-function (LoF) variants in Oxidative Resistance 1 (OXR1) in five individuals from three families; these individuals presented with a history of severe global developmental delay, current intellectual disability, language delay, cerebellar atrophy, and seizures. While OXR1 is known to play a role in oxidative stress resistance, its molecular functions are not well established. OXR1 contains three conserved domains: LysM, GRAM, and TLDc. The gene encodes at least six transcripts, including some that only consist of the C-terminal TLDc domain. We utilized Drosophila to assess the phenotypes associated with loss of mustard (mtd), the fly homolog of OXR1. Strong LoF mutants exhibit late pupal lethality or pupal eclosion defects. Interestingly, although mtd encodes 26 transcripts, severe LoF and null mutations can be rescued by a single short human OXR1 cDNA that only contains the TLDc domain. Similar rescue is observed with the TLDc domain of NCOA7, another human homolog of mtd. Loss of mtd in neurons leads to massive cell loss, early death, and an accumulation of aberrant lysosomal structures, similar to what we observe in fibroblasts of affected individuals. Our data indicate that mtd and OXR1 are required for proper lysosomal function; this is consistent with observations that NCOA7 is required for lysosomal acidification.
Collapse
Affiliation(s)
- Julia Wang
- Program in Developmental Biology, Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Justine Rousseau
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada
| | - Emily Kim
- Biochemistry and Cell Biology, Rice University, Houston, TX 77005, USA
| | - Sophie Ehresmann
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada
| | - Yi-Ting Cheng
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lita Duraine
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ye-Jin Park
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - David Li-Kroeger
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weimin Bi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lee-Jun Wong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jill Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joseph Gleeson
- Rady Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Eissa Faqeih
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 11525, Saudi Arabia
| | - Klaas J Wierenga
- Department of Pediatrics, Oklahoma University Health Sciences Center (OUHSC), Oklahoma City, OK 26901, USA; Department of Clinical Genomics, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Jiani Chen
- Department of Pediatrics, Oklahoma University Health Sciences Center (OUHSC), Oklahoma City, OK 26901, USA; Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandra Afenjar
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012, France; Département de Génétique et Embryologie Médicale, CRMR des Malformations et Maladies Congénitales du Cervelet, GRC ConCer-LD, Sorbonne Universités, Hôpital Trousseau, Paris, 75012 France
| | - Caroline Nava
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012, France
| | - Diane Doummar
- Assistance Publique des Hôpitaux de Paris, Service de Neuropédiatrie, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012 France
| | - Boris Keren
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012, France
| | | | - Markus Grompe
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97201, USA; Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97201, USA
| | - Hugo J Bellen
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute and Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Philippe M Campeau
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
134
|
Darzynkiewicz Z, Halicka DH, Zhao H, Li J. Assessment of DNA Susceptibility to Denaturation as a Marker of Chromatin Structure. ACTA ACUST UNITED AC 2019; 91:e65. [PMID: 31763788 DOI: 10.1002/cpcy.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The susceptibility of DNA in situ to denaturation is modulated by its interactions with histone and nonhistone proteins, as well as with other chromatin components related to the maintenance of the 3D nuclear structure. Measurement of DNA proclivity to denature by cytometry provides insight into chromatin structure and thus can be used to recognize cells in different phases of the cell cycle, including mitosis, quiescence (G0 ), and apoptosis, as well as to identify the effects of drugs that modify chromatin structure. Particularly useful is the method's ability to detect chromatin changes in sperm cells related to DNA fragmentation and infertility. This article presents a flow cytometric procedure for assessing DNA denaturation based on application of the metachromatic property of acridine orange (AO) to differentially stain single- versus double-stranded DNA. This approach circumvents limitations of biochemical methods of examining DNA denaturation, in particular the fact that the latter destroy higher orders of chromatin structure and that, being applied to bulk cell populations, they cannot detect heterogeneity of individual cells. Because the metachromatic properties of AO have also found application in other cytometric procedures, such as differential staining of RNA versus DNA and assessment of lysosomal proton pump including autophagy, to avert confusion between these approaches and the use of this dye in the DNA denaturation assay, these AO applications are briefly outlined in this unit as well. © 2019 by John Wiley & Sons, Inc. Basic Protocol: Differential staining of single- versus double-stranded DNA with acridine orange.
