1
|
Lu X, Chen D, Wang M, Song X, Ermine K, Hao S, Jha A, Huang Y, Kang Y, Qiu H, Lenz HJ, Li S, Jin Z, Yu J, Zhang L. Depletion of oxysterol-binding proteins by OSW-1 triggers RIP1/RIP3-independent necroptosis and sensitization to cancer immunotherapy. Cell Death Differ 2025:10.1038/s41418-025-01521-8. [PMID: 40329104 DOI: 10.1038/s41418-025-01521-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Oxysterol-binding proteins (OSBPs), lipid transfer proteins functioning at intracellular membrane contact sites, are recently found to be dysregulated in cancer and promote cancer cell survival. However, their role as potential targets in cancer therapy remains largely unexplored. In this study, we found OSW-1, a natural compound and OSBP inhibitor, potently and selectively kills colon cancer cells by activating a previously unknown necroptosis pathway that is independent of receptor-interacting protein 1 (RIP1) and RIP3. OSW-1 stabilizes p53 and degrades OSBPs to promote endoplasmic reticulum (ER) stress and glycogen synthase kinase 3β (GSK3β)/Tip60-mediated p53 acetylation at Lysine 120, which selectively induces its target PUMA. PUMA-mediated mitochondrial calcium influx activates calcium/calmodulin-dependent protein kinase IIδ (CamKIIδ) to promote mixed lineage kinase domain-like (MLKL) phosphorylation and necroptotic cell death. Furthermore, OSW-1-induced necroptosis is highly immunogenic and sensitizes syngeneic colorectal tumors to anti-PD-1 immunotherapy. Together, our results identified a novel RIP1/RIP3-independent necroptosis pathway underlying the extremely potent anticancer activity of OSW-1, which can be harnessed to develop new anticancer therapies by selectively stimulating antitumor immunity.
Collapse
Affiliation(s)
- Xinyan Lu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dongshi Chen
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Min Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiangping Song
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kaylee Ermine
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Suisui Hao
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anupma Jha
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yixian Huang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Kang
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Haibo Qiu
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Heinz-Josef Lenz
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Song Li
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhendong Jin
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Jian Yu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Jang J, Jung H, Jeong J, Jeon J, Lee K, Jang HR, Han JW, Lee J. Modeling doxorubicin-induced-cardiotoxicity through breast cancer patient specific iPSC-derived heart organoid. Heliyon 2024; 10:e38714. [PMID: 39640743 PMCID: PMC11620051 DOI: 10.1016/j.heliyon.2024.e38714] [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: 04/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 12/07/2024] Open
Abstract
Heart organoid (HO) technology has successfully overcome the limitations of two-dimensional (2D) disease modeling and drug testing, thereby emerging as a valuable tool in drug discovery for assessing toxicity and efficacy. However, its ability to distinguish drug responses among individuals remain unclear, which is crucial for developing predictive models. We addressed this gap by comparing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with human induced pluripotent stem cell-derived heart organoids (hiPSC-HOs) in the context of doxorubicin-induced cardiotoxicity (DIC). For this study, we utilized hiPSCs generated from breast cancer patients who had previously been treated with doxorubicin. By comparing groups with and without DIC, we examined various parameters, including cell viability, mRNA expression, protein expression and electrophysiological variations. The results of our analysis revealed significant differences between these groups, providing insights into hiPSC-HOs as a potential platform for testing differences in drug responses among patients.
Collapse
Affiliation(s)
- Jiye Jang
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyewon Jung
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaekyun Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Junseok Jeon
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyungho Lee
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jeung-Whan Han
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaecheol Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
3
|
Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159515. [PMID: 38844203 DOI: 10.1016/j.bbalip.2024.159515] [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: 02/28/2024] [Revised: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.
Collapse
Affiliation(s)
- Wojciech Białek
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
4
|
Wei T, Chen L, Shi P, Wang C, Peng Y, Yang J, Liao X, Yang B, Gao C. Platinum (IV) drugs with cannabidiol inducing mitochondrial dysfunction and synergistically enhancing anti-tumor effects. J Inorg Biochem 2024; 254:112515. [PMID: 38490045 DOI: 10.1016/j.jinorgbio.2024.112515] [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: 12/05/2023] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
Chemotherapy resistance is an insurmountable problem in clinical anticancer therapy. Although Oxaliplatin is an effective chemotherapeutic agent for the treatment of colorectal cancer (CRC), it still suffers from serious toxicities as well as drug resistance. In this work, three Oxaliplatin tetravalent platinum prodrugs(O1-O3) and three novel mixed ammine/amine analogs(C1-C3) were constructed, introducing cannabidiol with anti-tumor activity in their axial position. All Pt(IV) prodrugs exhibited potent antitumor effects in a variety of tumor cell lines, especially in HCT-116 cells, where complex O3 showed strong inhibitory effects with the half maximal inhibitory concentrations (IC50) value of 6.02 ± 0.69 μM and about 2.6 times higher than that of Oxaliplatin. Further studies revealed that complex O3 decreased cellular mitochondrial membrane potential in a concentration-dependent manner and enhanced reactive oxygen species (ROS) accumulation by decreasing the expression of catalase, superoxide dismutase 2 (SOD2) and superoxide dismutase 3 (SOD3). Complex O3 induces mitochondrial dysfunction and upregulates the pro-apoptotic protein Noxa, ultimately leading to severe DNA damage. The upregulation of Phosphorylated histone protein H2AX (γ-H2AX) expression is clear evidence. In addition, O3 inhibits the expression of RAD51 protein and prevents DNA damage repair, thus overcoming drug resistance. This strategy of combining bioactive molecules cannabidiol with platinum drugs to improve therapeutic efficacy and overcome drug resistance has been proven to be very effective and deserves further investigation.
