1
|
Łukowska-Chojnacka E, Fedorov E, Kowalkowska A, Wielechowska M, Sobiepanek A, Koronkiewicz M, Wińska P. Synthesis and evaluation of anticancer activity of new 4,5,6,7-tetrabromo-1H-benzimidazole derivatives. Bioorg Chem 2024; 153:107880. [PMID: 39476601 DOI: 10.1016/j.bioorg.2024.107880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/16/2024] [Accepted: 10/05/2024] [Indexed: 12/10/2024]
Abstract
An efficient method for the synthesis of new 4,5,6,7-tetrabromo-1H-benzimidazole derivatives has been developed. New ketones were obtained by N-alkylation of TBBi or 2-Me-TBBi with various phenacyl halides and then reduced to the corresponding alcohols. All compounds were obtained with satisfactory yields in the range of 40-91 %. The synthesized compounds appeared a weak CK2 and PIM-1 inhibitors but exhibit an interesting cytotoxic activity against cancer cell lines, i.e. MCF-7, PC-3, CCRF-CEM, K-562. 1-Phenyl-2-(4,5,6,7-tetrabromo-1H-benzimidazol-1-yl)ethanone 3aA exhibits the highest cytotoxic activity with IC50 value of 5.30 µM for MCF-7 and 6.80 µM for CCRF-CEM. Moreover, this compound shows the highest selectivity against both MCF-7 and CCRF-CEM with SI selectivity coefficients (against MRC-5 and Vero cells) equal 5.45 and 4.30 for MCF-7 and 4.25 and 3.35 for CCRF-CEM, respectively. Furthermore, it was shown that compound 3aA exhibits very good pro-apoptotic properties, through induction of the mitochondrial apoptotic pathway in CCRF-CEM cells. These results correlate with data showing the effect of 3aA on intracellular level of CK2α protein and CK2-mediated phosphorylation of Ser529 in NF-κBp65. Study of the effect of compound 3aA on mRNA levels of CK2α and CK2α' showed no significant differences in gene expression levels in control CCRF-CEM and cells treated with 3aA, indicating 3aA action at the protein level.
Collapse
Affiliation(s)
- Edyta Łukowska-Chojnacka
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland.
| | - Egor Fedorov
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Anna Kowalkowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Monika Wielechowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Anna Sobiepanek
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Mirosława Koronkiewicz
- Department of Biomedical Research, National Medicines Institute, Chełmska St. 30/34, 00-725 Warsaw, Poland
| | - Patrycja Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| |
Collapse
|
2
|
Takatani-Nakase T, Ikushima C, Sakitani M, Nakase I. Regulatory network of ferroptosis and autophagy by targeting oxidative stress defense using sulfasalazine in triple-negative breast cancer. Life Sci 2024; 339:122411. [PMID: 38184272 DOI: 10.1016/j.lfs.2023.122411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/14/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
AIMS The cellular defense system against oxidative stress is important for the survival ability and sensitization in chemotherapy; however, the regulatory mechanisms remain unknown in triple-negative breast cancer (TNBC) cells. This study aimed to investigate the relationship between ferroptosis and autophagy by targeting the defense of oxidative stress through the cystine transporter (xCT) using sulfasalazine (SASP), which is a widely employed xCT inhibitor. MAIN METHODS We analyzed the cell death process of SASP in human TNBC cells, and examined the effects of SASP on tumor progression by using xenograft mouse model. KEY FINDINGS TNBC cells demonstrated a high defense capacity against reactive oxidative species through xCT. SASP significantly attenuated oxidative stress resistance in MDA-MB-231, which is a generally used model cell as TNBC, through decreased glutathione levels, causing a marked iron-dependent ferroptotic cell death induction. Moreover, autophagy was required to trigger efficient SASP-induced ferroptosis at the early stage of cell death. Tamoxifen, which is currently in clinical use as the gold standard for endocrine therapy of estrogen receptor-positive breast cancer, was a beneficial tool as an autophagy regulator under ferroptotic cell death by SASP. Additionally, SASP suppressed tumor growth and metastasis progression through total glutathione reduction in the primary tumor, indicating high anticancer activity against TNBC without liver injury in vivo. SIGNIFICANCE We revealed that SASP can efficiently induce ferroptosis associated with autophagy and that an understanding of the mechanism of cell death regulation by SASP is a promising new strategy for TNBC therapy and drug repositioning.
Collapse
Affiliation(s)
- Tomoka Takatani-Nakase
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo 663-8179, Japan; Institute for Bioscience, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo 663-8179, Japan.
| | - Chinami Ikushima
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo 663-8179, Japan
| | - Manami Sakitani
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, 11-68, Koshien Kyuban-cho, Nishinomiya, Hyogo 663-8179, Japan
| | - Ikuhiko Nakase
- Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| |
Collapse
|
3
|
Dehghan S, Kheshtchin N, Hassannezhad S, Soleimani M. Cell death classification: A new insight based on molecular mechanisms. Exp Cell Res 2023; 433:113860. [PMID: 38013091 DOI: 10.1016/j.yexcr.2023.113860] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Cells tend to disintegrate themselves or are forced to undergo such destructive processes in critical circumstances. This complex cellular function necessitates various mechanisms and molecular pathways in order to be executed. The very nature of cell death is essentially important and vital for maintaining homeostasis, thus any type of disturbing occurrence might lead to different sorts of diseases and dysfunctions. Cell death has various modalities and yet, every now and then, a new type of this elegant procedure gets to be discovered. The diversity of cell death compels the need for a universal organizing system in order to facilitate further studies, therapeutic strategies and the invention of new methods of research. Considering all that, we attempted to review most of the known cell death mechanisms and sort them all into one arranging system that operates under a simple but subtle decision-making (If \ Else) order as a sorting algorithm, in which it decides to place and sort an input data (a type of cell death) into its proper set, then a subset and finally a group of cell death. By proposing this algorithm, the authors hope it may solve the problems regarding newer and/or undiscovered types of cell death and facilitate research and therapeutic applications of cell death.
Collapse
Affiliation(s)
- Sepehr Dehghan
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Nasim Kheshtchin
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Soleimani
- Department of Medical Basic Sciences, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Pathogenesis of Hepatocellular Carcinoma: The Interplay of Apoptosis and Autophagy. Biomedicines 2023; 11:1166. [PMID: 37189787 PMCID: PMC10135776 DOI: 10.3390/biomedicines11041166] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The pathogenesis of hepatocellular carcinoma (HCC) is a multifactorial process that has not yet been fully investigated. Autophagy and apoptosis are two important cellular pathways that are critical for cell survival or death. The balance between apoptosis and autophagy regulates liver cell turnover and maintains intracellular homeostasis. However, the balance is often dysregulated in many cancers, including HCC. Autophagy and apoptosis pathways may be either independent or parallel or one may influence the other. Autophagy may either inhibit or promote apoptosis, thus regulating the fate of the liver cancer cells. In this review, a concise overview of the pathogenesis of HCC is presented, with emphasis on new developments, including the role of endoplasmic reticulum stress, the implication of microRNAs and the role of gut microbiota. The characteristics of HCC associated with a specific liver disease are also described and a brief description of autophagy and apoptosis is provided. The role of autophagy and apoptosis in the initiation, progress and metastatic potential is reviewed and the experimental evidence indicating an interplay between the two is extensively analyzed. The role of ferroptosis, a recently described specific pathway of regulated cell death, is presented. Finally, the potential therapeutic implications of autophagy and apoptosis in drug resistance are examined.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete School of Medicine, 71500 Heraklion, Crete, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Central Macedonia, Greece
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Central Macedonia, Greece
| |
Collapse
|
5
|
Effects of autophagy inhibition by 3-methyladenine on encystation, morphology, and metabolites of Cryptocaryon irritans. Parasitol Res 2023; 122:509-517. [PMID: 36526927 DOI: 10.1007/s00436-022-07751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Encystment is crucial for defense and reproduction in Cryptocaryon irritans. Therefore, understanding the encystment-related events in the protomont stage can help prevent and control C. irritans. Autophagy promotes protozoan parasite encystation. However, 3MA can inhibit autophagy. In this study, the effects of autophagy inhibition on encystation, survival rate, ultrastructural features, and metabolomic profiles of C. irritans, were evaluated using protomonts treated with 3MA (20 mM). The treatment with 3MA for about 4 h significantly lowered survival and encystation rates of protomonts to about 86.44% and 76.08%, respectively. Microstructural observations showed that the 3MA-treated protomonts showed deformed cell membranes and the cytoplasmic content spill. Furthermore, observation of the ultrastructure of 3MA-treated protomonts showed the destruction of organelles (Golgi bodies and mucocyst) and a lack of autophagosomes. However, no abnormality was observed in the control experiments. Furthermore, the metabolic analysis revealed suppression of metabolites, such as lipids, amino acids, and carbohydrates. These results demonstrate that 3MA can inhibit autophagy in C. irritans, thus hindering encystation, suppressing the metabolism of metabolites, and altering morphological ultrastructure in these parasites.
Collapse
|
6
|
Plini ERG, Melnychuk MC, Harkin A, Dahl MJ, McAuslan M, Kühn S, Boyle RT, Whelan R, Andrews R, Düzel S, Drewelies J, Wagner GG, Lindenberger U, Norman K, Robertson IH, Dockree PM. Dietary Tyrosine Intake (FFQ) Is Associated with Locus Coeruleus, Attention and Grey Matter Maintenance: An MRI Structural Study on 398 Healthy Individuals of the Berlin Aging Study-II. J Nutr Health Aging 2023; 27:1174-1187. [PMID: 38151868 DOI: 10.1007/s12603-023-2005-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/19/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND AND OBJECTIVE It is documented that low protein and amino-acid dietary intake is related to poorer cognitive health and increased risk of dementia. Degradation of the neuromodulatory pathways, (comprising the cholinergic, dopaminergic, serotoninergic and noradrenergic systems) is observed in neurodegenerative diseases and impairs the proper biosynthesis of key neuromodulators from micro-nutrients and amino acids. How these micro-nutrients are linked to neuromodulatory pathways in healthy adults is less studied. The Locus Coeruleus-Noradrenergic System (LC-NA) is the earliest subcortical structure affected in Alzheimer's disease, showing marked neurodegeneration, but is also sensitive for age-related changes. The LC-NA system is critical for supporting attention and cognitive control, functions that are enhanced both by tyrosine administration and chronic tyrosine intake. The purpose of this study was to 1) investigate whether the dietary intake of tyrosine, the key precursor for noradrenaline (NA), is related to LC signal intensity 2) whether LC mediates the reported association between tyrosine intake and higher cognitive performance (measured with Trail Making Test - TMT), and 3) whether LC signal intensity relates to an objective measure of brain maintenance (BrainPAD). METHODS The analyses included 398 3T MRIs of healthy participants from the Berlin Aging Study II to investigate the relationship between LC signal intensity and habitual dietary tyrosine intake-daily average (HD-Tyr-IDA - measured with Food Frequency Questionnaire - FFQ). As a control procedure, the same analyses were repeated on other main seeds of the neuromodulators' subcortical system (Dorsal and Medial Raphe, Ventral Tegmental Area and Nucleus Basalis of Meynert). In the same way, the relationships between the five nuclei and BrainPAD were tested. RESULTS Results show that HD-Tyr-IDA is positively associated with LC signal intensity. Similarly, LC disproportionally relates to better brain maintenance (BrainPAD). Mediation analyses reveal that only LC, relative to the other nuclei tested, mediates the relationship between HD-Tyr-IDA I and performance in the TMT and between HD-Tyr-IDA and BrainPAD. CONCLUSIONS These findings provide the first evidence linking tyrosine intake with LC-NA system signal intensity and its correlation with neuropsychological performance. This study strengthens the role of diet for maintaining brain and cognitive health and supports the noradrenergic theory of cognitive reserve. Within this framework, adequate tyrosine intake might increase the resilience of LC-NA system functioning, by preventing degeneration and supporting noradrenergic metabolism required for LC function and neuropsychological performance.