Collapse
Affiliation(s)
| | - Dorota H Halicka
- Brander Cancer Research Institute, New York Medical College, Valhalla, New York
| | - Hong Zhao
- Brander Cancer Research Institute, New York Medical College, Valhalla, New York
| | - Jiangwei Li
- Brander Cancer Research Institute, New York Medical College, Valhalla, New York
| |
Collapse
|
135
|
Rabiee Motmaen S, Tavakol S, Joghataei MT, Barati M. Acidic pH derived from cancer cells as a double-edged knife modulates wound healing through DNA repair genes and autophagy. Int Wound J 2019; 17:137-148. [PMID: 31714008 DOI: 10.1111/iwj.13248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/18/2019] [Accepted: 09/22/2019] [Indexed: 12/16/2022] Open
Abstract
Wound healing is a sequester program that involves diverse cell signalling cascades. Notwithstanding, complete signal transduction pathways underpinning acidic milieu derived from cancer cells is not clear, yet. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, fluorescein diacetate/propidium iodide staining, and cell cycle flow cytometry revealed that acidic media decreased cell viability and cell number along with enhanced dead cells and S-phase arrest in normal fibroblasts. Notably, the trends of intracellular reactive oxygen species production and lactate dehydrogenase release significantly increased with time. It seems the downregulation of Klf4 is in part due to acidosis-induced DNA damage. It promoted cells towards S-phase arrest and diminished cell proliferation. Klf4 downregulation had a direct correlation with the P53 level while acidic microenvironment promotes cells towards cell death mechanisms including apoptosis and autophagy. Noteworthily, the unchanged levels of Rb and Mlh1 indicated in those genes had no dominant role in the repairing of DNA damage in fibroblasts treated with the acidic microenvironment. Therefore, cells owing to not entering to mitosis and accumulation of DNA damage were undergone cell death to preserve cell homeostasis. Since acidic media decreased the level of tumour suppressor and DNA repair genes and altered the normal survival pathways in fibroblasts, caution should be exercised to not lead to cancer rather than wound healing.
Collapse
Affiliation(s)
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad T Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Barati
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
136
|
Gao X, Wang Y, Li Y, Wang Y, Yan M, Sun H, Chen S, Pan X. Huganpian, a traditional chinese medicine, inhibits liver cancer growth in vitro and in vivo by inducing autophagy and cell cycle arrest. Biomed Pharmacother 2019; 120:109469. [PMID: 31698319 DOI: 10.1016/j.biopha.2019.109469] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/03/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Huganpian (HGP), a traditional chinese medicine composed of 6 herbs, possesses excellent therapeutic effects in clinical application. In this study, we aimed to elucidate the anti-tumor activity and the underlying mechanisms of HGP in liver cancer. The results of this study indicated that HGP effectively inhibited liver cancer growth in vitro and in vivo in a dose-dependent manner. Mechanistically, HGP exerted its anti-tumor effects by triggering autophagy with increased LC3Ⅱ and beclin1 levels and arrested the cell cycle on G0-G1 phase by downregulating the expressions of cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 4 (CDK4) and cyclinE1 in vitro and in vivo. Meanwhile, HGP did not induce apoptosis significantly. Importantly, we also confirmed that there were fewer side effects of HGP on immune system. Taken together, our findings suggest for the first time that HGP may become a promising drug or adjuvant drug with a lower toxicity for liver cancer treatment in the future.
Collapse
Affiliation(s)
- Xue Gao
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Yuyang Wang
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining 272067, China
| | - Yuxin Li
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Yansong Wang
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Miaomiao Yan
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Hongliu Sun
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Shayan Chen
- Department of Laboratory Science, Tianjin Medical University NanKai Hospital, Tianjin 300100, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Medical University NanKai Hospital, Tianjin 300100, China.
| | - Xiaohong Pan
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
137
|
Ou C, Liu H, Ding Z, Zhou L. Chloroquine promotes gefitinib‑induced apoptosis by inhibiting protective autophagy in cutaneous squamous cell carcinoma. Mol Med Rep 2019; 20:4855-4866. [PMID: 31638204 PMCID: PMC6854599 DOI: 10.3892/mmr.2019.10734] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 07/26/2019] [Indexed: 12/29/2022] Open
Abstract
Aberrant expression of the epidermal growth factor receptor (EGFR) plays vital roles in tumor development and progression. In the present study, ultraviolet irradiation induced the upregulation of EGFR in skin-derived keratinocytes, which may contribute to the development of cutaneous squamous cell carcinoma (CSCC). This was supported by the high expression of EGFR in CSCC clinical samples. Treating A431 CSCC cells with gefitinib, a tyrosine kinase inhibitor, activated the intrinsic mitochondrial apoptotic pathway while inducing protective autophagy. Combined application of chloroquine with gefitinib enhanced the treatment efficacy of gefitinib against CSCC by inhibiting autophagic flux. These findings demonstrated that autophagy inhibition may be an effective strategy for enhancing the sensitivity of EGFR-expressing cells to tyrosine kinase inhibitor treatment. Manipulating pro-survival autophagy by the combined application of chloroquine with gefitinib is a promising approach for improving the efficacy of EGFR inhibitors in cancer treatment. This may contribute to novel EGFR-targeted therapeutic strategies in the near future.