Collapse
Affiliation(s)
- Tangli Wei
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China
| | - Lihua Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China
| | - Pengmin Shi
- Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Changli Wang
- Faculty of Foreign Languages and Cultures, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China
| | - Yusheng Peng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China
| | - Jing Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China
| | - Xiali Liao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China
| | - Bo Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China.
| | - Chuanzhu Gao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, PR China.
| |
Collapse
|
5
|
Gebert M, Sławski J, Kalinowski L, Collawn JF, Bartoszewski R. The Unfolded Protein Response: A Double-Edged Sword for Brain Health. Antioxidants (Basel) 2023; 12:1648. [PMID: 37627643 PMCID: PMC10451475 DOI: 10.3390/antiox12081648] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Efficient brain function requires as much as 20% of the total oxygen intake to support normal neuronal cell function. This level of oxygen usage, however, leads to the generation of free radicals, and thus can lead to oxidative stress and potentially to age-related cognitive decay and even neurodegenerative diseases. The regulation of this system requires a complex monitoring network to maintain proper oxygen homeostasis. Furthermore, the high content of mitochondria in the brain has elevated glucose demands, and thus requires a normal redox balance. Maintaining this is mediated by adaptive stress response pathways that permit cells to survive oxidative stress and to minimize cellular damage. These stress pathways rely on the proper function of the endoplasmic reticulum (ER) and the activation of the unfolded protein response (UPR), a cellular pathway responsible for normal ER function and cell survival. Interestingly, the UPR has two opposing signaling pathways, one that promotes cell survival and one that induces apoptosis. In this narrative review, we discuss the opposing roles of the UPR signaling pathways and how a better understanding of these stress pathways could potentially allow for the development of effective strategies to prevent age-related cognitive decay as well as treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-134 Gdansk, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-134 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland
| | - James F. Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
| |
Collapse
|
6
|
Bodnar-Wachtel M, Huber AL, Gorry J, Hacot S, Burlet D, Gérossier L, Guey B, Goutagny N, Bartosch B, Ballot E, Lecuelle J, Truntzer C, Ghiringhelli F, Py BF, Couté Y, Ballesta A, Lantuejoul S, Hall J, Tissier A, Petrilli V. Inflammasome-independent NLRP3 function enforces ATM activity in response to genotoxic stress. Life Sci Alliance 2023; 6:e202201494. [PMID: 36746533 PMCID: PMC9904227 DOI: 10.26508/lsa.202201494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/08/2023] Open
Abstract
NLRP3 is a pattern recognition receptor with a well-documented role in inducing inflammasome assembly in response to cellular stress. Deregulation of its activity leads to many inflammatory disorders including gouty arthritis, Alzheimer disease, and cancer. Whereas its role in the context of cancer has been mostly explored in the immune compartment, whether NLRP3 exerts functions unrelated to immunity in cancer development remains unexplored. Here, we demonstrate that NLRP3 interacts with the ATM kinase to control the activation of the DNA damage response, independently of its inflammasome activity. NLRP3 down-regulation in both broncho- and mammary human epithelial cells significantly impairs ATM pathway activation, leading to lower p53 activation, and provides cells with the ability to resist apoptosis induced by acute genotoxic stress. Interestingly, NLRP3 expression is down-regulated in non-small cell lung cancers and breast cancers, and its expression positively correlates with patient overall survival. Our findings identify a novel non-immune function for NLRP3 in maintaining genome integrity and strengthen the concept of a functional link between innate immunity and DNA damage sensing pathways to maintain cell integrity.