Collapse
Affiliation(s)
- E R G Plini
- Emanuele RG Plini, Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Sehgal SA, Wu H, Sajid M, Sohail S, Ahsan M, Parveen G, Riaz M, Khan MS, Iqbal MN, Malik A. Pharmacological Progress of Mitophagy Regulation. Curr Neuropharmacol 2023; 21:1026-1041. [PMID: 36918785 PMCID: PMC10286582 DOI: 10.2174/1570159x21666230314140528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 03/16/2023] Open
Abstract
With the advancement in novel drug discovery, biologically active compounds are considered pharmacological tools to understand complex biological mechanisms and the identification of potent therapeutic agents. Mitochondria boast a central role in different integral biological processes and mitochondrial dysfunction is associated with multiple pathologies. It is, therefore, prudent to target mitochondrial quality control mechanisms by using pharmacological approaches. However, there is a scarcity of biologically active molecules, which can interact with mitochondria directly. Currently, the chemical compounds used to induce mitophagy include oligomycin and antimycin A for impaired respiration and acute dissipation of mitochondrial membrane potential by using CCCP/FCCP, the mitochondrial uncouplers. These chemical probes alter the homeostasis of the mitochondria and limit our understanding of the energy regulatory mechanisms. Efforts are underway to find molecules that can bring about selective removal of defective mitochondria without compromising normal mitochondrial respiration. In this report, we have tried to summarize and status of the recently reported modulators of mitophagy.
Collapse
Affiliation(s)
- Sheikh Arslan Sehgal
- Department of Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Punjab, Pakistan
- Department of Bioinformatics, University of Okara, Okara, Pakistan
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, China
| | - Muhammad Sajid
- Department of Biotechnology, University of Okara, Okara, Pakistan
| | - Summar Sohail
- Department of Forestry, Kohsar University Murree, Pakistan
| | - Muhammad Ahsan
- Institute of Environmental and Agricultural Sciences, University of Okara, Okara, Punjab, Pakistan
| | | | - Mehreen Riaz
- Department of Zoology, Women University, Swabi, Pakistan
| | | | - Muhammad Nasir Iqbal
- Department of Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Punjab, Pakistan
| | - Abbeha Malik
- Department of Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Punjab, Pakistan
| |
Collapse
|
8
|
ZC3H4 regulates infiltrating monocytes, attenuating pulmonary fibrosis through IL-10. Respir Res 2022; 23:204. [PMID: 35962397 PMCID: PMC9375388 DOI: 10.1186/s12931-022-02134-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Silicosis is a pulmonary fibrosis-associated disease caused by the inhalation of large amounts of free silicon dioxide (SiO2) that mainly manifests as early inflammation and late pulmonary fibrosis. As macrophage precursors, monocytes accumulate in the lung during early inflammation, but their role in the development of silicosis is unclear. Single-cell sequencing (cell numbers = 25,002), Western blotting, quantitative real-time PCR, ELISA and cell functional experiments were used to explore the specific effects of monocytes on fibroblasts. The CRISPR/Cas9 system was used to specifically knock down ZC3H4, a novel member of the CCCH zinc finger protein family, and was combined with pharmacological methods to explore the mechanism by which ZC3H4 affects chemokine and cytokine secretion. The results indicated that (1) SiO2 induced an infiltrating phenotype in monocytes; (2) infiltrating monocytes inhibited the activation, viability and migration of fibroblasts by regulating IL-10 but not IL-8; and (3) SiO2 downregulated IL-10 via ZC3H4-induced autophagy. This study revealed that ZC3H4 regulated the secretion function of monocytes, which, in turn, inhibited fibroblast function in early inflammation through autophagy signaling, thereby reducing pulmonary fibrosis. These findings provide a new idea for the clinical treatment of silicosis.
Collapse
|
9
|
Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther 2022; 7:218. [PMID: 35798726 PMCID: PMC9261907 DOI: 10.1038/s41392-022-01080-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Molecular oxygen (O2) is essential for most biological reactions in mammalian cells. When the intracellular oxygen content decreases, it is called hypoxia. The process of hypoxia is linked to several biological processes, including pathogenic microbe infection, metabolic adaptation, cancer, acute and chronic diseases, and other stress responses. The mechanism underlying cells respond to oxygen changes to mediate subsequent signal response is the central question during hypoxia. Hypoxia-inducible factors (HIFs) sense hypoxia to regulate the expressions of a series of downstream genes expression, which participate in multiple processes including cell metabolism, cell growth/death, cell proliferation, glycolysis, immune response, microbe infection, tumorigenesis, and metastasis. Importantly, hypoxia signaling also interacts with other cellular pathways, such as phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-B (NF-κB) pathway, extracellular signal-regulated kinases (ERK) signaling, and endoplasmic reticulum (ER) stress. This paper systematically reviews the mechanisms of hypoxia signaling activation, the control of HIF signaling, and the function of HIF signaling in human health and diseases. In addition, the therapeutic targets involved in HIF signaling to balance health and diseases are summarized and highlighted, which would provide novel strategies for the design and development of therapeutic drugs.
Collapse
|
10
|
Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components. Cancers (Basel) 2022; 14:cancers14061462. [PMID: 35326612 PMCID: PMC8945922 DOI: 10.3390/cancers14061462] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chemotherapy resistance is a common occurrence during cancer treatment that cancer researchers are attempting to understand and overcome. Mitochondria are a crucial intracellular signaling core that are becoming important determinants of numerous aspects of cancer genesis and progression, such as metabolic reprogramming, metastatic capability, and chemotherapeutic resistance. Mitophagy, or selective autophagy of mitochondria, can influence both the efficacy of tumor chemotherapy and the degree of drug resistance. Regardless of the fact that mitochondria are well-known for coordinating ATP synthesis from cellular respiration in cellular bioenergetics, little is known its mitophagy regulation in chemoresistance. Recent advancements in mitochondrial research, mitophagy regulatory mechanisms, and their implications for our understanding of chemotherapy resistance are discussed in this review. Abstract Cancer chemotherapy resistance is one of the most critical obstacles in cancer therapy. One of the well-known mechanisms of chemotherapy resistance is the change in the mitochondrial death pathways which occur when cells are under stressful situations, such as chemotherapy. Mitophagy, or mitochondrial selective autophagy, is critical for cell quality control because it can efficiently break down, remove, and recycle defective or damaged mitochondria. As cancer cells use mitophagy to rapidly sweep away damaged mitochondria in order to mediate their own drug resistance, it influences the efficacy of tumor chemotherapy as well as the degree of drug resistance. Yet despite the importance of mitochondria and mitophagy in chemotherapy resistance, little is known about the precise mechanisms involved. As a consequence, identifying potential therapeutic targets by analyzing the signal pathways that govern mitophagy has become a vital research goal. In this paper, we review recent advances in mitochondrial research, mitophagy control mechanisms, and their implications for our understanding of chemotherapy resistance.
Collapse
|
11
|
Sun Y, Wen F, Yan C, Su L, Luo J, Chi W, Zhang S. Mitophagy Protects the Retina Against Anti-Vascular Endothelial Growth Factor Therapy-Driven Hypoxia via Hypoxia-Inducible Factor-1α Signaling. Front Cell Dev Biol 2021; 9:727822. [PMID: 34790659 PMCID: PMC8591297 DOI: 10.3389/fcell.2021.727822] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
Anti-VEGF drugs are first-line treatments for retinal neovascular diseases, but these anti-angiogenic agents may also aggravate retinal damage by inducing hypoxia. Mitophagy can protect against hypoxia by maintaining mitochondrial quality, thereby sustaining metabolic homeostasis and reducing reactive oxygen species (ROS) generation. Here we report that the anti-VEGF agent bevacizumab upregulated the hypoxic cell marker HIF-1α in photoreceptors, Müller cells, and vascular endothelial cells of oxygen-induced retinopathy (OIR) model mice, as well as in hypoxic cultured 661W photoreceptors, MIO-MI Müller cells, and human vascular endothelial cells. Bevacizumab also increased expression of mitophagy-related proteins, and mitophagosome formation both in vivo and in vitro, but did not influence cellular ROS production or apoptosis rate. The HIF-1α inhibitor LW6 blocked mitophagy, augmented ROS production, and triggered apoptosis. Induction of HIF-1α and mitophagy were associated with upregulation of BCL2/adenovirus E1B 19-kDa protein-interacting protein 3 (BNIP3) and FUN14 domain containing 1 (FUNDC1), and overexpression of these proteins in culture reversed the effects of HIF-1α inhibition. These findings suggest that bevacizumab does induce retinal hypoxia, but that concomitant activation of the HIF-1α-BNIP3/FUNDC1 signaling pathway also induces mitophagy, which can mitigate the deleterious effects by reducing oxidative stress secondary. Promoting HIF-1α-BNIP3/FUNDC1-mediated mitophagy may enhance the safety of anti-VEGF therapy for retinal neovascular diseases and indicate new explanation and possible new target of the anti-VEGF therapy with suboptimal effect.
Collapse
Affiliation(s)
- Yimeng Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chun Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lishi Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiawen Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shaochong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Shenzhen Key Laboratory of Ophthalmology, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
| |
Collapse
|
12
|
Chojnacki K, Wińska P, Karatsai O, Koronkiewicz M, Milner-Krawczyk M, Wielechowska M, Rędowicz MJ, Bretner M, Borowiecki P. Synthesis of Novel Acyl Derivatives of 3-(4,5,6,7-Tetrabromo-1 H-benzimidazol-1-yl)propan-1-ols-Intracellular TBBi-Based CK2 Inhibitors with Proapoptotic Properties. Int J Mol Sci 2021; 22:6261. [PMID: 34200807 PMCID: PMC8230474 DOI: 10.3390/ijms22126261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/09/2022] Open
Abstract
Protein kinase CK2 has been considered as an attractive drug target for anti-cancer therapy. The synthesis of N-hydroxypropyl TBBi and 2MeTBBi derivatives as well as their respective esters was carried out by using chemoenzymatic methods. Concomitantly with kinetic studies toward recombinant CK2, the influence of the obtained compounds on the viability of two human breast carcinoma cell lines (MCF-7 and MDA-MB-231) was evaluated using MTT assay. Additionally, an intracellular inhibition of CK2 as well as an induction of apoptosis in the examined cells after the treatment with the most active compounds were studied by Western blot analysis, phase-contrast microscopy and flow cytometry method. The results of the MTT test revealed potent cytotoxic activities for most of the newly synthesized compounds (EC50 4.90 to 32.77 µM), corresponding to their solubility in biological media. We concluded that derivatives with the methyl group decrease the viability of both cell lines more efficiently than their non-methylated analogs. Furthermore, inhibition of CK2 in breast cancer cells treated with the tested compounds at the concentrations equal to their EC50 values correlates well with their lipophilicity since derivatives with higher values of logP are more potent intracellular inhibitors of CK2 with better proapoptotic properties than their parental hydroxyl compounds.
Collapse
Affiliation(s)
- Konrad Chojnacki
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Patrycja Wińska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (O.K.); (M.J.R.)
| | - Mirosława Koronkiewicz
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Małgorzata Milner-Krawczyk
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Monika Wielechowska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (O.K.); (M.J.R.)