Collapse
Affiliation(s)
- Chengshan Ou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hongxia Liu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhenhua Ding
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Liang Zhou
- Department of Toxicology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
138
|
Serbian I, Siewert B, Al-Harrasi A, Csuk R. 2-O-(2-chlorobenzoyl) maslinic acid triggers apoptosis in A2780 human ovarian carcinoma cells. Eur J Med Chem 2019; 180:457-464. [DOI: 10.1016/j.ejmech.2019.07.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/28/2019] [Accepted: 07/15/2019] [Indexed: 02/08/2023]
|
139
|
Martinez-Lopez A, Persaud M, Chavez MP, Zhang H, Rong L, Liu S, Wang TT, Sarafianos SG, Diaz-Griffero F. Glycosylated diphyllin as a broad-spectrum antiviral agent against Zika virus. EBioMedicine 2019; 47:269-283. [PMID: 31501074 PMCID: PMC6796538 DOI: 10.1016/j.ebiom.2019.08.060] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/24/2019] [Accepted: 08/24/2019] [Indexed: 11/27/2022] Open
Abstract
Background Flaviviruses such as Zika cause sporadic pandemic outbreaks worldwide. There is an urgent need for anti-Zika virus (ZIKV) drugs to prevent mother-to-child transmission of ZIKV, new infections in high-risk populations, and the infection of medical personnel in ZIKV-affected areas. Methods Here, we showed that the small molecule 6-deoxyglucose-diphyllin (DGP) exhibited anti-ZIKV activity both in vitro and in vivo. DGP potently blocked ZIKV infection across all human and monkey cell lines tested. DGP also displayed broad-spectrum antiviral activity against other flaviviruses. Remarkably, DGP prevented ZIKV-induced mortality in mice lacking the type I interferon receptor (Ifnar1−/−). Cellular and virological experiments showed that DGP blocked ZIKV at a pre-fusion step or during fusion, which prevented the delivery of viral contents into the cytosol of the target cell. Mechanistic studies revealed that DGP prevented the acidification of endosomal/lysosomal compartments in target cells, thus inhibiting ZIKV fusion with cellular membranes and infection. Findings These investigations revealed that DGP inhibits ZIKV infection in vitro and in vivo. Interpretation The small molecule DGP has great potential for preclinical studies and the ability to inhibit ZIKV infection in humans.
Collapse
Affiliation(s)
- Alicia Martinez-Lopez
- Department of Microbiology and Immunology, Albert Einstein College of Medicine Bronx, NY 10461, USA
| | - Mirjana Persaud
- Department of Microbiology and Immunology, Albert Einstein College of Medicine Bronx, NY 10461, USA
| | - Maritza Puray Chavez
- Laboratory of Biochemical Pharmacology Emory University, Emory University, Atlanta, GA 30322, USA
| | - Hongjie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong SAR, People's Republic of China
| | - Lijun Rong
- Microbiology and Immunology College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - Shufeng Liu
- Laboratory of Vector-borne Viral Diseases, Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Tony T Wang
- Laboratory of Vector-borne Viral Diseases, Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology Emory University, Emory University, Atlanta, GA 30322, USA
| | - Felipe Diaz-Griffero
- Department of Microbiology and Immunology, Albert Einstein College of Medicine Bronx, NY 10461, USA.
| |
Collapse
|
140
|
Kurze AK, Buhs S, Eggert D, Oliveira-Ferrer L, Müller V, Niendorf A, Wagener C, Nollau P. Immature O-glycans recognized by the macrophage glycoreceptor CLEC10A (MGL) are induced by 4-hydroxy-tamoxifen, oxidative stress and DNA-damage in breast cancer cells. Cell Commun Signal 2019; 17:107. [PMID: 31455323 PMCID: PMC6712659 DOI: 10.1186/s12964-019-0420-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/14/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ligands of the C-type lectin CLEC10A such as Tn and sialyl-Tn representing early intermediates of O-glycosylation are hallmarks of many human malignancies. A variety of regulatory mechanisms underlying their expression are being discussed. METHODS CLEC10A ligands were detected in various tissues and cells using the recombinant glycan-binding domain of CLEC10A. In normal breast and endometrium, presence of ligands was correlated to the female cycle. Estrogen- and stress dependent induction of CLEC10A ligands was analyzed in MCF7 and T47D cells exposed to 4-hydroxy-tamoxifen (Tam), zeocin and hydrogen peroxide. The expression and localization of CLEC10A ligands was analyzed by Western blot and immunofluorescence. In breast cancer patients CLEC10A ligand expression and survival was correlated by Kaplan-Meyer analysis. RESULT We observed binding of CLEC10A in normal endometrial and breast tissues during the late phase of the female hormonal cycle suggesting a suppressive effect of female sex hormones on CLEC10A ligand expression. Accordingly, CLEC10A ligands were induced in MCF7- and T47D breast cancer cells after Tam treatment and accumulated on the cell surface and in the endosomal/lysosomal compartment. Phagocytosis experiments indicate that macrophages preferentially internalize CLEC10A ligands coated beads and Tam treated MCF7 cells. CLEC10A ligands were also expressed after the addition of zeocin and hydrogen-peroxide. Each substance induced the production of ROS indicating reactive oxygen species as a unifying mechanism of CLEC10A ligand induction. Mechanistically, increased expression of GalNAc-transferase 6 (GalNT6) and translocation of GalNT2 and GalNT6 from cis- towards trans-Golgi compartment was observed, while protein levels of COSMC and T-synthase remained unaffected. In breast cancer patients, positivity for CLEC10A staining in tumor tissues was associated with improved outcome and survival. CONCLUSION CLEC10A ligands are inducible by hormone depletion, 4-hydroxy-tamoxifen and agents inducing DNA damage and oxidative stress. Our results indicate that CLEC10A acts as a receptor for damaged and dead cells and may play an important role in the uptake of cell debris by macrophages and dendritic cells.