Collapse
Affiliation(s)
- Mélanie Bodnar-Wachtel
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Anne-Laure Huber
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Julie Gorry
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Sabine Hacot
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Delphine Burlet
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Laetitia Gérossier
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Baptiste Guey
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Nadège Goutagny
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Birke Bartosch
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Elise Ballot
- Département d'oncologie Médicale, INSERM 1231, Université de Bourgogne, Dijon, France
| | - Julie Lecuelle
- Département d'oncologie Médicale, INSERM 1231, Université de Bourgogne, Dijon, France
| | - Caroline Truntzer
- Département d'oncologie Médicale, INSERM 1231, Université de Bourgogne, Dijon, France
| | - François Ghiringhelli
- Département d'oncologie Médicale, INSERM 1231, Université de Bourgogne, Dijon, France
| | - Bénédicte F Py
- CIRI, Centre International de Recherche en Infectiologie, University Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Yohann Couté
- Université Grenoble Alpes, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Annabelle Ballesta
- INSERM and Université Paris Sud, UMRS 935, Campus CNRS, Villejuif, France & Honorary Position, University of Warwick, Coventry, UK
| | - Sylvie Lantuejoul
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
- Département de Pathologie, Pôle de Biologie et de Pathologie, Centre Hospitalier Universitaire, Inserm U823, Institut A Bonniot-Université J Fourier, Grenoble, France
| | - Janet Hall
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Agnès Tissier
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| | - Virginie Petrilli
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Département de Biopathologie, Centre Léon Bérard, Lyon, France
| |
Collapse
|
7
|
Yiang GT, Wu CC, Lu CL, Hu WC, Tsai YJ, Huang YM, Su WL, Lu KC. Endoplasmic Reticulum Stress in Elderly Patients with COVID-19: Potential of Melatonin Treatment. Viruses 2023; 15:156. [PMID: 36680196 PMCID: PMC9863214 DOI: 10.3390/v15010156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Aging processes, including immunosenescence, inflammation, inflammasome formation, genomic instability, telomeric attrition, and altered autophagy, are involved in viral infections and they may contribute to increased pathophysiological responses to the SARS-CoV-2 infection in the elderly; this poses additional risks of accelerated aging, which could be found even after recovery. Aging is associated with oxidative damage. Moreover, SARS-CoV-2 infections may increase the production of reactive oxygen species and such infections will disturb the Ca++ balance via an endoplasmic reticulum (ER) stress-mediated unfolded protein response. Although vaccine development and anti-inflammation therapy lower the severity of COVID-19, the prevalence and mortality rates are still alarming in some countries worldwide. In this review, we describe the involvement of viral proteins in activating ER stress transducers and their downstream signals and in inducing inflammation and inflammasome formation. Furthermore, we propose the potential of melatonin as an ER stress modulator, owing to its antioxidant, anti-inflammatory, and immunoregulatory effects in viral infections. Considering its strong safety profile, we suggest that additive melatonin supplementation in the elderly could be beneficial in treating COVID-19.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-Lin Lu
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei 24352, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Yi-Ju Tsai
- Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei 243, Taiwan
| | - Yiao-Mien Huang
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Wen-Lin Su
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei 24352, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| |
Collapse
|
8
|
Identification of New Molecular Biomarkers in Ovarian Cancer Using the Gene Expression Profile. J Clin Med 2022; 11:jcm11133888. [PMID: 35807169 PMCID: PMC9267752 DOI: 10.3390/jcm11133888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is a common cause of death among women worldwide. The current diagnostic and prognostic procedures available for the treatment of ovarian cancer are either not specific or are very expensive. Gene expression profiling has proved to be a very effective tool in the exploration of new molecular markers in patients with ovarian cancer, although the link between such markers and patient survival and clinical outcomes is still elusive. We are looking for genes that may function in the development and progression of ovarian cancer. The aim of our study was to evaluate the expression of selected suppressor genes (ATM, BRCA1, BRCA2), proto-oncogenes (KRAS, c-JUN, c-FOS), pro-apoptotic genes (NOXA, PUMA), genes related to chromatin remodeling (MEN1), and genes related to carcinogenesis (NOD2, CHEK2, EGFR). Tissue samples from 30 normal ovaries and 60 ovarian carcinoma tumors were provided for analysis of the gene and protein expression. Gene expression analysis was performed using the real-time PCR method. The protein concentrations from tissue homogenates were determined using the ELISA technique according to the manufacturers’ protocols. An increase in the expression level of mRNA and protein in women with ovarian cancer was observed for KRAS, c-FOS, PUMA, and EGFR. No significant changes in the transcriptional levels we observed for BRCA1, BRCA2, NOD2, or CHEK2. In conclusion, we suggest that KRAS, NOXA, PUMA, c-FOS, and c-JUN may be associated with poor prognosis in ovarian cancer.