| | - Maria Bretner
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Paweł Borowiecki
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| |
Collapse
|
13
|
Behl T, Sehgal A, Bala R, Chadha S. Understanding the molecular mechanisms and role of autophagy in obesity. Mol Biol Rep 2021; 48:2881-2895. [PMID: 33797660 DOI: 10.1007/s11033-021-06298-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/17/2021] [Indexed: 12/14/2022]
Abstract
Vital for growth, proliferation, subsistence, and thermogenesis, autophagy is the biological cascade, which confers defence against aging and various pathologies. Current research has demonstrated de novo activity of autophagy in stimulation of biological events. There exists a significant association between autophagy activation and obesity, encompassing expansion of adipocytes which facilitates β cell activity. The main objective of the manuscript is to enumerate intrinsic role of autophagy in obesity and associated complications. The peer review articles published till date were searched using medical databases like PubMed and MEDLINE for research, primarily in English language. Obesity is characterized by adipocytic hypertrophy and hyperplasia, which leads to imbalance of lipid absorption, free fatty acid release, and mitochondrial activity. Detailed evaluation of obesity progression is necessary for its treatment and related comorbidities. Data collected in regard to etiological sustaining of obesity, has revealed hypothesized energy misbalance and neuro-humoral dysfunction, which is stimulated by autophagy. Autophagy regulates chief salvaging events for protein clustering, excessive triglycerides, and impaired mitochondria which is accompanied by oxidative and genotoxic stress in mammals. Autophagy is a homeostatic event, which regulates biological process by eliminating lethal cells and reprocessing physiological constituents, comprising of proteins and fat. Unquestionably, autophagy impairment is involved in metabolic syndromes, like obesity. According to an individual's metabolic outline, autophagy activation is essential for metabolism and activity of the adipose tissue and to retard metabolic syndrome i.e. obesity. The manuscript summarizes the perception of current knowledge on autophagy stimulation and its effect on the obesity.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rajni Bala
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
14
|
Wei YX, Dong SM, Wang YY, Zhang P, Sun MY, Wei YX, Meng XC, Wang Y. Autophagy participates in the protection role of 1,25-dihydroxyvitamin D3 in acute myocardial infarction via PI3K/AKT/mTOR pathway. Cell Biol Int 2020; 45:394-403. [PMID: 33146448 DOI: 10.1002/cbin.11495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/09/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022]
Abstract
Vitamin D deficiency is associated with acute myocardial infarction (AMI); thus we aimed to explore improvement effects of 1,25-dihydroxyvitamin D3 (VD3) on the AMI and its potential mechanism. AMI models were constructed using male C57/BL6J mice and randomly treated with normal saline or VD3, using sham rats as control. Heart functions, myocardial damage, apoptosis, and inflammation were evaluated. Cardiomyocytes isolated from 3-day-old suckling mice were used for in vitro verification. After VD3 treatment, AMI-induced cardiac dysfunction was reversed with better cardiac function parameters. VD3 treatment reduced inflammatory cell infiltration and myocardial infarction area accompanied by the reduction of inflammatory factors and myocardial infarction markers compared with the AMI group. VD3 treatment obviously alleviated AMI-induced myocardial apoptosis, along with Bcl-2 upregulation and downregulation of caspase-3, caspase-9, and Bax. Both in vivo and in vitro experiments revealed that VD3 enhanced the expression of LC3II and Beclin-1 and decreased soluble p62. Furthermore, VD3 enhanced the AMI-caused inhibition of PI3K, p-AKT, and p-mTOR expression, which was conversely reversed by the addition of 3-methyladenine in vitro. The study highlights the improvement effects of VD3 on cardiac functions. We proposed a potential mechanism that VD3 protects against myocardial damage, inflammation, and apoptosis by promoting autophagy through PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yun-Xia Wei
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.,Department of Cardiovascular, Shijiazhuang HuaYao Hospital, Shijiazhuang, Hebei, China
| | - Shi-Min Dong
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuan-Yuan Wang
- Department of Respiratory, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pu Zhang
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ming-Yu Sun
- Department of Cardiovascular, Shijiazhuang HuaYao Hospital, Shijiazhuang, Hebei, China
| | - Yun-Xiao Wei
- Department of Neurology, Shijiazhuang HuaYao Hospital, Shijiazhuang, Hebei, China
| | - Xian-Ce Meng
- Department of Neurology, Shijiazhuang HuaYao Hospital, Shijiazhuang, Hebei, China
| | - Yue Wang
- Department of Respiratory, Shijiazhuang HuaYao Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
15
|
Maruszewska A, Tarasiuk J. Quercetin Triggers Induction of Apoptotic and Lysosomal Death of Sensitive and Multidrug Resistant Leukaemia HL60 Cells. Nutr Cancer 2020; 73:484-501. [PMID: 32329631 DOI: 10.1080/01635581.2020.1752745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multidrug resistance (MDR) constitutes the major cause of the failure in anticancer therapy. One of the most important mechanisms leading to the occurrence of MDR is related to the modulation of cellular death pathways. The aim of this study was to determine the effect of quercetin (Q) on triggering the programed death of human promyelocytic leukemia sensitive cells HL60 as well as multidrug resistant HL60/VINC cells overexpressing P-glycoprotein and HL60/MX2 cells characterized by the presence of mutated α isoform of topoisomerase II and the absence of β isoform of this enzyme. Q exerted comparable cytotoxic activities toward sensitive HL60 cells and their MDR counterparts. It was also found that this compound modulated the cellular level of reactive oxygen species (ROS) and led to the marked decrease in cellular GSH level. Furthermore, it was demonstrated that Q used at IC50 and IC90 significantly increased the percentage of sub-G1 subpopulation of all studied leukemia cells causing oligonucleosomal DNA fragmentation. The present study also indicated that Q used at IC90 triggers predominantly programed cell death of sensitive HL60 cells and their MDR counterparts by induction of apoptosis occurring with the involvement of caspase-3 and caspase-8 as well as by lysosome membrane permeabilization-dependent mechanisms.
Collapse
Affiliation(s)
- Agnieszka Maruszewska
- Department of Biochemistry, Faculty of Biology, University of Szczecin, Szczecin, Poland.,Molecular Biology and Biotechnology Center, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | - Jolanta Tarasiuk
- Department of Biochemistry, Faculty of Biology, University of Szczecin, Szczecin, Poland.,Molecular Biology and Biotechnology Center, Faculty of Biology, University of Szczecin, Szczecin, Poland
| |
Collapse
|
16
|
Gnatyshyna L, Khoma V, Mishchuk O, Martinyuk V, Spriņģe G, Stoliar O. Multi-marker study of the responses of the Unio tumidus from the areas of small and micro hydropower plants at the Dniester River Basin, Ukraine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11038-11049. [PMID: 31955329 DOI: 10.1007/s11356-020-07698-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
The impact of the hydropower plants (HPPs) on the aquatic life is expected, but the biochemical markers of ecotoxicity have not been investigated in relation to HPP proximity. The aim of this study was to compare the responses of mollusk Unio tumidus from the small HPP (reservoir (Ku) and downstream of the dam (Kd)) and micro HPP (upstream (Zu) and downstream of the dam (Zd)). In total, 11 indexes (n) from digestive gland, hemocytes (lysosomal integrity), and gonads (alkali-labile phosphates, ALP) were analyzed. The mollusks from the reservoir demonstrate the typical signs of toxic impact: cholinesterase and glutathione depletion, the highest glutathione S-transferase activity, and ratio of extralysosomal cathepsin D compare to all other groups. The specimens from the micro HPP have the highest levels of glutathione (Zd) and lipid peroxidation (Zu) and lesser Cu/Zn-SOD activity (Zu) than other groups. These indications of stressful conditions may derive from the regular oscillations in the water flow regimes at the micro HPP. For both HPPs, the responses of upstream and downstream groups are distinct. The calculated IBR/n (4.17, 3.85, 3.12, and 0.26 for Ku, Kd, Zu, and Zd correspondingly) gives a quantitative basis for the evaluation of environmental impact of HPPs. Graphical abstract .
Collapse
Affiliation(s)
- Lesya Gnatyshyna
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine
- I.Ya. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Vira Khoma
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine
| | | | - Viktoria Martinyuk
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine
| | - Gunta Spriņģe
- University of Latvia, Institute of Biology, Salaspils, Latvia
| | - Oksana Stoliar
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Kryvonosa Str 2, Ternopil, 46027, Ukraine.
| |
Collapse
|
17
|
Nirmala JG, Lopus M. Cell death mechanisms in eukaryotes. Cell Biol Toxicol 2019; 36:145-164. [PMID: 31820165 DOI: 10.1007/s10565-019-09496-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
Like the organism they constitute, the cells also die in different ways. The death can be predetermined, programmed, and cleanly executed, as in the case of apoptosis, or it can be traumatic, inflammatory, and sudden as many types of necrosis exemplify. Nevertheless, there are a number of cell deaths-some of them bearing a resemblance to apoptosis and/or necrosis, and many, distinct from each-that serve a multitude of roles in either supporting or disrupting the homoeostasis. Apoptosis is coordinated by death ligands, caspases, b-cell lymphoma-2 (Bcl-2) family proteins, and their downstream effectors. Events that can lead to apoptosis include mitotic catastrophe and anoikis. Necrosis, although it has been considered an abrupt and uncoordinated cell death, has many molecular events associated with it. There are cell death mechanisms that share some standard features with necrosis. These include methuosis, necroptosis, NETosis, pyronecrosis, and pyroptosis. Autophagy, generally a catabolic pathway that operates to ensure cell survival, can also kill the cell through mechanisms such as autosis. Other cell-death mechanisms include entosis, ferroptosis, lysosome-dependent cell death, and parthanatos.
Collapse
Affiliation(s)
- J Grace Nirmala
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, 400098, India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, 400098, India.
| |
Collapse
|
18
|
Falfushynska HI, Wu F, Ye F, Kasianchuk N, Dutta J, Dobretsov S, Sokolova IM. The effects of ZnO nanostructures of different morphology on bioenergetics and stress response biomarkers of the blue mussels Mytilus edulis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 694:133717. [PMID: 31400676 DOI: 10.1016/j.scitotenv.2019.133717] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Biofouling causes massive economical losses in the maritime sector creating an urgent need for effective and ecologically non-harmful antifouling materials. Zinc oxide (ZnO) nanorod coatings show promise as an antifouling material; however, the toxicity of ZnO nanorods to marine organisms is not known. We compared the toxicity of suspended ZnO nanorods (NR) with that of ZnO nanoparticles (NP) and ionic Zn2+ in a marine bivalve Mytilus edulis exposed for two weeks to 10 or 100 μg Zn L-1 of ZnO NPs, NRs or Zn2+, or to immobilized NRs. The multi-biomarker assessment included bioenergetics markers (tissue energy reserves, activity of mitochondrial electron transport system and autophagic enzymes), expression of apoptotic and inflammatory genes, and general stress biomarkers (oxidative lesions, lysosomal membrane stability and metallothionein expression). Exposure to ZnO NPs, NRs and Zn2+ caused accumulation of oxidative lesions in proteins and lipids, stimulated autophagy, and led to lysosomal membrane destabilization indicating toxicity. However, these responses were not specific for the form of Zn (NPs, NR or Zn2+) and showed no monotonous increase with increasing Zn concentrations in the experimental exposures. No major disturbance of the energy status was found in the mussels exposed to ZnO NPs, NRs, or Zn2+. Exposure to ZnO NPs and NRs led to a strong induction of apoptosis- and inflammation-related genes, which was not seen in Zn2+ exposures. Based on the integrated biomarker response, the overall toxicity as well as the pro-apoptotic and pro-inflammatory action was stronger in ZnO NPs compared with the NRs. Given the stability of ZnO NR coatings and the relatively low toxicity of suspended ZnO NR, ZnO NR coating might be considered a promising low-toxicity material for antifouling paints.
Collapse
Affiliation(s)
- Halina I Falfushynska
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Department of Human Health, Physical Rehabilitation and Vital Activity, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Fangli Wu
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany
| | - Fei Ye
- KTH Royal Institute of Technology, Material and Nanophysics Applied Physics Department, School of Science, Stockholm, Sweden
| | - Nadiia Kasianchuk
- Department of Human Health, Physical Rehabilitation and Vital Activity, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Joydeep Dutta
- KTH Royal Institute of Technology, Material and Nanophysics Applied Physics Department, School of Science, Stockholm, Sweden
| | - Sergey Dobretsov
- Department of Marine Science and Fisheries, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al Khoud 123, PO Box 34, Muscat, Oman; Center of Excellence in Marine Biotechnology, Sultan Qaboos University, Al Khoud 123, PO Box 50, Muscat, Oman
| | - Inna M Sokolova
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, Rostock, Germany.
| |
Collapse
|
19
|
Wang X, Sun D, Hu Y, Xu X, Jiang W, Shang H, Cui D. The roles of oxidative stress and Beclin-1 in the autophagosome clearance impairment triggered by cardiac arrest. Free Radic Biol Med 2019; 136:87-95. [PMID: 30951836 DOI: 10.1016/j.freeradbiomed.2018.12.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 02/03/2023]
Abstract
During cardiac arrest and return of spontaneous circulation (CA-ROSC), autophagosome clearance in the cortex is progressively impaired, but the role of reactive oxygen species (ROS) in this process and the mechanism underlying the autophagy impairment remain unknown. In this study, we investigated the impacts of ROS on the autophagy-lysosome pathway after CA-ROSC in rats. Cortices from CA-ROSC rats revealed accumulation of LC3, p62 and ubiquitin, indicating impaired autophagic flux. Furthermore, impairment of autophagic flux was related to lysosomal lesion, as indicated by decreased cathepsin D and lysosomal-associated membrane protein 2 (LAMP2) levels after CA-ROSC. In vitro, the resulting ROS generation blocked autophagosome processing and caused accumulation of LC3-II, ubiquitin, and p62, leading to mitochondrial dysfunction and cell death; this outcome was alleviated by cyclosporine A (CsA) pretreatment. Interestingly, ischemia/reperfusion injury was connected with ROS-mediated Beclin-1 upregulation and a reduction in LAMP2, which is a pivotal protein in the autophagy-lysosome pathway. Recovery of the LAMP2 levels and partial Beclin-1 silencing restored the autophagic flux and reduced cell death after CA-ROSC. Taken together, our data indicate that CA-ROSC injury impairs autophagosome clearance partially through a ROS-induced decline in LAMP2 and increase in Beclin-1, leading to increased neuronal cell death.