Collapse
Affiliation(s)
- Anna-Katharina Kurze
- Research Institute Children's Cancer Center and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Sophia Buhs
- Research Institute Children's Cancer Center and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Dennis Eggert
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Leticia Oliveira-Ferrer
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Axel Niendorf
- MVZ Prof. Dr. med. A. Niendorf Pathologie Hamburg-West GmbH, Institut für Histologie, Zytologie und molekulare Diagnostik, Lornsenstraße 4, 22767, Hamburg, Germany
| | - Christoph Wagener
- Research Institute Children's Cancer Center and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Peter Nollau
- Research Institute Children's Cancer Center and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany.
| |
Collapse
|
141
|
Adipose-derived stromal cell secretome disrupts autophagy in glioblastoma. J Mol Med (Berl) 2019; 97:1491-1506. [PMID: 31401659 DOI: 10.1007/s00109-019-01829-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 07/21/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
Mesenchymal stromal cells (MSCs) are frequently recruited to tumor sites to play a part in the tumor microenvironment (TME). However, their real impact on cancer cell behavior remains obscure. Here we investigated the effects of human adipose-derived stromal cell (hADSC) secretome in autophagy of glioblastoma (GBM), as a way to better comprehend how hADSCs influence the TME. GBM U-87 MG cells were treated with conditioned medium (CM) from hADSCs and autophagic flux was evaluated. hADSC CM treatment blocked the autophagic flux in tumor cells, as indicated by the accumulation of autophagosomes in the cytosol, the high LC3-II and p62/SQSTM1 protein levels, and the lack of increase in the amount of acidic vesicular organelles. These effects were further detected in other GBM cell lines tested and also in co-cultures of hADSCs and U-87 MG. hADSC CM did not compromise lysosomal acidification; however, it was able to activate mTORC1 signaling and, as a consequence, led to a decrease in the nuclear translocation of TFEB, a master transcriptional regulator of lysosomal biogenesis and autophagy, thereby contributing to a defective autophagic process. hADSCs secrete transforming growth factor beta 1 (TGFβ1) and this cytokine is an important mediator of CM effects on autophagy. A comprehensive knowledge of MSC roles in tumor biology is of great importance to shed light on the complex dialog between these cells and to explore such interactions therapeutically. The present results help to elucidate the paracrine effects of MSCs in tumors and bring attention to the potential to be explored in MSC secretome. KEY MESSAGES: hADSC secretome specifically affects the biology of GBM cells. hADSCs block the late steps of autophagic flux in GBM cells. hADSC secretome activates mTORC1 signaling and reduces TFEB nuclear translocation in GBM cells.
Collapse
|
142
|
Dutta D, Paul B, Mukherjee B, Mondal L, Sen S, Chowdhury C, Debnath MC. Nanoencapsulated betulinic acid analogue distinctively improves colorectal carcinoma in vitro and in vivo. Sci Rep 2019; 9:11506. [PMID: 31395908 PMCID: PMC6687831 DOI: 10.1038/s41598-019-47743-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/17/2019] [Indexed: 12/19/2022] Open
Abstract
Betulinic acid, a plant secondary metabolite, has gained significant attention due to its antiproliferative activity over a range of cancer cells. A promising betulinic acid analogue (2c) with better therapeutic efficacy than parent molecule to colon carcinoma cells has been reported. Despite impressive biological applications, low aqueous solubility and bioavailability create difficulties for its therapeutic applications. To overcome these lacunae and make it as a promising drug candidate we have encapsulated the lead betulinic acid derivative (2c) in a polymeric nanocarrier system (2c-NP) and evaluated its in vitro and in vivo therapeutic efficacy. Apoptosis that induces in vitro antiproliferative activity was significantly increased by 2c-NP compared to free-drug (2c), as assured by MTT assay, Annexin V positivity, JC1 analysis and cell cycle study. The therapeutic potential measured in vitro and in vivo reflects ability of 2c-NP as an effective therapeutic agent for treatment of colon carcinoma and future translation to clinical trials.