Collapse
|
9
|
Su WL, Wu CC, Wu SFV, Lee MC, Liao MT, Lu KC, Lu CL. A Review of the Potential Effects of Melatonin in Compromised Mitochondrial Redox Activities in Elderly Patients With COVID-19. Front Nutr 2022; 9:865321. [PMID: 35795579 PMCID: PMC9251345 DOI: 10.3389/fnut.2022.865321] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/23/2022] [Indexed: 12/17/2022] Open
Abstract
Melatonin, an endogenous indoleamine, is an antioxidant and anti-inflammatory molecule widely distributed in the body. It efficiently regulates pro-inflammatory and anti-inflammatory cytokines under various pathophysiological conditions. The melatonin rhythm, which is strongly associated with oxidative lesions and mitochondrial dysfunction, is also observed during the biological process of aging. Melatonin levels decline considerably with age and are related to numerous age-related illnesses. The signs of aging, including immune aging, increased basal inflammation, mitochondrial dysfunction, significant telomeric abrasion, and disrupted autophagy, contribute to the increased severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These characteristics can worsen the pathophysiological response of the elderly to SARS-CoV-2 and pose an additional risk of accelerating biological aging even after recovery. This review explains that the death rate of coronavirus disease (COVID-19) increases with chronic diseases and age, and the decline in melatonin levels, which is closely related to the mitochondrial dysfunction in the patient, affects the virus-related death rate. Further, melatonin can enhance mitochondrial function and limit virus-related diseases. Hence, melatonin supplementation in older people may be beneficial for the treatment of COVID-19.
Collapse
Affiliation(s)
- Wen-Lin Su
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Fang Vivienne Wu
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Mei-Chen Lee
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chien-Lin Lu
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
10
|
Downregulation of CPT2 promotes proliferation and inhibits apoptosis through p53 pathway in colorectal cancer. Cell Signal 2022; 92:110267. [DOI: 10.1016/j.cellsig.2022.110267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023]
|
11
|
Coccè V, Rimoldi I, Facchetti G, Ciusani E, Alessandri G, Signorini L, Sisto F, Giannì A, Paino F, Pessina A. In Vitro Activity of Monofunctional Pt-II Complex Based on 8-Aminoquinoline against Human Glioblastoma. Pharmaceutics 2021; 13:pharmaceutics13122101. [PMID: 34959382 PMCID: PMC8704014 DOI: 10.3390/pharmaceutics13122101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/18/2022] Open
Abstract
A new cationic Pt(II) complex bearing 8-aminoquinoline as chelating ligand (called Pt-8AQ) was evaluated against two human carcinomas, one mesothelioma, and three glioblastoma cell lines. The in vitro comparison to the clinically approved CisPt showed a minor activity of Pt-8AQ against carcinoma and mesothelioma, whereas a significant activity of Pt-8AQ was observed on the proliferation of the three glioblastoma cell lines (U87-MG IC50 = 3.68 ± 0.69 µM; U373-MG IC50 = 11.53 ± 0.16 µM; U138-MG IC50 = 8.05 ± 0.23 µM) that was higher than that observed with the clinically approved CisPt (U87-MG IC50 = 7.27 + 1.80 µM; U373-MG IC50 = 22.69 ± 0.05 µM; U138-MG IC50 = 32.1 ± 4.44 µM). Cell cycle analysis proved that Pt-8AQ significantly affected the cell cycle pattern by increasing the apoptotic cells represented by the sub G0/G1 region related with a downregulation of p53 and Bcl-2. Moreover, an NMR investigation of Pt-8AQ interaction with 9-EtG, GSH, and Mets7 excluded DNA as the main target, suggesting a novel mechanism of action. Our study demonstrated the high stability of Pt-8AQ after incubation at 37 °C and a significant antineoplastic activity on glioblastomas. These features also make Pt-8AQ a good candidate for developing a new selective advanced cell chemotherapy approach in combination with MSCs.
Collapse
Affiliation(s)
- Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Isabella Rimoldi
- Department of Pharmaceutical Science, University of Milan, Via Golgi 19, 20133 Milan, Italy;
| | - Giorgio Facchetti
- Department of Pharmaceutical Science, University of Milan, Via Golgi 19, 20133 Milan, Italy;
- Correspondence: (G.F.); (A.P.)
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, Fondazione IRCCS Neurological Institute Carlo Besta, 20133 Milan, Italy;
| | - Giulio Alessandri
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy;
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Aldo Giannì
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
- Maxillo-Facial and Dental Unit, Fondazione Ca’ Granda IRCCS Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Paino
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
- Correspondence: (G.F.); (A.P.)