Collapse
Affiliation(s)
- Xintao Wang
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, China
| | - Dawei Sun
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, China
| | - Yue Hu
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, China
| | - Xiaotao Xu
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, China
| | - Wei Jiang
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, China
| | - Hanbing Shang
- Department of Neurosurgery, Shanghai Ruijin Hospital Affiliated with Medical School of Shanghai Jiaotong University, China.
| | - Derong Cui
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, China.
| |
Collapse
|
20
|
Falfushynska HI, Gnatyshyna LL, Ivanina AV, Khoma VV, Stoliar OB, Sokolova IM. Bioenergetic responses of freshwater mussels Unio tumidus to the combined effects of nano-ZnO and temperature regime. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 650:1440-1450. [PMID: 30308831 DOI: 10.1016/j.scitotenv.2018.09.136] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/01/2018] [Accepted: 09/10/2018] [Indexed: 06/08/2023]
Abstract
Bivalves from the cooling reservoirs of electrical power plants (PP) are exposed to the chronic heating and chemical pollution making them a suitable model to study the combined effects of these stressors. We investigated the effect of in situ exposures to chemical and thermal pollution in the PP cooling ponds on the metabolic responses of unionid bivalves (Unio tumidus) to a novel widespread pollutant, ZnO nanoparticles (nZnO). Male U. tumidus from the reservoirs of Dobrotvir and Burshtyn PPs (DPP and BPP) were maintained in clean water at 18 °C, or exposed for 14 days to one of the following conditions: nZnO (3.1 μM) or Zn2+ (3.1 μM, a positive control for Zn impacts) at 18 °C, elevated temperature (T, 25 °C), or nZnO at 25 °C (nZnO + T). Baseline levels of glycogen, lipids and ATP were similar in the two studied populations, whereas the levels of proteins, lactate/pyruvate ratio (L/P) and extralysosomal cathepsin D level were higher in the tissues of BPP mussels. The levels of glycogen and glucose declined in most experimental exposures indicating elevated energy demand except for a slight increase in the digestive gland of warming-exposed BPP mussels and in the gills of the nZnO + T-exposed DPP-mussels. Experimental exposures stimulated cathepsin D activity likely reflecting onset of autophagic processes to compensate for stress-induced energy demand. No depletion of ATP in Zn-containing exposures was observed indicating that the cellular metabolic adjustments were sufficient for such compensation. Unexpectedly, experimental warming mitigated most metabolic responses to nZnO in co-exposures. Our data thus indicate that metabolic effects of nZnO strongly depend on the environmental context of the mussels (such as temperature and acclimation history) which must be taken into account for the molecular and cellular biomarker-based assessment of the nanoparticle effects in the field.
Collapse
Affiliation(s)
- Halina I Falfushynska
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Lesya L Gnatyshyna
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine; Department of General Chemistry, I.Ya. Horbachevsky Ternopil State Medical University, Ternopil, Ukraine
| | - Anna V Ivanina
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, USA
| | - Vira V Khoma
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Oksana B Stoliar
- Research Laboratory of Comparative Biochemistry and Molecular Biology, Ternopil V. Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Inna M Sokolova
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, Rostock, Germany; Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, USA.
| |
Collapse
|
21
|
Porcu C, Sideri S, Martini M, Cocomazzi A, Galli A, Tarantino G, Balsano C. Oleuropein Induces AMPK-Dependent Autophagy in NAFLD Mice, Regardless of the Gender. Int J Mol Sci 2018; 19:3948. [PMID: 30544824 PMCID: PMC6321282 DOI: 10.3390/ijms19123948] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Oleuropein (Ole) is one of the most plentiful phenolic compounds with antioxidant, anti-inflammatory, anti-atherogenic, hypoglycemic and hypolipidemic effects. The aim of our study was to establish whether the positive Ole-related effects on liver steatosis could be associated with autophagy. Female and male C57BL/6J mice were fed normal diet (ND) or high-fat diet (HFD) for eight weeks, and Ole was added or not for the following eight weeks. The autophagy-related proteins Akt, mTOR, AMPK, ULK1, Beclin-1, LC3B and p62/Sqstm1 were analyzed. Interestingly, Ole induced a different regulation of the Akt/mTOR pathway in female compared to male mice, but was able to activate the autophagic process in ND and HFD mice through AMPK-dependent phosphorylation of ULK1 at Ser555, regardless of the gender. Our work reveals the ability of Ole to induce, in liver of ND and HFD mice, autophagy independently by gender-specific mTOR activation. We highlight Ole as a novel therapeutic approach to counteract unhealthy diet-related liver steatosis by targeting autophagy.
Collapse
Affiliation(s)
- Cristiana Porcu
- F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy.
- MESVA Department, University of L'Aquila, Piazza S. Salvatore Tommasi 1, 67100 Coppito, L'Aquila, Italy and F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy.
| | - Silvia Sideri
- MESVA Department, University of L'Aquila, Piazza S. Salvatore Tommasi 1, 67100 Coppito, L'Aquila, Italy and F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy.
| | - Maurizio Martini
- Fondazione Policlinico A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore Rome, Italy.
| | - Alessandra Cocomazzi
- Fondazione Policlinico A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore Rome, Italy.
| | - Andrea Galli
- Gastroenterology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy.
| | - Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, 80131 Naples, Italy.
| | - Clara Balsano
- F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy.
- MESVA Department, University of L'Aquila, Piazza S. Salvatore Tommasi 1, 67100 Coppito, L'Aquila, Italy and F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy.
| |
Collapse
|
22
|
Fortenbery GW, Sarathy B, Carraway KR, Mansfield KD. Hypoxic stabilization of mRNA is HIF-independent but requires mtROS. Cell Mol Biol Lett 2018; 23:48. [PMID: 30305827 PMCID: PMC6172842 DOI: 10.1186/s11658-018-0112-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/25/2018] [Indexed: 01/09/2023] Open
Abstract
Background Tissue ischemia can arise in response to numerous physiologic and pathologic conditions. The cellular response to decreased perfusion, most notably a decrease in glucose and oxygen, is important for cellular survival. In response to oxygen deprivation or hypoxia, one of the key response elements is hypoxia inducible factor (HIF) and a key protein induced by hypoxia is vascular endothelial growth factor (VEGF). Under hypoxia, we and others have reported an increase in the half-life of VEGF and other hypoxia related mRNAs including MYC and CYR61; however, the mediator of this response has yet to be identified. For this study, we sought to determine if HIF-mediated transcriptional activity is involved in the mRNA stabilization induced by hypoxia. Methods HEK293T or C6 cells were cultured in either normoxic or hypoxic (1% oxygen) conditions in the presence of 1 g/L glucose for all experiments. Pharmacological treatments were used to mimic hypoxia (desferroxamine, dimethyloxaloglutamate, CoCl2), inhibit mitochondrial respiration (rotenone, myxothiazol), scavenge reactive oxygen species (ROS; ebselen), or generate mitochondrial ROS (antimycin A). siRNAs were used to knock down components of the HIF transcriptional apparatus. mRNA half-life was determined via actinomycin D decay and real time PCR and western blotting was used to determine mRNA and protein levels respectively. Results Treatment of HEK293T or C6 cells with hypoxic mimetics, desferroxamine, dimethyloxaloglutamate, or CoCl2 showed similar induction of HIF compared to hypoxia treatment, however, in contrast to hypoxia, the mimetics caused no significant increase in VEGF, MYC or CYR61 mRNA half-life. Knockdown of HIF-alpha or ARNT via siRNA also had no effect on hypoxic mRNA stabilization. Interestingly, treatment of HEK293T cells with the mitochondrial inhibitors rotenone and myxothiazol, or the glutathione peroxidase mimetic ebselen did prevent the hypoxic stabilization of VEGF, MYC, and CYR61, suggesting a role for mtROS in the process. Additionally, treatment with antimycin A, which has been shown to generate mtROS, was able to drive the normoxic stabilization of these mRNAs. Conclusion Overall these data suggest that hypoxic mRNA stabilization is independent of HIF transcriptional activity but requires mtROS.
Collapse
Affiliation(s)
- Grey W Fortenbery
- 1Brody School of Medicine, East Carolina University, Greenville, NC 27834 USA
| | - Brinda Sarathy
- 2Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834 USA
| | - Kristen R Carraway
- 2Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834 USA
| | - Kyle D Mansfield
- 2Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834 USA
| |
Collapse
|
23
|
Bursch W. A cell's agony of choice: how to cross the Styx? : From morphological to molecular approaches to disclose its decision. Wien Med Wochenschr 2018; 168:300-306. [PMID: 30141112 PMCID: PMC6132567 DOI: 10.1007/s10354-018-0652-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022]
Abstract
The original “apoptosis–necrosis” concept was based on morphology and (patho)physiological conditions of the occurrence of cell death: (1) apoptosis, with nuclear and cytoplasmic condensation/fragmentation prominent, exclusion of autolysis, considered to result from coordinated self-destruction of a cell; (2) necrosis, with cell lysis prominent, caused by violent environmental perturbation leading to collapse of internal homeostasis. This suggestion initiated a controversial discussion within the scientific community and it soon became clear that the “apoptosis–necrosis dichotomy” was not generally applicable. Nowadays, there is sufficient evidence that cells may activate diverse suicide pathways, thereby allowing a flexible response to environmental changes, either physiological or pathological. The present paper commemorates electron microscopic and cytochemical studies on cell death of cultured human mammary carcinoma cells performed by Adi Ellinger, adding a significant contribution to recognize that autophagy can be involved in regulated cell death, thereby challenging the apoptosis–necrosis dichotomy still predominant in the 1990s.
Collapse
Affiliation(s)
- Wilfried Bursch
- Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| |
Collapse
|
24
|
Abstract
Obesity poses a severe threat to human health, including the increased prevalence of hypertension, insulin resistance, diabetes mellitus, cancer, inflammation, sleep apnoea and other chronic diseases. Current therapies focus mainly on suppressing caloric intake, but the efficacy of this approach remains poor. A better understanding of the pathophysiology of obesity will be essential for the management of obesity and its complications. Knowledge gained over the past three decades regarding the aetiological mechanisms underpinning obesity has provided a framework that emphasizes energy imbalance and neurohormonal dysregulation, which are tightly regulated by autophagy. Accordingly, there is an emerging interest in the role of autophagy, a conserved homeostatic process for cellular quality control through the disposal and recycling of cellular components, in the maintenance of cellular homeostasis and organ function by selectively ridding cells of potentially toxic proteins, lipids and organelles. Indeed, defects in autophagy homeostasis are implicated in metabolic disorders, including obesity, insulin resistance, diabetes mellitus and atherosclerosis. In this Review, the alterations in autophagy that occur in response to nutrient stress, and how these changes alter the course of obesogenesis and obesity-related complications, are discussed. The potential of pharmacological modulation of autophagy for the management of obesity is also addressed.
Collapse
Affiliation(s)
- Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| |
Collapse
|
25
|
Nobiletin (NOB) suppresses autophagic degradation via over-expressing AKT pathway and enhances apoptosis in multidrug-resistant SKOV3/TAX ovarian cancer cells. Biomed Pharmacother 2018; 103:29-37. [PMID: 29635125 DOI: 10.1016/j.biopha.2018.03.126] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy could be used as an effective therapeutic treatment for ovarian cancer and subsequent peritoneal metastasis. However, the occurrence of drug resistance reduced the treatment effect originated from cancer chemotherapy. Accumulating evidences indicated the significant role of autophagy in tumor cell resistance to chemotherapy. Thus, inhibition of autophagy using natural compounds could be a promising candidate to overcome multidrug resistance in human ovarian cancer cells. Nobiletin (NOB), a polymethoxyflavonoid found in citrus fruits such as Citrus depressa and Citrus reticulate, exhibits a number of bioactivities. In the present study, NOB selectively suppressed the growth and proliferation of human SKOV3/TAX cells, inducing G0/G1 phase arrest and reducing G2/M phase, along with the increase of p53 and p21. In addition, NOB induced significant apoptosis in SKOV3/TAX cells through the intrinsic apoptosis pathway, as evidenced by the up-regulation of cleaved Caspase-9/-3 and PARP. Further, NOB impaired the autophagic degradation in SKOV3/TAX cells, resulting in autophagic flux inhibition. Moreover, the impaired autophagic flux enhanced NOB-induced apoptosis in SKOV3/TAX cells. Importantly, AKT signaling was activated by NOB, which was involved in autophagic degradation and apoptotic cell death. In conclusion, the findings here supplied the illustration that NOB could overcome multidrug resistance in human ovarian cancer cells through AKT-regulated suppression of autophagic degradation.