Collapse
Affiliation(s)
- Debasmita Dutta
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Brahamacharry Paul
- Infectious Diseases and Immunology Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Biswajit Mukherjee
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Laboni Mondal
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Suparna Sen
- Organic and Medicinal Chemistry Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Chinmay Chowdhury
- Organic and Medicinal Chemistry Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Mita Chatterjee Debnath
- Infectious Diseases and Immunology Division, CSIR- Indian Institute of Chemical Biology, Kolkata, 700032, India
| |
Collapse
|
143
|
Arun RP, Sivanesan D, Patra B, Varadaraj S, Verma RS. Simulated microgravity increases polyploid giant cancer cells and nuclear localization of YAP. Sci Rep 2019; 9:10684. [PMID: 31337825 PMCID: PMC6650394 DOI: 10.1038/s41598-019-47116-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/25/2019] [Indexed: 12/18/2022] Open
Abstract
Physical cues are vital in determining cellular fate in cancer. In vitro 3D culture do not replicate forces present in vivo. These forces including tumor interstitial fluid pressure and matrix stiffness behave as switches in differentiation and metastasis, which are intricate features of cancer stem cells (CSCs). Gravity determines the effect of these physical factors on cell fate and functions as evident from microgravity experiments on space and ground simulations. Here, we described the role of simulation of microgravity (SMG) using rotary cell culture system (RCCS) in increasing stemness in human colorectal cancer cell HCT116. We observed distinct features of cancer stem cells including CD133/CD44 dual positive cells and migration in SMG which was not altered by autophagy induction or inhibition. 3D and SMG increased autophagy, but the flux was staggered under SMG. Increased unique giant cancer cells housing complete nuclear localization of YAP were observed in SMG. This study highlights the role of microgravity in regulating stemness in CSC and importance of physical factors in determining the same.
Collapse
Affiliation(s)
- Raj Pranap Arun
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036 TN, India
| | - Divya Sivanesan
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036 TN, India
| | - Bamadeb Patra
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036 TN, India
| | - Sudha Varadaraj
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036 TN, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036 TN, India.
| |
Collapse
|
144
|
Machado-Neto JA, Coelho-Silva JL, Santos FPDS, Scheucher PS, Campregher PV, Hamerschlak N, Rego EM, Traina F. Autophagy inhibition potentiates ruxolitinib-induced apoptosis in JAK2 V617F cells. Invest New Drugs 2019; 38:733-745. [PMID: 31286322 DOI: 10.1007/s10637-019-00812-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
JAK2V617F can mimic growth factor signaling, leading to PI3K/AKT/mTOR activation and inhibition of autophagy. We hypothesized that selective inhibition of JAK1/2 by ruxolitinib could induce autophagy and limit drug efficacy in myeloproliferative neoplasms (MPN). Therefore, we investigated the effects of ruxolitinib treatment on autophagy-related genes and cellular processes, to determine the potential benefit of autophagy inhibitors plus ruxolitinib in JAK2V617F cells, and to verify the frequency and clinical impact of autophagy-related gene mutations in patients with MPNs. In SET2 JAK2V617F cells, ruxolitinib treatment induced autophagy and modulated 26 out of 79 autophagy-related genes. Ruxolitinib treatment reduced the expressions of important autophagy regulators, including mTOR/p70S6K/4EBP1 and the STAT/BCL2 axis, in a dose- and time-dependent manner. Pharmacological inhibition of autophagy was able to significantly suppress ruxolitinib-induced autophagy and increased ruxolitinib-induced apoptosis. Mutations in autophagy-related genes were found in 15.5% of MPN patients and were associated with increased age and a trend towards worse survival. In conclusion, ruxolitinib induces autophagy in JAK2V617F cells, potentially by modulation of mTOR-, STAT- and BCL2-mediated signaling. This may lead to inhibition of apoptosis. Our results suggest that the combination of ruxolitinib with pharmacological inhibitors of autophagy, such as chloroquine, may be a promising strategy to treat patients with JAK2V617F-mutated MPNs.