| |
Collapse
|
12
|
Catlin JP, Marziali LN, Rein B, Yan Z, Feltri ML, Schaner Tooley CE. Age-related neurodegeneration and cognitive impairments of NRMT1 knockout mice are preceded by misregulation of RB and abnormal neural stem cell development. Cell Death Dis 2021; 12:1014. [PMID: 34711807 PMCID: PMC8553844 DOI: 10.1038/s41419-021-04316-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023]
Abstract
N-terminal methylation is an important posttranslational modification that regulates protein/DNA interactions and plays a role in many cellular processes, including DNA damage repair, mitosis, and transcriptional regulation. Our generation of a constitutive knockout mouse for the N-terminal methyltransferase NRMT1 demonstrated its loss results in severe developmental abnormalities and premature aging phenotypes. As premature aging is often accompanied by neurodegeneration, we more specifically examined how NRMT1 loss affects neural pathology and cognitive behaviors. Here we find that Nrmt1-/- mice exhibit postnatal enlargement of the lateral ventricles, age-dependent striatal and hippocampal neurodegeneration, memory impairments, and hyperactivity. These morphological and behavior abnormalities are preceded by alterations in neural stem cell (NSC) development. Early expansion and differentiation of the quiescent NSC pool in Nrmt1-/- mice is followed by its subsequent depletion and many of the resulting neurons remain in the cell cycle and ultimately undergo apoptosis. These cell cycle phenotypes are reminiscent to those seen with loss of the NRMT1 target retinoblastoma protein (RB). Accordingly, we find misregulation of RB phosphorylation and degradation in Nrmt1-/- mice, and significant de-repression of RB target genes involved in cell cycle. We also identify novel de-repression of Noxa, an RB target gene that promotes apoptosis. These data identify Nα-methylation as a novel regulatory modification of RB transcriptional repression during neurogenesis and indicate that NRMT1 and RB work together to promote NSC quiescence and prevent neuronal apoptosis.
Collapse
Affiliation(s)
- James P Catlin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Leandro N Marziali
- Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Benjamin Rein
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - M Laura Feltri
- Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Christine E Schaner Tooley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
13
|
Toriyama K, Takano N, Kokuba H, Kazama H, Moriya S, Hiramoto M, Abe S, Miyazawa K. Azithromycin enhances the cytotoxicity of DNA-damaging drugs via lysosomal membrane permeabilization in lung cancer cells. Cancer Sci 2021; 112:3324-3337. [PMID: 34051014 PMCID: PMC8353917 DOI: 10.1111/cas.14992] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer cells use autophagy for growth, survival, and cytoprotection from chemotherapy. Therefore, autophagy inhibitors appear to be good candidates for cancer treatment. Our group previously reported that macrolide antibiotics, especially azithromycin (AZM), have potent autophagy inhibitory effects, and combination treatment with tyrosine kinase inhibitors or proteasome inhibitors enhances their anti-cancer activity. In this study, we evaluated the effect of combination therapy with DNA-damaging drugs and AZM in non-small-cell lung cancer (NSCLC) cells. We found that the cytotoxic activities of DNA-damaging drugs, such as doxorubicin (DOX), etoposide, and carboplatin, were enhanced in the presence of AZM in NSCLC cell lines, whereas AZM alone exhibited almost no cytotoxicity. This enhanced cell death was dependent on wild-type-p53 status and autophagosome-forming ability because TP53 knockout (KO) and ATG5-KO cells attenuated AZM-enhanced cytotoxicity. DOX treatment upregulated lysosomal biogenesis by activating TFEB and led to lysosomal membrane damage as assessed by galectin 3 puncta assay and cytoplasmic leakage of lysosomal enzymes. In contrast, AZM treatment blocked autophagy, which resulted in the accumulation of lysosomes/autolysosomes. Thus, the effects of DOX and AZM were integrated into the marked increase in damaged lysosomes/autolysosomes, leading to prominent lysosomal membrane permeabilization (LMP) for apoptosis induction. Our data suggest that concomitant treatment with DNA-damaging drugs and AZM is a promising strategy for NSCLC treatment via pronounced LMP induction.