Collapse
|
26
|
Jung S, Moon HI, Lee BS, Kim S, Quynh NTN, Yu J, Le DDT, Sandag Z, Lee H, Lee H, Anh NH, Yang Y, Lim JS, Kim KI, Lee MS. Anti-cancerous effect of cis-khellactone from Angelica amurensis through the induction of three programmed cell deaths. Oncotarget 2018; 9:16744-16757. [PMID: 29682182 PMCID: PMC5908283 DOI: 10.18632/oncotarget.24686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/27/2018] [Indexed: 11/29/2022] Open
Abstract
Angelica amurensis has traditionally been used to treat various medical problems. In this report, we introduce cis-khellactone as a new anti-cancer agent, which was isolated from the chloroform soluble fraction of the rhizomes of Angelica amurensis. Its anti-cancerous effect was at first tested in MCF7 and MDA-MB-231 breast cell lines, in which MCF7 is well known to be resistant to many anti-cancer drugs; MCF10A normal breast cell line was used as a control. In vitro experiments showed that cis-khellactone suppressed cell growth and proliferation at a relatively low concentrations (<5 μg/ml) and decreased cell viability at high concentrations (>10 μg/ml) in both cancer cell lines in a time- and concentration-dependent manner. This anti-cancerous effect was also checked in additional 16 different types of normal and cancer cell lines. Cis-khellactone treatment significantly suppressed cell proliferation and enhanced cell death in all tested cancer cell lines. Furthermore, Western blot analysis showed that cis-khellactone induced three types of programmed cell death (PCD): apoptosis, autophagy-mediated cell death, and necrosis/necroptosis. Cis-khellactone concentration-dependently decreased cell viability by increasing the level of reactive oxygen species (ROS) and decreasing mitochondrial membrane potential (MMP), which are related to all three types of PCD. Mitochondrial fractionation data revealed that cis-khellactone induced the translocation of BAX and BAK into mitochondria as well as the overexpression of VDAC1, which probably accelerates MMP disruption and finally cell death. Importantly, our extended in vivo studies with xenograft model further confirmed these findings of anti-cancerous effects and showed no harmful effects in normal tissues, suggesting that there would be no side effects in humans.
Collapse
Affiliation(s)
- Samil Jung
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Hyung-In Moon
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Beom Suk Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Subeen Kim
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Nguyen Thi Ngoc Quynh
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Jimin Yu
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, 49315, South Korea
| | - Dan-Diem Thi Le
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Zolzaya Sandag
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Hyegyeong Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Hyojeong Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Nguyen Hai Anh
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Young Yang
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Jong-Seok Lim
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Keun-Il Kim
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| | - Myeong-Sok Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 14310, South Korea
| |
Collapse
|
27
|
Efficacy of 2-hydroxy-3-phenylsulfanylmethyl-[1,4]-naphthoquinone derivatives against different Trypanosoma cruzi discrete type units: Identification of a promising hit compound. Eur J Med Chem 2017; 144:572-581. [PMID: 29289882 DOI: 10.1016/j.ejmech.2017.12.052] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/07/2017] [Accepted: 12/14/2017] [Indexed: 11/23/2022]
Abstract
The limited efficacy of benznidazole (Bz) indicated by failures of current Phase II clinical trials emphasizes the urgent need to identify new drugs with improved safety and efficacy for treatment of Chagas disease (CD). Herein, we analyzed the efficacy of a series of 2-hydroxy-3-phenylsulfanylmethyl-[1,4]-naphthoquinones against different Trypanosoma cruzi discrete type units (DTUs) of relevant clinical forms of CD. Cytotoxic and trypanocidal effect of naphthoquinone derivatives were assessed in mammalian cells, trypomastigotes and intracellular amastigotes using, luminescent assays (CellTiter-Glo and T. cruzi Dm28c-luciferase) and/or counting with a light microscope. Reactive oxygen species (ROS) production and intracellular targets of promising compounds were assessed with 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) probe and ultrastructural analysis, respectively. ADMET properties were analyzed by in silico modeling. Most of the compounds showed low cytotoxic effect. Only two compounds (Compounds 2 and 11) had IC50 values lower than Bz, showing higher susceptibility of bloodstream trypomastigotes. Compound 2 exhibited greater efficacy against trypomastigotes from different T. cruzi DTUs, even better than Bz against Brazil and CL strains. Ultrastructural analysis revealed changes in intracellular compartments, suggesting autophagy as one possible mechanism of action. Oxidative stress, induced by Compound 2, resulted in elevated level of ROS, leading to parasite death. Compound 2 was also effective against intracellular amastigotes, showing high selectivity index. ADMET analysis predicted good oral bioavailability, reduced drug metabolism and no carcinogenic potential for Compound 2. The data highlight Compound 2 as a hit compound and stimulate further structural and pharmacological optimization to potentiate its trypanocidal activity and selectivity.
Collapse
|
28
|
Granchi D, Savarino LM, Ciapetti G, Baldini N. Biological effects of metal degradation in hip arthroplasties. Crit Rev Toxicol 2017; 48:170-193. [PMID: 29130357 DOI: 10.1080/10408444.2017.1392927] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Metals and metal alloys are the most used materials in orthopedic implants. The focus is on total hip arthroplasty (THA) that, though well tolerated, may be associated with local and remote adverse effects in the medium-long term. This review aims to summarize data on the biological consequences of the metal implant degradation that have been attributed predominantly to metal-on-metal (MoM) THA. Local responses to metals consist of a broad clinical spectrum ranging from small asymptomatic tissue lesions to severe destruction of bone and soft tissues, which are designated as metallosis, adverse reactions to metal debris (ARMD), aseptic lymphocytic vasculitis associated lesion (ALVAL), and pseudotumors. In addition, the dissemination of metal particles and ions throughout the body has been associated with systemic adverse effects, including organ toxicity, cancerogenesis, teratogenicity, and immunotoxicity. As proved by the multitude of studies in this field, metal degradation may increase safety issues associated with THA, especially with MoM hip systems. Data collection regarding local, systemic and long-term effects plays an essential role to better define any safety risks and to generate scientifically based recommendations.
Collapse
Affiliation(s)
- Donatella Granchi
- a Orthopedic Pathophysiology and Regenerative Medicine Unit , Rizzoli Orthopedic Institute , Bologna , Italy
| | - Lucia Maria Savarino
- a Orthopedic Pathophysiology and Regenerative Medicine Unit , Rizzoli Orthopedic Institute , Bologna , Italy
| | - Gabriela Ciapetti
- a Orthopedic Pathophysiology and Regenerative Medicine Unit , Rizzoli Orthopedic Institute , Bologna , Italy
| | - Nicola Baldini
- a Orthopedic Pathophysiology and Regenerative Medicine Unit , Rizzoli Orthopedic Institute , Bologna , Italy.,b Department of Biomedical and Neuromotor Science , University of Bologna , Bologna , Italy
| |
Collapse
|
29
|
Chaicharoenaudomrung N, Jaroonwitchawan T, Noisa P. Cordycepin induces apoptotic cell death of human brain cancer through the modulation of autophagy. Toxicol In Vitro 2017; 46:113-121. [PMID: 28987792 DOI: 10.1016/j.tiv.2017.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 08/09/2017] [Accepted: 10/02/2017] [Indexed: 01/19/2023]
Abstract
Brain cancer, in particular neuroblastoma and glioblastoma, is a global challenge to human health. Cordycepin, extracted from Cordyceps ssp., has been revealed as a strong anticancer agent through several ways; however, the mechanism, by which cordycepin counteracts brain cancers, is still poorly understood. In this study, the underlying mechanisms of cordycepin against human brain cancer cells were explored. SH-SY5Y and U251 cells were being a model to represent human neuroblastoma and glioblastoma, respectively. Here, it was found that cordycepin inhibited cell growth, and induced apoptosis in a dose-dependent manner in both SH-SY5Y and U-251 cell lines. The expression of pro-apoptotic genes, including P53, BAX, Caspase-3, and Caspase-9, were upregulated, whereas the expression of anti-apoptotic gene, BCL-2, was suppressed. Besides, cordycepin induced the generation of reactive oxygen species (ROS) along with the suppression of antioxidant genes, including GPX, SOD, and Catalase. Importantly, cordycepin was shown to involve in the activation of autophagy, which was evidenced by the increment of LC3I/II. The combination of cordycepin with chloroquine, an autophagy inhibitor, further inhibited the growth, and enhanced the death of brain cancer cells. Altogether, this finding suggested that cordycepin induced apoptosis of human brain cancer cells through mitochondrial-mediated intrinsic pathway and the modulation of autophagy. Therefore, cordycepin could be a promising candidate for the development of anticancer drugs targeting human brain cancers.
Collapse
Affiliation(s)
- Nipha Chaicharoenaudomrung
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Nakhon Ratchasima 30000, Thailand
| | - Thiranut Jaroonwitchawan
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Nakhon Ratchasima 30000, Thailand
| | - Parinya Noisa
- Laboratory of Cell-Based Assays and Innovations, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
30
|
Carraway KR, Johnson EM, Kauffmann TC, Fry NJ, Mansfield KD. Hypoxia and Hypoglycemia synergistically regulate mRNA stability. RNA Biol 2017; 14:938-951. [PMID: 28362162 PMCID: PMC5546718 DOI: 10.1080/15476286.2017.1311456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ischemic events, common in many diseases, result from decreased blood flow and impaired delivery of oxygen and glucose to tissues of the body. While much is known about the cellular transcriptional response to ischemia, much less is known about the posttranscriptional response to oxygen and glucose deprivation. The goal of this project was to investigate one such posttranscriptional response, the regulation of mRNA stability. To that end, we have identified several novel ischemia-related mRNAs that are synergistically stabilized by oxygen and glucose deprivation including VEGF, MYC, MDM2, and CYR61. This increase in mRNA half-life requires the synergistic effects of both low oxygen (1%) as well as low glucose (≤ 1 g/L) conditions. Oxygen or glucose deprivation alone fails to initiate the response, as exposure to either high glucose (4 g/L) or normoxic conditions inhibits the response. Furthermore, in response to hypoxia/hypoglycemia, the identified mRNAs are released from the RNA binding protein KHSRP which likely contributes to their stabilization.
Collapse
Affiliation(s)
- Kristen R Carraway
- a Biochemistry and Molecular Biology, Brody School of Medicine , East Carolina University , Greenville , NC , USA
| | - Ellen M Johnson
- a Biochemistry and Molecular Biology, Brody School of Medicine , East Carolina University , Greenville , NC , USA
| | - Travis C Kauffmann
- b Brody School of Medicine , East Carolina University , Greenville , NC , USA
| | - Nate J Fry
- a Biochemistry and Molecular Biology, Brody School of Medicine , East Carolina University , Greenville , NC , USA
| | - Kyle D Mansfield
- a Biochemistry and Molecular Biology, Brody School of Medicine , East Carolina University , Greenville , NC , USA
| |
Collapse
|
31
|
Lien JC, Lin MW, Chang SJ, Lai KC, Huang AC, Yu FS, Chung JG. Tetrandrine induces programmed cell death in human oral cancer CAL 27 cells through the reactive oxygen species production and caspase-dependent pathways and associated with beclin-1-induced cell autophagy. ENVIRONMENTAL TOXICOLOGY 2017; 32:329-343. [PMID: 26822499 DOI: 10.1002/tox.22238] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 06/05/2023]
Abstract
Tetrandrine, a bisbenzylisoquinoline alkaloid, is extracted from the root of the Chinese herb Radix Stephania tetrandra S Moore. This compound has antitumor activity in different cancer cell types. In this study, the effects of tetrandrine on human oral cancer CAL 27 cells were examined. Results indicated that tetrandrine induced cytotoxic activity in CAL 27 cells. Effects were due to cell death by the induction of apoptosis and accompany with autophagy and these effects were concentration- and time-dependent manners. Tetrandrine induced apoptosis was accompanied by alterations in cell morphology, chromatin fragmentation, and caspase activation in CAL 27 cells. Tetrandrine treatment also induced intracellular accumulation of reactive oxygen species (ROS). The generation of ROS may play an important role in tetrandrine-induced apoptosis. Tetrandrine triggered LC3B expression and induced autophagy in CAL 27 cells. Tetrandrine induced apoptosis and autophagy were significantly attenuated by N-acetylcysteine pretreatment that supports the involvement of ROS production. Tetrandrine induced cell death may act through caspase-dependent apoptosis with Beclin-1-induced autophagy in human oral cancer cells. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 329-343, 2017.