Collapse
Affiliation(s)
- João Agostinho Machado-Neto
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Av. Bandeirante, Ribeirão Preto, SP, 3900, Brazil.,Department of Pharmacology, Institute of Biomedical Sciences of the University of São Paulo, São Paulo, Brazil
| | - Juan Luiz Coelho-Silva
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Av. Bandeirante, Ribeirão Preto, SP, 3900, Brazil
| | - Fábio Pires de Souza Santos
- Centro de Oncologia e Hematologia Familia Dayan-Daycoval, Hospital Israelita Albert Einstein São Paulo, São Paulo, Brazil.,Instituto de Ensino e Pesquisa, Hospital Israelita Albert Einstein São Paulo, São Paulo, Brazil
| | - Priscila Santos Scheucher
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Av. Bandeirante, Ribeirão Preto, SP, 3900, Brazil
| | - Paulo Vidal Campregher
- Centro de Oncologia e Hematologia Familia Dayan-Daycoval, Hospital Israelita Albert Einstein São Paulo, São Paulo, Brazil.,Instituto de Ensino e Pesquisa, Hospital Israelita Albert Einstein São Paulo, São Paulo, Brazil
| | - Nelson Hamerschlak
- Centro de Oncologia e Hematologia Familia Dayan-Daycoval, Hospital Israelita Albert Einstein São Paulo, São Paulo, Brazil
| | - Eduardo Magalhães Rego
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Av. Bandeirante, Ribeirão Preto, SP, 3900, Brazil
| | - Fabiola Traina
- Department of Medical Images, Hematology and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Av. Bandeirante, Ribeirão Preto, SP, 3900, Brazil.
| |
Collapse
|
145
|
de Miranda Ramos V, Gasparotto J, Figueiró F, de Fraga Dias A, Rostirolla DC, Somensi N, da Rosa HT, Grun LK, Barbé-Tuana FM, Gelain DP, Moreira JCF. Retinoic acid downregulates thiol antioxidant defences and homologous recombination while promotes A549 cells sensitization to cisplatin. Cell Signal 2019; 62:109356. [PMID: 31288066 DOI: 10.1016/j.cellsig.2019.109356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/24/2019] [Accepted: 07/05/2019] [Indexed: 01/09/2023]
Abstract
Recent studies have investigated the use of retinoic acid (RA) molecule in combined chemotherapies to cancer cells as an attempt to increase treatment efficiency and circumvent cell resistance. Positive results were obtained in clinical trials from lung cancer patients treated with RA and cisplatin. Meanwhile, the signalling process that results from the interaction of both molecules remains unclear. One of the pathways that RA is able to modulate is the activity of NRF2 transcription factor, which is highly associated with tumour progression and resistance. Therefore, the aim of this work was to investigate molecular mechanism of RA and cisplatin co-treatment in A549 cells, focusing in NRF2 pathway. To this end, we investigated NRF2 and NRF2-target genes expression, cellular redox status, cisplatin-induced apoptosis, autophagy and DNA repair through homologous recombination. RA demonstrated to have an inhibitory effect over NRF2 activation, which regulates the expression of thiol antioxidants enzymes. Moreover, RA increased reactive species production associated with increased oxidation of thiol groups within the cells. The expression of proteins associated with DNA repair through homologous recombination was also suppressed by RA pre-treatment. All combined, these effects appear to create a more sensitive cellular environment to cisplatin treatment, increasing apoptosis frequency. Interestingly, autophagy was also increased by combination therapy, suggesting a resistance mechanism by A549 cells. In conclusion, these results provided new information about molecular mechanisms of RA and cisplatin treatment contributing to chemotherapy optimization.
Collapse
Affiliation(s)
- Vitor de Miranda Ramos
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Juciano Gasparotto
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Departamento de Civil y Ambiental, Universidad de la Costa, Barranquilla, Atlántico, Colombia; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fabrício Figueiró
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratório de Sinalização Purinérgica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Amanda de Fraga Dias
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Laboratório de Sinalização Purinérgica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Diana Carolina Rostirolla
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Nauana Somensi
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Helen Tais da Rosa
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas Kich Grun
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Group of Inflammation and Cellular Senescence, School of Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Brazil
| | - Florencia María Barbé-Tuana
- Group of Inflammation and Cellular Senescence, School of Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Brazil; Postgraduate Program in Cellular and Molecular Biology, School of Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Brazil
| | - Daniel Pens Gelain
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil; Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
146
|
High-throughput quantitative detection of basal autophagy and autophagic flux using image cytometry. Biotechniques 2019; 67:70-73. [PMID: 31238709 DOI: 10.2144/btn-2019-0044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Quantitative assessment of changes in macro-autophagy is often performed through manual quantification of the number of LC3B foci in immunofluorescence microscopy images. This method is highly laborious, subject to image-field selection and foci-counting bias, and is not sensitive for analyzing changes in basal autophagy. Alternative methods such as flow cytometry and transmission electron microscopy require highly specialized, expensive instruments and time-consuming sample preparation. Immunoblots are prone to exposure-related variations and noise that prevents accurate quantification. We report a high-throughput, inexpensive, reliable and objective method for studying basal level and flux changes in late-stage autophagy using image cytometry and acridine orange staining.