Collapse
Affiliation(s)
- Kazutoshi Toriyama
- Department of Respiratory Medicine, Tokyo Medical University Hospital, Tokyo, Japan
| | - Naoharu Takano
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Hiroko Kokuba
- Laboratory of Electron Microscopy, Tokyo Medical University, Tokyo, Japan
| | - Hiromi Kazama
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Shota Moriya
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Shinji Abe
- Department of Respiratory Medicine, Tokyo Medical University Hospital, Tokyo, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
14
|
Muyskens JB, Winbush A, Foote DM, Turnbull DW, Dreyer HC. Essential amino acid supplementation alters the p53 transcriptional response and cytokine gene expression following total knee arthroplasty. J Appl Physiol (1985) 2020; 129:980-991. [PMID: 32881622 DOI: 10.1152/japplphysiol.00022.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Reducing muscle atrophy following orthopedic surgery is critical during the postoperative period. Our previous work in patients who underwent total knee arthroplasty (TKA) showed that the vast majority of atrophy occurs within 2 wk following surgery and that essential amino acid (EAA) supplementation attenuates this atrophy. We used RNA-sequencing (RNA-seq) to identify genes associated with atrophy after TKA with and without EAAs. Analysis of overrepresented gene-ontology terms revealed that p53 signaling and the cytokine-cytokine receptor pathways were highly upregulated after TKA. Relative to the placebo group, the EAA group had altered expression of p53 regulators such as MDM2. This altered expression may account for differences between groups in timing of upregulation of some p53 targets such as apoptosis genes, and may account for the reduction in muscle loss in the subjects receiving EAAs. Furthermore, we observed altered expression of a large number of cytokine-signaling genes including TNFRSF12A, which plays a critical role in muscle atrophy, myogenesis, fibrosis, and the noncanonical NF-κB pathway.NEW & NOTEWORTHY Total knee arthroplasty is the most frequently performed inpatient surgical procedure for those over 45 yr in the United States. Following surgery, patients lose a large amount of muscle, which impacts functional mobility. Previously, our laboratory found that supplementing patients' diets with essential amino acids (EAAs) reduces postsurgical muscle loss. Here, our goal was to characterize the transcriptional changes associated with surgery with and without EAA supplementation to uncover the underlying mechanisms by which EAAs attenuate this muscle loss.
Collapse
Affiliation(s)
| | - Ari Winbush
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon
| | - Douglas M Foote
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Douglas W Turnbull
- Genomics and Cell Characterization Core Facility, University of Oregon, Eugene, Oregon
| | - Hans C Dreyer
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| |
Collapse
|
15
|
Abstract
Within the past several decades, the emergence of new viral diseases with severe health complications and mortality is evidence of an age-dependent, compromised bodily response to abrupt stress with concomitantly reduced immunity. The new severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, causes coronavirus disease 2019 (COVID-19). It has increased morbidity and mortality in persons with underlying chronic diseases and those with a compromised immune system regardless of age and in older adults who are more likely to have these conditions. While SARS-CoV-2 is highly virulent, there is variability in the severity of the disease and its complications in humans. Severe pneumonia, acute respiratory distress syndrome, lung fibrosis, cardiovascular events, acute kidney injury, stroke, hospitalization, and mortality have been reported that result from pathogen-host interactions. Hallmarks of aging, interacting with one another, have been proposed to influence health span in older adults, possibly via mechanisms regulating the immune system. Here, we review the potential roles of the hallmarks of aging, coupled with host-coronavirus interactions. Of these hallmarks, we focused on those that directly or indirectly interact with viral infections, including immunosenescence, inflammation and inflammasomes, adaptive immunosenescence, genomic instability, mitochondrial dysfunction, epigenetic alterations, telomere attrition, and impaired autophagy. These hallmarks likely contribute to the increased pathophysiological responses to SARS-CoV-2 among older adults and may play roles as an additive risk of accelerated biological aging even after recovery. We also briefly discuss the role of antiaging drug candidates that require paramount attention in COVID-19 research.
Collapse
Affiliation(s)
- Shabnam Salimi
- Department of Epidemiology and Public Health, Division of Gerontology, University of Maryland School of Medicine, Baltimore
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
16
|
Mallick DJ, Eastman A. AT101 [(-)-Gossypol] Selectively Inhibits MCL1 and Sensitizes Carcinoma to BH3 Mimetics by Inducing and Stabilizing NOXA. Cancers (Basel) 2020; 12:E2298. [PMID: 32824203 PMCID: PMC7464284 DOI: 10.3390/cancers12082298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/04/2020] [Accepted: 08/12/2020] [Indexed: 12/17/2022] Open
Abstract
Anti-apoptotic BCL2 proteins are important targets for cancer therapy as cancers depend on their activity for survival. Direct inhibitors of MCL1 have entered clinical trials, although their efficacy may be limited by toxicity. An alternative approach may be to induce the pro-apoptotic protein NOXA which selectively inhibits MCL1 in cells. Many compounds originally proposed as inhibitors of the BCL2 family were subsequently found to induce the pro-apoptotic protein NOXA through the unfolded protein response. In the present study, we compared various putative BH3 mimetics across a panel of carcinoma cell lines and measured expression of NOXA protein and mRNA, as well as the kinetics of NOXA induction. We found that AT101 [(-)-gossypol] induces high levels of NOXA in carcinoma cell lines yet cells survive. When combined with an appropriate BCL2 or BCL-XL inhibitor, NOXA-dependent sensitization occurs. NOXA protein continues to accumulate for many hours after AT101 is removed, providing a window for administering these combinations. As MCL1 promotes drug resistance and overall survival, we propose that NOXA induction is an alternative therapeutic strategy to target MCL1 and either kill cancer cells that are dependent on MCL1 or sensitize cancer cells to other BCL2 inhibitors.