Collapse
Affiliation(s)
- Jin-Cherng Lien
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung, 404, Taiwan
| | - Meng-Wei Lin
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei, 231, Taiwan
| | - Shu-Jen Chang
- School of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Kuang-Chi Lai
- Department of Surgery, China Medical University Beigang Hospital, Yunlin, 651, Taiwan
- School of Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - An-Cheng Huang
- Department of Nursing, St. Mary's Medicine Nursing and Management College, Yilan, 266, Taiwan
| | - Fu-Shun Yu
- Department of Dentist, China Medical University, Taichung, 404, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
32
|
Rendina-Ruedy E, Graef J, Lightfoot S, Ritchey J, Clarke S, Lucas E, Smith B. Impaired glucose tolerance attenuates bone accrual by promoting the maturation of osteoblasts: Role of Beclin1-mediated autophagy. Bone Rep 2016; 5:199-207. [PMID: 28580387 PMCID: PMC5440954 DOI: 10.1016/j.bonr.2016.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/07/2016] [Indexed: 11/22/2022] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) experience a 1.5-3.5 fold increase in fracture risk, but the mechanisms responsible for these alterations in bone biomechanical properties remain elusive. Macroautophagy, often referred to as autophagy, is regulated by signaling downstream of the insulin receptor. Metabolic changes associated with the progression of glucose intolerance have been shown to alter autophagy in various tissues, but limited information is available in relation to bone cells. The aim of this study was to (a) investigate whether autophagy is altered in bone tissue during impaired glucose tolerance, and (b) determine how autophagy impacts osteoblast differentiation, activity, and maturation. Four-week-old, male C57BL/6 mice were fed a control (Con) or high fat (HF) diet for 2, 8, or 16 wks. Mice on the HF diet demonstrated elevated fasting blood glucose and impaired glucose tolerance. Reduced trabecular bone in the femoral neck was evident in the mice on the HF diet by 8 wks compared to Con mice. Histological evaluation of the tibia suggested that the high fat diet promoted terminal differentiation of the osteoblast to an osteocyte. This shift of the osteoblasts towards a non-mineralizing, osteocyte phenotype appears to be coordinated by Beclin1-mediated autophagy. Consistent with these changes in the osteoblast in vivo, the induction of autophagy was able to direct MC3T3-E1 cells towards a more mature osteoblast phenotype. Although these data are somewhat observational, further investigation is warranted to determine if Beclin1-mediated autophagy is essential for the terminal differentiation of the osteoblasts and whether autophagy is having a protective or deleterious effect on bone in T2DM.
Collapse
Key Words
- AGEs, advanced glycation end products
- AIN, American Institute of Nutrition
- AMPK, adenosine monophosphate-activated protein kinase
- Ambra1, vacuole sorting protein (Vps34/15), activating molecule in Beclin-1 regulator autophagy
- Atg, autophagy-related proteins
- BafA1, bafilomycinA1
- Beclin1, Bcl-2-interacting myosin-like coiled-coil protein
- FIP200, focal adhesion
- Hyperglycemia
- IR, insulin receptor
- Insulin
- LC3, microtubule associated light chain
- Macroautophagy
- Osteocyte
- PE, phosphatidylethanolamine
- ROCK1, rho kinase 1
- Rap, rapamycin
- T2DM, type 2 diabetes mellitus
- ULK1/2, unc-like kinase
- UVRAG, ultraviolet radiation resistance-associated gene
- mTORC1, mammalian or mechanistic target of rapamycin complex
Collapse
Affiliation(s)
- E. Rendina-Ruedy
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - J.L. Graef
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - S.A. Lightfoot
- Center for Cancer Prevention and Drug Development, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - J.W. Ritchey
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK, United States
| | - S.L. Clarke
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - E.A. Lucas
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| | - B.J. Smith
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
33
|
McKeown CR, Thompson CK, Cline HT. Reversible developmental stasis in response to nutrient availability in the Xenopus laevis central nervous system. ACTA ACUST UNITED AC 2016; 220:358-368. [PMID: 27875263 DOI: 10.1242/jeb.151043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/04/2016] [Indexed: 12/31/2022]
Abstract
Many organisms confront intermittent nutrient restriction (NR), but the mechanisms to cope with nutrient fluctuations during development are not well understood. This is particularly true of the brain, the development and function of which is energy intensive. Here we examine the effects of nutrient availability on visual system development in Xenopus laevis tadpoles. During the first week of development, tadpoles draw nutrients from maternally provided yolk. Upon yolk depletion, animals forage for food. By altering access to external nutrients after yolk depletion, we identified a period of reversible stasis during tadpole development. We demonstrate that NR results in developmental stasis characterized by a decrease in overall growth of the animals, a failure to progress through developmental stages, and a decrease in volume of the optic tectum. During NR, neural progenitors virtually cease proliferation, but tadpoles swim and behave normally. Introducing food after temporary NR increased neural progenitor cell proliferation more than 10-fold relative to NR tadpoles, and cell proliferation was comparable to that of fed counterparts 1 week after delayed feeding. Delayed feeding also rescued NR-induced body length and tectal volume deficits and partially rescued developmental progression defects. Tadpoles recover from developmental stasis if food is provided within the first 9 days of NR, after which access to food fails to increase cell proliferation. These results show that early stages of tadpole brain development are acutely sensitive to fluctuations in nutrient availability and that NR induces developmental stasis from which animals can recover if food becomes available within a critical window.
Collapse
Affiliation(s)
- C R McKeown
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - C K Thompson
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - H T Cline
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
34
|
Jung S, Li C, Duan J, Lee S, Kim K, Park Y, Yang Y, Kim KI, Lim JS, Cheon CI, Kang YS, Lee MS. TRIP-Br1 oncoprotein inhibits autophagy, apoptosis, and necroptosis under nutrient/serum-deprived condition. Oncotarget 2016; 6:29060-75. [PMID: 26334958 PMCID: PMC4745711 DOI: 10.18632/oncotarget.5072] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/10/2015] [Indexed: 11/25/2022] Open
Abstract
TRIP-Br1 oncogenic protein has been shown to have multiple biological functions in cells. In this study, we demonstrate that TRIP-Br1 functions as an oncoprotein by inhibiting autophagy, apoptosis, and necroptosis of cancer cells and eventually helping them to survive under the nutrient/serum starved condition. TRIP-Br1 expression level was significantly increased in conditions with low levels of nutrients. Nutrient depleted conditions were induced by culturing cancer cells until they were overcrowded with high cell density or in media deprived of glucose, amino acids, or serum. Among them, serum starvation significantly enhanced the expression of TRIP-Br1 only in all tested breast cancer cell lines (MCF7, MDA-MB-231, T47D, MDA-MB-435, Hs578D, BT549, and MDA-MB-435) but not in the three normal cell lines (MCF10A, HfCH8, and NIH3T3). As compared with the control cells, the introduction of TRIP-Br1 silencing siRNA into MCF7 and MDA-MB-231 cells accelerated cell death by inducing apoptosis and necroptosis. In this process, TRIP-Br1 confers resistance to serum starvation-induced cell deaths by stabilizing the XIAP protein and inhibiting cellular ROS production. Moreover, our data also show that the intracellular increase of TRIP-Br1 protein resulting from serum starvation seems to occur in part through the blockage of PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Samil Jung
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Chengping Li
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Jingjing Duan
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Soonduck Lee
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Kyeri Kim
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Yeonji Park
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Young Yang
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Keun-Il Kim
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Jong-Seok Lim
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Chung-Il Cheon
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Young-Sook Kang
- College of Pharmacy, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Myeong-Sok Lee
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| |
Collapse
|
35
|
Li C, Jung S, Lee S, Jeong D, Yang Y, Kim KI, Lim JS, Cheon CI, Kim C, Kang YS, Lee MS. Nutrient/serum starvation derived TRIP-Br3 down-regulation accelerates apoptosis by destabilizing XIAP. Oncotarget 2016; 6:7522-35. [PMID: 25691055 PMCID: PMC4480697 DOI: 10.18632/oncotarget.3112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/08/2015] [Indexed: 12/19/2022] Open
Abstract
TRIP-Br3 and TRIP-Br1 have shown to have important biological functions. However, the function of TRIP-Br3 in tumorigenesis is not well characterized compared to oncogenic TRIP-Br1. Here, we investigated the function of TRIP-Br3 in tumorigenesis by comparing with that of TRIP-Br1. Under nutrient/serum starvation, TRIP-Br3 expression was down-regulated slightly in cancer cells and significantly in normal cells. Unexpectedly, TRIP-Br1 expression was greatly up-regulated in cancer cells but not in normal cells. Moreover, TRIP-Br3 activated autophagy while TRIP-Br1 inactivated it under serum starvation. In spite of different expression and roles of TRIP-Br3 and TRIP-Br1, both of them alleviate cell death by directly binding to and stabilizing XIAP, a potent apoptosis inhibitor, through blocking its ubiquitination. Taken together, we propose that TRIP-Br3 primarily activates the autophagy and suppresses apoptosis in nutrient sufficient condition. However, the prolonged extreme stressful condition of nutrient starvation causes a dramatic decrease of TRIP-Br3, which in turn induces apoptosis by destabilizing XIAP. Up-regulated TRIP-Br1 in cancer cells compensates this effect and delays apoptosis. This can be explained by the competitive alternative binding of TRIP-Br3 and TRIP-Br1 to the BIR2 domain of XIAP. In an extended study, our immunohistochemical analysis revealed a markedly lower level of TRIP-Br3 protein in human carcinoma tissues compared to normal epithelial tissues, implying the role of TRIP-Br3 as a tumor suppressor rather than onco-protein.
Collapse
Affiliation(s)
- Chengping Li
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Samil Jung
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Soonduck Lee
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Dongjun Jeong
- Department of Pathology, College of Medicine, Soonchunhyang University, Chonan, 330-090, South Korea
| | - Young Yang
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Keun-Il Kim
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Jong-Seok Lim
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Chung-Il Cheon
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Changjin Kim
- Department of Pathology, College of Medicine, Soonchunhyang University, Chonan, 330-090, South Korea
| | - Young-Sook Kang
- College of Pharmacy, Sookmyung Women's University, Seoul, 140-742, South Korea
| | - Myeong-Sok Lee
- Department of Life Systems, Sookmyung Women's University, Seoul, 140-742, South Korea
| |
Collapse
|
36
|
Inhibitory role of TRIP-Br1 oncoprotein in hypoxia-induced apoptosis in breast cancer cell lines. Int J Oncol 2016; 48:2639-46. [PMID: 27035851 DOI: 10.3892/ijo.2016.3454] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/18/2016] [Indexed: 11/05/2022] Open
Abstract
TRIP-Br1 oncoprotein is known to be involved in many vital cellular functions. In this study, we examined the role of TRIP-Br1 in hypoxia-induced cell death. Exposure to the overcrowded and CoCl2-induced hypoxic conditions increased TRIP-Br1 expression at the protein level in six breast cancer cell lines (MCF7, MDA-MB-231, T47D, Hs578D, BT549, and MDA-MB-435) but resulted in no significant change in three normal cell lines (MCF10A, MEF and NIH3T3). Our result revealed that CoCl2-induced hypoxia stimulated apoptosis and autophagy, in which TRIP-Br1 expression was found to be upregulated. Interestingly, TRIP-Br1 silencing in the MCF7 and MDA-MB-231 cancer cells accelerated apoptosis and destabilization of XIAP under the CoCl2-induced hypoxic condition, implying that TRIP-Br1 may render cancer cells resistant to apoptosis through the stabilization of XIAP. We also propose that TRIP-Br1 seems to be upregulated at least partly as a result of the inhibition of PI3K/AKT signaling pathway and the overexpression of HIF-1α. In conclusion, our findings suggest that TRIP-Br1 functions as an oncogenic protein by providing cancer cells resistance to the hypoxia-induced cell death.