Collapse
|
147
|
Aldaghi SA, Jalal R. Concentration-Dependent Dual Effects of Ciprofloxacin on SB-590885-Resistant BRAF V600E A375 Melanoma Cells. Chem Res Toxicol 2019; 32:645-658. [PMID: 30829029 DOI: 10.1021/acs.chemrestox.8b00335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BRAF inhibitors (BRAFi) have been applied to treat melanoma harboring V600E mutations. Several studies showed that BRAFi-resistant melanomas are dependent on mitochondrial biogenesis. Therefore, the present study aimed to investigate the influence of ciprofloxacin (CIP), a mitochondria-targeting antibiotic, on SB-590885-resistant BRAFV600E A375 melanoma (A375/SB) cells. The cytotoxicity activity of CIP and SB-590885, a potent and specific BRAFi, on A375 and A375/SB cells was evaluated by MTT, colony formation, migration, and spheroid formation assays. Moreover, SB-590885-induced cell death in A375 cells was analyzed. SB-590885 showed time- and concentration-dependent cytotoxic effects on A375 cells. Twenty-five μg/mL CIP decreased the cell viability of A375 and A375/SB cells in a time-dependent manner. This concentration of CIP markedly decreased clonogenicity in both cells and caused a reduction in the growth of A375/SB spheroids. The cytotoxicity of 5 μg/mL CIP on A375/SB cells was less than that of A375 cells. The colony formation and migration ability of A375/SB cells was increased in the presence of 5 μg/mL CIP. Ten μM SB-590885 induced a massive vacuolization in A375 cells. Cell death assays suggested a simultaneous activation of autophagy, paraptosis, apoptosis, and necrosis. For the first time, this study reveals that CIP at the maximum concentration in serum (5 μg/mL) can enhance the colony formation and migration abilities in BRAFi-resistant melanoma cells, while it has cytotoxic activity against these cells at a higher concentration than serum level. This study suggests that CIP may promote aggressive growth properties in BRAFi-resistant melanomas, at a concentration present in serum.
Collapse
Affiliation(s)
- Seyyede Araste Aldaghi
- Department of Chemistry, Faculty of Science , Ferdowsi University of Mashhad , Mashhad , Iran
| | - Razieh Jalal
- Department of Chemistry, Faculty of Science , Ferdowsi University of Mashhad , Mashhad , Iran.,Department of Research Cell and Molecular Biology, Institute of Biotechnology , Ferdowsi University of Mashhad , Mashhad , Iran
| |
Collapse
|
148
|
Colin M, Delporte C, Janky R, Lechon AS, Renard G, Van Antwerpen P, Maltese WA, Mathieu V. Dysregulation of Macropinocytosis Processes in Glioblastomas May Be Exploited to Increase Intracellular Anti-Cancer Drug Levels: The Example of Temozolomide. Cancers (Basel) 2019; 11:cancers11030411. [PMID: 30909495 PMCID: PMC6468498 DOI: 10.3390/cancers11030411] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 12/16/2022] Open
Abstract
Macropinocytosis is a clathrin-independent endocytosis of extracellular fluid that may contribute to cancer aggressiveness through nutrient supply, recycling of plasma membrane and receptors, and exosome internalization. Macropinocytosis may be notably triggered by epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR), two well-known markers for glioblastoma aggressiveness. Therefore, we studied whether the expression of key actors of macropinocytosis is modified in human glioma datasets. Strong deregulation has been evidenced at the mRNA level according to the grade of the tumor, and 38 macropinocytosis-related gene signatures allowed discrimination of the glioblastoma (GBM) samples. Honokiol-induced vacuolization was then compared to vacquinol-1 and MOMIPP, two known macropinocytosis inducers. Despite high phase-contrast morphological similarities, honokiol-induced vacuoles appeared to originate from both endocytosis and ER. Also, acridine orange staining suggested differences in the macropinosomes’ fate: their fusion with lysosomes appeared very limited in 3-(5-methoxy -2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (MOMIPP)-treated cells. Nevertheless, each of the compounds markedly increased temozolomide uptake by glioma cells, as evidenced by LC-MS. In conclusion, the observed deregulation of macropinocytosis in GBM makes them prone to respond to various compounds affecting their formation and/or intracellular fate. Considering that sustained macropinocytosis may also trigger cell death of both sensitive and resistant GBM cells, we propose to envisage macropinocytosis inducers in combination approaches to obtain dual benefits: increased drug uptake and additive/synergistic effects.