Collapse
Affiliation(s)
- David J. Mallick
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
| | - Alan Eastman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
- Department of Molecular and Systems Biology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| |
Collapse
|
17
|
Núñez-Vázquez S, Sánchez-Vera I, Saura-Esteller J, Cosialls AM, Noisier AFM, Albericio F, Lavilla R, Pons G, Iglesias-Serret D, Gil J. NOXA upregulation by the prohibitin-binding compound fluorizoline is transcriptionally regulated by integrated stress response-induced ATF3 and ATF4. FEBS J 2020; 288:1271-1285. [PMID: 32648994 DOI: 10.1111/febs.15480] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/17/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023]
Abstract
Fluorizoline is a new synthetic molecule that induces p53-independent apoptosis, in several tumor cell lines and in primary leukemia cells, by selectively targeting prohibitins (PHBs). In this study, we describe how fluorizoline induces BCL-2 homology 3-only protein NOXA, without modulating the protein levels of anti-apoptotic B-cell lymphoma-2 (BCL-2) family members prior to caspase activation, as well as how it synergizes with the BCL-2 and BCL-XL inhibitor ABT-737 to induce apoptosis. Interestingly, fluorizolinetreatment triggers the activation of the integrated stress response (ISR) in HeLa and HAP1 cells, with increased eukaryotic translation initiation factor 2α phosphorylation, and induction of ATF3, ATF4, and CHOP. Moreover, PHB downregulation induces similar ISR activation and apoptosis as with fluorizoline treatment. In addition, we studied the essential role of the pro-apoptotic protein NOXA in fluorizoline-induced apoptosis and we describe its mechanism of induction in HeLa and HAP1 cells. Moreover, we identified ATF3 and ATF4 as the transcription factors that bind to NOXA promoter upon fluorizoline treatment. Furthermore, using ATF3 and ATF4 CRISPR HeLa and HAP1 cells, we confirmed that both factors mediate the induction of NOXA and apoptosis by fluorizoline. In conclusion, fluorizoline treatment triggers the activation of the ISR that results in the induction of ATF3 and ATF4, important regulators of NOXA transcription in fluorizoline-induced apoptosis.
Collapse
Affiliation(s)
- Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Ismael Sánchez-Vera
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - José Saura-Esteller
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Ana M Cosialls
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Anaïs F M Noisier
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences and Institute of Medicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Fernando Albericio
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona, Barcelona, Spain
| | - Rodolfo Lavilla
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences and Institute of Medicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Daniel Iglesias-Serret
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain.,Facultat de Medicina, Universitat de Vic - Universitat Central de Catalunya (UVic-UCC), Vic, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Strasser A, Vaux DL. Cell Death in the Origin and Treatment of Cancer. Mol Cell 2020; 78:1045-1054. [PMID: 32516599 DOI: 10.1016/j.molcel.2020.05.014] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
Cell death, or, more specifically, cell suicide, is a process of fundamental importance to human health. Throughout our lives, over a million cells are produced every second. When organismal growth has stopped, to balance cell division, a similar number of cells must be removed. This is achieved by activation of molecular mechanisms that have evolved so that cells can destroy themselves. The first clues regarding the nature of one of these mechanisms came from studying genes associated with cancer, in particular the gene for BCL-2. Subsequent studies revealed that mutations or other defects that inhibit cell death allow cells to accumulate, prevent removal of cells with damaged DNA, and increase the resistance of malignant cells to chemotherapy. Knowledge of this mechanism has allowed development of drugs that kill cancer cells by directly activating the cell death machinery and by synergizing with conventional chemotherapy as well as targeted agents to achieve improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3052, Australia.
| | - David L Vaux
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3052, Australia.
| |
Collapse
|
19
|
Abo Elwafa R, Abd Elrahman A, Ghallab O. Long intergenic non-coding RNA-p21 is associated with poor prognosis in chronic lymphocytic leukemia. Clin Transl Oncol 2020; 23:92-99. [PMID: 32468342 DOI: 10.1007/s12094-020-02398-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/12/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Long non-coding RNAs (LncRNAs) are RNA transcripts longer than 200 nucleotides. They are new players in transcriptional regulation and cancer research. LincRNA-p21 is a p53-regulated lncRNA involved in the p53 transcriptional network. It has an important role in regulating cellular proliferation and apoptosis. Chronic lymphocytic leukemia is derived by a typical defect in apoptosis and characterized by clonal proliferation and accumulation of mature B cells. The aim of the present study was to assess the expression pattern of the lincRNA-p21 and investigate its potential role as a new prognostic marker in CLL. METHODS The study was conducted on 80 newly diagnosed CLL patients and 80 age- and sex-matched controls. The analysis of LincRNA-p21 and the p53 downstream proapoptotic target genes (MDM2, PUMA, BAX, and NOXA) was performed by real-time PCR. The cytogenetic abrasions and expression of ZAP70 and CD38 were detected by FISH and Flow cytometry, respectively. RESULTS LincRNA-p21 was significantly downregulated in CLL patients compared to controls. The downstream proapoptotic targets were significantly downregulated in CLL patients and positively correlated with lincRNA-p21. Low expression of lincRNA-p21 was associated with poor prognostic markers (advanced stages of CLL, del 17p13, ZAP70, and CD38 expression), failure of complete remission, shorter progression free survival, and overall survival. Low lincRNA-p21 expression was independently prognostic for shorter time to treatment. CONCLUSION Low expression of lincRNA-p21 demarcates a more aggressive form of CLL with poor prognosis. Therefore, it could be considered as a new prognostic marker to predict disease outcome in CLL.