Collapse
|
37
|
Yu FS, Yu CS, Chen JC, Yang JL, Lu HF, Chang SJ, Lin MW, Chung JG. Tetrandrine induces apoptosis Via caspase-8, -9, and -3 and poly (ADP ribose) polymerase dependent pathways and autophagy through beclin-1/ LC3-I, II signaling pathways in human oral cancer HSC-3 cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:395-406. [PMID: 25266202 DOI: 10.1002/tox.22053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 06/03/2023]
Abstract
Tetrandrine is a bisbenzylisoquinoline alkaloid that was found in the Radix Stephania tetrandra S Moore. It had been reported to induce cytotoxic effects on many human cancer cells. In this study, we investigated the cytotoxic effects of tetrandrine on human oral cancer HSC-3 cells in vitro. Treatments of HSC-3 cells with tetrandrine significantly decreased the percentage of viable cells through the induction of autophagy and apoptosis and these effects are in concentration-dependent manner. To define the mechanism underlying the cytotoxic effects of tetrandrine, we investigated the critical molecular events known to regulate the apoptotic and autophagic machinery. Tetrandrine induced chromatin condensation, internucleosomal DNA fragmentation, activation of caspases-3, -8, and -9, and cleavage of poly (ADP ribose) polymerase (PARP) that were associated with apoptosis, and it also enhanced the expression of LC3-I and -II that were associated with the induction of autophagy in human squamous carcinoma cell line (HSC-3) cells. Tetrandrine induced autophagy in HSC-3 cells was significantly attenuated by bafilomycin A1 (inhibitor of autophagy) pre-treatment that confirmed tetrandrine induced cell death may be associated with the autophagy. In conclusion, we suggest that tetrandrine induced cell death may be through the induction of apoptosis as well as autophagy in human oral cancer HSC-3 cells via PARP, caspases/Becline I/LC3-I/II signaling pathways.
Collapse
Affiliation(s)
- Fu-Shun Yu
- Department of Dentist, China Medical University, Taichung, 404, Taiwan
| | - Chun-Shu Yu
- School of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Jaw-Chyun Chen
- Department of Medicinal Botany and Health Applications, Da-Yeh University, Changhua, Taiwan
| | - Jiun-Long Yang
- Department of Chinese Medicine Resources, China Medical University, Taichung, 404, Taiwan
| | - Hsu-Feng Lu
- Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Shu-Jen Chang
- School of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Meng-Wei Lin
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, Yilan, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
38
|
Costa L, Amaral C, Teixeira N, Correia-da-Silva G, Fonseca BM. Cannabinoid-induced autophagy: Protective or death role? Prostaglandins Other Lipid Mediat 2015; 122:54-63. [PMID: 26732541 DOI: 10.1016/j.prostaglandins.2015.12.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 12/07/2015] [Accepted: 12/21/2015] [Indexed: 12/11/2022]
Abstract
Autophagy, the "self-digestion" mechanism of the cells, is an evolutionary conserved catabolic process that targets portions of cytoplasm, damaged organelles and proteins for lysosomal degradation, which plays a crucial role in development and disease. Cannabinoids are active compounds of Cannabis sativa and the most prevalent psychoactive substance is Δ(9)-tetrahydrocannabinol (THC). Cannabinoid compounds can be divided in three types: the plant-derived natural products (phytocannabinoids), the cannabinoids produced endogenously (endocannabinoids) and the synthesized compounds (synthetic cannabinoids). Various studies reported a cannabinoid-induced autophagy mechanism in cancer and non-cancer cells. In this review we focus on the recent advances in the cannabinoid-induced autophagy and highlight the molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Lia Costa
- Departamento de Biologia, Universidade de Aveiro, Portugal; UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Portugal
| | - Cristina Amaral
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Portugal
| | - Natércia Teixeira
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Portugal
| | - Bruno M Fonseca
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Portugal.
| |
Collapse
|
39
|
Macrophage migration inhibitory factor has a permissive role in concanavalin A-induced cell death of human hepatoma cells through autophagy. Cell Death Dis 2015; 6:e2008. [PMID: 26633714 PMCID: PMC4720884 DOI: 10.1038/cddis.2015.349] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 02/06/2023]
Abstract
Concanavalin A (ConA) is a lectin and T-cell mitogen that can activate immune responses. In recent times, ConA-induced cell death of hepatoma cells through autophagy has been reported and its therapeutic effect was confirmed in a murine in situ hepatoma model. However, the molecular mechanism of ConA-induced autophagy is still unclear. As macrophage migration inhibitory factor (MIF), which is a proinflammatory cytokine, can trigger autophagy in human hepatoma cells, the possible involvement of MIF in ConA-induced autophagy was investigated in this study. We demonstrated that cell death is followed by an increment in MIF expression and secretion in the ConA-stimulated human hepatoma cell lines, HuH-7 and Hep G2. In addition, ConA-induced autophagy and cell death of hepatoma cells were blocked in the presence of an MIF inhibitor. Knockdown of endogenous MIF by small hairpin RNA confirmed that MIF is required for both ConA-induced autophagy and death of hepatoma cells. Furthermore, signal pathway studies demonstrated that ConA induces signal transducer and activator of transcription 3 (STAT3) phosphorylation to trigger MIF upregulation, which in turn promotes Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (BNIP3)-dependent autophagy. By using a murine in situ hepatoma model, we further demonstrated that MIF contributes to anti-hepatoma activity of ConA by regulating STAT3-MIF-BNIP3-dependent autophagy. In summary, our findings uncover a novel role of MIF in lectin-mediated anti-hepatoma activities by regulating autophagy.
Collapse
|
40
|
Golden EB, Cho HY, Hofman FM, Louie SG, Schönthal AH, Chen TC. Quinoline-based antimalarial drugs: a novel class of autophagy inhibitors. Neurosurg Focus 2015; 38:E12. [PMID: 25727221 DOI: 10.3171/2014.12.focus14748] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Chloroquine (CQ) is a quinoline-based drug widely used for the prevention and treatment of malaria. More recent studies have provided evidence that this drug may also harbor antitumor properties, whereby CQ possesses the ability to accumulate in lysosomes and blocks the cellular process of autophagy. Therefore, the authors of this study set out to investigate whether CQ analogs, in particular clinically established antimalaria drugs, would also be able to exert antitumor properties, with a specific focus on glioma cells. METHODS Toward this goal, the authors treated different glioma cell lines with quinine (QN), quinacrine (QNX), mefloquine (MFQ), and hydroxychloroquine (HCQ) and investigated endoplasmic reticulum (ER) stress-induced cell death, autophagy, and cell death. RESULTS All agents blocked cellular autophagy and exerted cytotoxic effects on drug-sensitive and drug-resistant glioma cells with varying degrees of potency (QNX > MFQ > HCQ > CQ > QN). Furthermore, all quinoline-based drugs killed glioma cells that were highly resistant to temozolomide (TMZ), the current standard of care for patients with glioma. The cytotoxic mechanism involved the induction of apoptosis and ER stress, as indicated by poly(ADP-ribose) polymerase (PARP) cleavage and CHOP/GADD153. The induction of ER stress and resulting apoptosis could be confirmed in the in vivo setting, in which tumor tissues from animals treated with quinoline-based drugs showed increased expression of CHOP/GADD153, along with elevated TUNEL staining, a measure of apoptosis. CONCLUSIONS Thus, the antimalarial compounds investigated in this study hold promise as a novel class of autophagy inhibitors for the treatment of newly diagnosed TMZ-sensitive and recurrent TMZ-resistant gliomas.
Collapse
Affiliation(s)
- Encouse B Golden
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
| | | | | | | | | | | |
Collapse
|
41
|
Chen M, Suzuki A, Borlak J, Andrade RJ, Lucena MI. Drug-induced liver injury: Interactions between drug properties and host factors. J Hepatol 2015; 63:503-14. [PMID: 25912521 DOI: 10.1016/j.jhep.2015.04.016] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a common cause for drug withdrawal from the market and although infrequent, DILI can result in serious clinical outcomes including acute liver failure and the need for liver transplantation. Eliminating the iatrogenic "harm" caused by a therapeutic intent is a priority in patient care. However, identifying culprit drugs and individuals at risk for DILI remains challenging. Apart from genetic factors predisposing individuals at risk, the role of the drugs' physicochemical and toxicological properties and their interactions with host and environmental factors need to be considered. The influence of these factors on mechanisms involved in DILI is multi-layered. In this review, we summarize current knowledge on 1) drug properties associated with hepatotoxicity, 2) host factors considered to modify an individuals' risk for DILI and clinical phenotypes, and 3) drug-host interactions. We aim at clarifying knowledge gaps needed to be filled in as to improve risk stratification in patient care. We therefore broadly discuss relevant areas of future research. Emerging insight will stimulate new investigational approaches to facilitate the discovery of clinical DILI risk modifiers in the context of disease complexity and associated interactions with drug properties, and hence will be able to move towards safety personalized medicine.
Collapse
Affiliation(s)
- Minjun Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Ayako Suzuki
- Gastroenterology, Central Arkansas Veterans Healthcare System, Little Rock, AR, United States; Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jürgen Borlak
- Center of Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Raúl J Andrade
- Unidad de Gestión Clínica de Enfermedades Digestivas, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| | - M Isabel Lucena
- Unidad de Gestión Clínica de Enfermedades Digestivas, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| |
Collapse
|
42
|
Netea-Maier RT, Plantinga TS, van de Veerdonk FL, Smit JW, Netea MG. Modulation of inflammation by autophagy: Consequences for human disease. Autophagy 2015. [PMID: 26222012 PMCID: PMC4836004 DOI: 10.1080/15548627.2015.1071759] [Citation(s) in RCA: 278] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Autophagy and inflammation are 2 fundamental biological processes involved in both physiological and pathological conditions. Through its crucial role in maintaining cellular homeostasis, autophagy is involved in modulation of cell metabolism, cell survival, and host defense. Defective autophagy is associated with pathological conditions such as cancer, autoimmune disease, neurodegenerative disease, and senescence. Inflammation represents a crucial line of defense against microorganisms and other pathogens, and there is increasing evidence that autophagy has important effects on the induction and modulation of the inflammatory reaction; understanding the balance between these 2 processes may point to important possibilities for therapeutic targeting. This review focuses on the crosstalk between autophagy and inflammation as an emerging field with major implications for understanding the host defense on the one hand, and for the pathogenesis and treatment of immune-mediated diseases on the other hand.
Collapse
Affiliation(s)
- Romana T Netea-Maier
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,b Division of Endocrinology, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Theo S Plantinga
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Frank L van de Veerdonk
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,c Radboud Center for Infectious Diseases, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Johannes W Smit
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,b Division of Endocrinology, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Mihai G Netea
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,c Radboud Center for Infectious Diseases, Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|
43
|
The role of autophagy and lipolysis in survival of astrocytes under nutrient deprivation. Neurosci Lett 2015; 595:128-33. [PMID: 25888813 DOI: 10.1016/j.neulet.2015.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/03/2015] [Accepted: 04/11/2015] [Indexed: 10/23/2022]
Abstract
Astrocytes can survive nutrient deprivation (ND) for days. However, the pro-survival strategy of astrocytes under such a metabolic challenge is still not clear. In the present study, we examined the effects of inhibition of two potential steps in energy acquisition during ND: autophagy (using chloroquine) and lipolysis (using orlistat). The inhibition of autophagy did not show significant effects on cell viability until 8-9h of ND. From that point onwards, the number of dead cells gradually increased, reaching ∼60% between 10 and 12h of ND. In addition, early inhibition of autophagy made astrocytes more vulnerable to the latter ND. The inhibition of lipolysis decreased the viability of cells exposed to ND, but this appeared much later compared to the inhibition of autophagy. The application of orlistat prevented ND-related hyperpolarization of the mitochondrial membrane, and mitochondria became swollen. This study clearly shows that autophagy and lipolysis are essential for the survival of astrocytes under ND conditions, which might be related to their role as neuron-supporting cells.
Collapse
|
44
|
Li X, Lao Y, Zhang H, Wang X, Tan H, Lin Z, Xu H. The natural compound Guttiferone F sensitizes prostate cancer to starvation induced apoptosis via calcium and JNK elevation. BMC Cancer 2015; 15:254. [PMID: 25885018 PMCID: PMC4394563 DOI: 10.1186/s12885-015-1292-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 03/30/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In a cytotoxicity screen in serum-free medium, Guttiferone F showed strong growth inhibitory effect against prostate cancer cells. METHODS Prostate cancer cells LNCaP and PC3 were treated with Guttiferone F in serum depleted medium. Sub-G1 phase distributions were estimated with flow cytometry. Mitochondrial disruption was observed under confocal microscope using Mitotracker Red staining. Gene and protein expression changes were detected by real-time PCR and Western blotting. Ca(2+) elevation was examined by Fluo-4 staining under fluorescence microscope. PC3 xenografts in mice were examined by immunohistochemical analysis. RESULTS Guttiferone F had strong growth inhibitory effect against prostate cancer cell lines under serum starvation. It induced a significant increase in sub-G1 fraction and DNA fragmentation. In serum-free medium, Guttiferone F triggered mitochondria dependent apoptosis by regulating Bcl-2 family proteins. In addition, Guttiferone F attenuated the androgen receptor expression and phosphorylation of ERK1/2, while activating the phosphorylation of JNK and Ca(2+) flux. Combination of caloric restriction with Guttiferone F in vivo could increase the antitumor effect without causing toxicity. CONCLUSIONS Guttiferone F induced prostate cancer cell apoptosis under serum starvation via Ca(2+) elevation and JNK activation. Combined with caloric restriction, Guttiferone F exerted significant growth inhibition of PC3 cells xenograft in vivo. Guttiferone F is therefore a potential anti-cancer compound.