Collapse
Affiliation(s)
- Margaux Colin
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - Cédric Delporte
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | | | - Anne-Sophie Lechon
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - Gwendoline Renard
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - Pierre Van Antwerpen
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - William A Maltese
- Department of Cancer Biology, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
- ULB Cancer Research Center, Université libre de Bruxelles (ULB), 1050 Bruxelles, Belgium.
| |
Collapse
|
149
|
Sharma G, Guardia CM, Roy A, Vassilev A, Saric A, Griner LN, Marugan J, Ferrer M, Bonifacino JS, DePamphilis ML. A family of PIKFYVE inhibitors with therapeutic potential against autophagy-dependent cancer cells disrupt multiple events in lysosome homeostasis. Autophagy 2019; 15:1694-1718. [PMID: 30806145 DOI: 10.1080/15548627.2019.1586257] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High-throughput screening identified 5 chemical analogs (termed the WX8-family) that disrupted 3 events in lysosome homeostasis: (1) lysosome fission via tubulation without preventing homotypic lysosome fusion; (2) trafficking of molecules into lysosomes without altering lysosomal acidity, and (3) heterotypic fusion between lysosomes and autophagosomes. Remarkably, these compounds did not prevent homotypic fusion between lysosomes, despite the fact that homotypic fusion required some of the same machinery essential for heterotypic fusion. These effects varied 400-fold among WX8-family members, were time and concentration dependent, reversible, and resulted primarily from their ability to bind specifically to the PIKFYVE phosphoinositide kinase. The ability of the WX8-family to prevent lysosomes from participating in macroautophagy/autophagy suggested they have therapeutic potential in treating autophagy-dependent diseases. In fact, the most potent family member (WX8) was 100-times more lethal to 'autophagy-addicted' melanoma A375 cells than the lysosomal inhibitors hydroxychloroquine and chloroquine. In contrast, cells that were insensitive to hydroxychloroquine and chloroquine were also insensitive to WX8. Therefore, the WX8-family of PIKFYVE inhibitors provides a basis for developing drugs that could selectively kill autophagy-dependent cancer cells, as well as increasing the effectiveness of established anti-cancer therapies through combinatorial treatments. Abbreviations: ACTB: actin beta; Baf: bafilomycin A1; BECN1: beclin 1; BODIPY: boron-dipyrromethene; BORC: BLOC-1 related complex; BRAF: B-Raf proto-oncogene, serine/threonine kinase; BSA: bovine serum albumin; CTSD: cathepsin D; CQ: chloroquine; DNA: deoxyribonucleic acid; EC50: half maximal effective concentration; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HCQ: hydroxychloroquine; HOPS complex: homotypic fusion and protein sorting complex; Kd: equilibrium binding constant; IC50: half maximal inhibitory concentration; KO: knockout; LAMP1: lysosomal associated membrane protein 1; MAP1LC3A: microtubule associated protein 1 light chain 3 alpha; MES: 2-(N-morpholino)ethanesulphonic acid; MTOR: mechanistic target of rapamycin kinase; μM: micromolar; NDF: 3-methylbenzaldehyde (2,6-dimorpholin-4-ylpyrimidin-4-yl)hydrazine;NEM: N-ethylmaleimide; NSF: N-ethylmaleimide sensitive factor; PBS: phosphate-buffered saline; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PIP4K2C: phosphatidylinositol-5-phosphate 4-kinase type 2 gamma; PtdIns3P: phosphatidylinositol 3-phosphate; PtdIns(3,5)P2: phosphatidylinositol 3,5-biphosphate; RFP: red fluorescent protein; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; SQSTM1: sequestosome 1; TWEEN 20: polysorbate 20; V-ATPase: vacuolar-type H+-translocating ATPase; VPS39: VPS39 subunit of HOPS complex; VPS41: VPS41 subunit of HOPS complex; WWL: benzaldehyde [2,6-di(4-morpholinyl)-4-pyrimidinyl]hydrazone; WX8: 1H-indole-3-carbaldehyde [4-anilino-6-(4-morpholinyl)-1,3,5-triazin-2-yl]hydrazine; XBA: N-(3-chloro-4-fluorophenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride; XB6: N-(4-ethylphenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride.
Collapse
Affiliation(s)
- Gaurav Sharma
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Ajit Roy
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Alex Vassilev
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Amra Saric
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Lori N Griner
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Juan Marugan
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Marc Ferrer
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Melvin L DePamphilis
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
150
|
Despres HW, Sabra A, Anderson P, Hemraz UD, Boluk Y, Sunasee R, Ckless K. Mechanisms of the immune response cause by cationic and anionic surface functionalized cellulose nanocrystals using cell-based assays. Toxicol In Vitro 2019; 55:124-133. [DOI: 10.1016/j.tiv.2018.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
|