Collapse
Affiliation(s)
- Xin Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China.
| | - Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China.
| | - Hong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China.
| | - Xiaoyu Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China.
| | - Hongsheng Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China.
| | - Zhixiu Lin
- School of Chinese Medicine, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, SAR, China.
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, P.R. China.
| |
Collapse
|
45
|
Vergilio C, Moreira R, Carvalho C, Melo E. Evolution of cadmium effects in the testis and sperm of the tropical fish Gymnotus carapo. Tissue Cell 2015; 47:132-9. [DOI: 10.1016/j.tice.2015.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/27/2022]
|
46
|
Cytoplasmic PELP1 and ERRgamma protect human mammary epithelial cells from Tam-induced cell death. PLoS One 2015; 10:e0121206. [PMID: 25789479 PMCID: PMC4366195 DOI: 10.1371/journal.pone.0121206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/28/2015] [Indexed: 01/24/2023] Open
Abstract
Tamoxifen (Tam) is the only FDA-approved chemoprevention agent for pre-menopausal women at high risk for developing breast cancer. While Tam reduces a woman's risk of developing estrogen receptor positive (ER+) breast cancer, the molecular mechanisms associated with risk reduction are poorly understood. Prior studies have shown that cytoplasmic proline, glutamic acid and leucine rich protein 1 (PELP1) promotes Tam resistance in breast cancer cell lines. Herein, we tested for PELP1 localization in breast epithelial cells from women at high risk for developing breast cancer and found that PELP1 was localized to the cytoplasm in 36% of samples. In vitro, immortalized HMECs expressing a nuclear localization signal (NLS) mutant of PELP1 (PELP1-cyto) were resistant to Tam-induced death. Furthermore, PELP1-cyto signaling through estrogen-related receptor gamma (ERRγ) promoted cell survival in the presence of Tam. Overexpression of ERRγ in immortalized HMECs protected cells from Tam-induced death, while knockdown of ERRγ sensitized PELP1-cyto expressing HMECs to Tam. Moreover, Tam-induced HMEC cell death was independent of apoptosis and involved accumulation of the autophagy marker LC3-II. Expression of PELP1-cyto and ERRγ reduced Tam-induced LC3-II accumulation, and knockdown of ERRγ increased LC3-II levels in response to Tam. Additionally, PELP1-cyto expression led to the upregulation of MMP-3 and MAOB, known PELP1 and ERRγ target genes, respectively. Our data indicate that cytoplasmic PELP1 induces signaling pathways that converge on ERRγ to promote cell survival in the presence of Tam. These data suggest that PELP1 localization and/or ERRγ activation could be developed as tissue biomarkers for Tam responsiveness.
Collapse
|
47
|
Bagul M, Kakumanu S, Wilson TA. Crude Garlic Extract Inhibits Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis of Cancer Cells In Vitro. J Med Food 2015; 18:731-7. [PMID: 25608085 DOI: 10.1089/jmf.2014.0064] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Garlic and its lipid-based extracts have played an important medicinal role in humans for centuries that includes antimicrobial, hypoglycemic, and lipid-lowering properties. The present study was to investigate the effects of crude garlic extract (CGE) on the proliferation of human breast, prostate, hepatic, and colon cancer cell lines and mouse macrophageal cells, not previously studied. The human cancer cell lines, such as hepatic (Hep-G2), colon (Caco-2), prostate (PC-3), and breast (MCF-7), were propagated at 37°C; air/CO2 (95:5 v/v) using the ATCC-formulated RPMI-1640 Medium and 10% fetal bovine serum (FBS), while the mouse macrophage cell line (TIB-71) was propagated at 37°C; air/CO2 (95:5 v/v) using the ATCC-formulated DMEM and 10% FBS. All cells were plated at a density of ∼5000 cells/well. After overnight incubation, the cells were treated with 0.125, 0.25, 0.5, or 1 μg/mL of CGE an additional 72 h. Inhibition of cell proliferation of 80-90% was observed for Hep-G2, MCF-7, TIB-71, and PC-3 cells, but only 40-55% for the Caco-2 cells when treated with 0.25, 0.5, or 1 μg/mL. In a coculture study of Caco-2 and TIB-71 cells, inhibition of cell proliferation of 90% was observed for Caco-2 cells compared to the 40-55% when cultured separately. CGE also induced cell cycle arrest and had a fourfold increase in caspase activity (apoptosis) in PC-3 cells when treated at a dose of 0.5 or 1 μg/mL. This investigation of CGE clearly highlights the fact that the lipid bioactive compounds in CGE have the potential as promising anticancer agents.
Collapse
Affiliation(s)
- Mukta Bagul
- 1 Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell , Lowell, Massachusetts, USA
| | - Srikanth Kakumanu
- 1 Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell , Lowell, Massachusetts, USA
| | - Thomas A Wilson
- 1 Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell , Lowell, Massachusetts, USA .,2 Department of Clinical Laboratory and Nutritional Sciences, Center for Health and Disease Research, University of Massachusetts Lowell , Lowell, Massachusetts, USA
| |
Collapse
|
48
|
Netea-Maier RT, Klück V, Plantinga TS, Smit JWA. Autophagy in thyroid cancer: present knowledge and future perspectives. Front Endocrinol (Lausanne) 2015; 6:22. [PMID: 25741318 PMCID: PMC4332359 DOI: 10.3389/fendo.2015.00022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/05/2015] [Indexed: 01/01/2023] Open
Abstract
Thyroid cancer is the most common endocrine malignancy. Despite having a good prognosis in the majority of cases, when the tumor is dedifferentiated it does no longer respond to conventional treatment with radioactive iodine, the prognosis worsens significantly. Treatment options for advanced, dedifferentiated disease are limited and do not cure the disease. Autophagy, a process of self-digestion in which damaged molecules or organelles are degraded and recycled, has emerged as an important player in the pathogenesis of different diseases, including cancer. The role of autophagy in thyroid cancer pathogenesis is not yet elucidated. However, the available data indicate that autophagy is involved in several steps of thyroid tumor initiation and progression as well as in therapy resistance and therefore could be exploited for therapeutic applications. The present review summarizes the most recent data on the role of autophagy in the pathogenesis of thyroid cancer and we will provide a perspective on how this process can be targeted for potential therapeutic approaches and could be further explored in the context of multimodality treatment in cancer and personalized medicine.
Collapse
Affiliation(s)
- Romana T. Netea-Maier
- Department of Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| | - Viola Klück
- Department of Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| | - Theo S. Plantinga
- Department of Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| | - Johannes W. A. Smit
- Department of Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
- *Correspondence: Johannes W. A. Smit, Department of Medicine, Division of Endocrinology, Radboud University Nijmegen Medical Center, Geert Grooteplein 8, PO Box 9101, Nijmegen 6500 HB, Netherlands e-mail:
| |
Collapse
|
49
|
Kawashima A, Sekizawa A, Koide K, Hasegawa J, Satoh K, Arakaki T, Takenaka S, Matsuoka R. Vitamin C Induces the Reduction of Oxidative Stress and Paradoxically Stimulates the Apoptotic Gene Expression in Extravillous Trophoblasts Derived From First-Trimester Tissue. Reprod Sci 2014; 22:783-90. [PMID: 25519716 DOI: 10.1177/1933719114561561] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
AIM To investigate the effects of vitamin C on the expression of the genes related to apoptosis in extravillous trophoblasts (EVTs) in the first trimester. METHODS Extravillous trophoblasts were cultured under 2% O2 followed by 2% O2 or 8% O2 with or without vitamin C. The level of reactive oxygen species (ROS) in the cultured medium was estimated using electron spin resonance spectroscopy. The expression levels of the genes TP53, BCL2, and BAX were quantified using real-time quantitative polymerase chain reaction. RESULTS Reactive oxygen species were found to be decreased after adding vitamin C under increasing oxygen concentrations. In addition, the ratio of BAX/BCL2 also increased after adding vitamin C under conditions of 2% O2, while the gene expression level of BCL2 increased after adding vitamin C under increasing oxygen concentrations. In contrast, the gene expression level of TP53 and the ratio of BAX/BCL2 both decreased. CONCLUSION We have revealed that vitamin C reduces ROS and may promote the apoptosis of EVTs under conditions of 2% O2 while paradoxically preventing apoptosis under increasing oxygen concentrations.
Collapse
Affiliation(s)
- Akihiro Kawashima
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| | - Akihiko Sekizawa
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| | - Keiko Koide
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| | - Junichi Hasegawa
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| | - Kazue Satoh
- Department of Anatomy, Showa University School of Medicine, Shinagawa, Japan
| | - Tatsuya Arakaki
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| | - Shin Takenaka
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| | - Ryu Matsuoka
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Shinagawa, Japan
| |
Collapse
|
50
|
Garoby-Salom S, Rouahi M, Mucher E, Auge N, Salvayre R, Negre-Salvayre A. Hyaluronan synthase-2 upregulation protects smpd3-deficient fibroblasts against cell death induced by nutrient deprivation, but not against apoptosis evoked by oxidized LDL. Redox Biol 2014; 4:118-26. [PMID: 25555205 PMCID: PMC4309855 DOI: 10.1016/j.redox.2014.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 01/02/2023] Open
Abstract
The neutral type 2 sphingomyelinase (nSMase2) hydrolyzes sphingomyelin and generates ceramide, a major bioactive sphingolipid mediator, involved in growth arrest and apoptosis. The role of nSMase2 in apoptosis is debated, and apparently contradictory results have been observed on fibroblasts isolated from nSMase2-deficient fragilitas ossium (homozygous fro/fro) mice. These mice exhibit a severe neonatal dysplasia, a lack of long bone mineralization and delayed apoptosis patterns of hypertrophic chondrocytes in the growth plate. We hypothesized that apoptosis induced by nutrient deprivation, which mimics the environmental modifications of the growth plate, requires nSMase2 activation. In this study, we have compared the resistance of fro/fro fibroblasts to different death inducers (oxidized LDL, hydrogen peroxide and nutrient starvation). The data show that nSMase2-deficient fro/fro cells resist to apoptosis evoked by nutrient starvation (fetal calf serum/glucose/pyruvate-free DMEM), whereas wt fibroblasts die after 48 h incubation in this medium. In contrast, oxidized LDL and hydrogen peroxide are similarly toxic to fro/fro and wt fibroblasts, indicating that nSMase2 is not involved in the mechanism of toxicity evoked by these agents. Interestingly, wt fibroblasts treated with the SMase inhibitor GW4869 were more resistant to starvation-induced apoptosis. The resistance of fro/fro cells to starvation-induced apoptosis is associated with an increased expression of hyaluronan synthase 2 (HAS2) mRNAs and protein, which is inhibited by ceramide. In wt fibroblasts, this HAS2 rise and its protective effect did not occur, but exogenously added HA exhibited a protective effect against starvation-induced apoptosis. The protective mechanism of HAS2 involves an increased expression of the heat-shock protein Hsp72, a chaperone with antiapoptotic activity. Taken together, these results highlight the role of nSMase2 in apoptosis evoked by nutrient starvation that could contribute to the delayed apoptosis of hypertrophic chondrocytes in the growth plate, and emphasize the antiapoptotic properties of HAS2. Apoptosis evoked by oxidized LDL and H2O2 is comparable in fro/fro and wt fibroblasts. fro/fro fibroblasts resist to apoptosis evoked by nutrient starvation. HAS2 increased expression protects fro/fro fibroblasts against apoptosis. HAS2 regulates the expression of the antiapoptotic heat-shock protein HsP72.
Collapse
Affiliation(s)
- Sandra Garoby-Salom
- INSERM UMR-1048, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Toulouse, France
| | - Myriam Rouahi
- INSERM UMR-1048, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Toulouse, France
| | - Elodie Mucher
- INSERM UMR-1048, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Toulouse, France
| | - Nathalie Auge
- INSERM UMR-1048, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Toulouse, France
| | - Robert Salvayre
- INSERM UMR-1048, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Toulouse, France
| | - Anne Negre-Salvayre
- INSERM UMR-1048, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Toulouse, France.
| |
Collapse
|