Collapse
Affiliation(s)
- R Abo Elwafa
- Clinical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - A Abd Elrahman
- Internal Medicine Department (Hematology Unit), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - O Ghallab
- Internal Medicine Department (Hematology Unit), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
20
|
Króliczewski J, Bartoszewska S, Dudkowska M, Janiszewska D, Biernatowska A, Crossman DK, Krzymiński K, Wysocka M, Romanowska A, Baginski M, Markuszewski M, Ochocka RJ, Collawn JF, Sikorski AF, Sikora E, Bartoszewski R. Utilizing Genome-Wide mRNA Profiling to Identify the Cytotoxic Chemotherapeutic Mechanism of Triazoloacridone C-1305 as Direct Microtubule Stabilization. Cancers (Basel) 2020; 12:cancers12040864. [PMID: 32252403 PMCID: PMC7226417 DOI: 10.3390/cancers12040864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/21/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Rational drug design and in vitro pharmacology profiling constitute the gold standard in drug development pipelines. Problems arise, however, because this process is often difficult due to limited information regarding the complete identification of a molecule’s biological activities. The increasing affordability of genome-wide next-generation technologies now provides an excellent opportunity to understand a compound’s diverse effects on gene regulation. Here, we used an unbiased approach in lung and colon cancer cell lines to identify the early transcriptomic signatures of C-1305 cytotoxicity that highlight the novel pathways responsible for its biological activity. Our results demonstrate that C-1305 promotes direct microtubule stabilization as a part of its mechanism of action that leads to apoptosis. Furthermore, we show that C-1305 promotes G2 cell cycle arrest by modulating gene expression. The results indicate that C-1305 is the first microtubule stabilizing agent that also is a topoisomerase II inhibitor. This study provides a novel approach and methodology for delineating the antitumor mechanisms of other putative anticancer drug candidates.
Collapse
Affiliation(s)
- Jarosław Króliczewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, 80-416 Gdansk, Poland; (J.K.); (R.J.O.)
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland;
| | - Magdalena Dudkowska
- Laboratory of the Molecular Bases of Ageing, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland (D.J.); (E.S.)
| | - Dorota Janiszewska
- Laboratory of the Molecular Bases of Ageing, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland (D.J.); (E.S.)
| | - Agnieszka Biernatowska
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw Poland;
| | - David K. Crossman
- Department of Genetics, UAB Genomics Core Facility, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Karol Krzymiński
- Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (K.K.); (M.W.); (A.R.)
| | - Małgorzata Wysocka
- Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (K.K.); (M.W.); (A.R.)
| | - Anna Romanowska
- Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (K.K.); (M.W.); (A.R.)
| | - Maciej Baginski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland;
| | - Michal Markuszewski
- Department of Biopharmacy and Pharmacodynamics, Medical University of Gdansk, 80-416 Gdansk, Poland;
| | - Renata J. Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, 80-416 Gdansk, Poland; (J.K.); (R.J.O.)
| | - James F. Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | | | - Ewa Sikora
- Laboratory of the Molecular Bases of Ageing, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland (D.J.); (E.S.)
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, 80-416 Gdansk, Poland; (J.K.); (R.J.O.)
- Correspondence: ; Tel.: +48-58-349-32-14; Fax: +48-58-349-32-11
| |
Collapse
|
21
|
Poofery J, Sripanidkulchai B, Banjerdpongchai R. Extracts of Bridelia ovata and Croton oblongifolius induce apoptosis in human MDA‑MB‑231 breast cancer cells via oxidative stress and mitochondrial pathways. Int J Oncol 2020; 56:969-985. [PMID: 32319560 DOI: 10.3892/ijo.2020.4973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/12/2019] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is the most common type of cancer and is also the second leading cause of cancer‑associated death in women worldwide. Thus, there is an urgent requirement for the development of effective treatments for this disease. Bridelia ovata and Croton oblongifolius are herbs used in Thai traditional medicine that have been used to treat various health problems; B. ovata has traditionally been used as a purgative, an antipyretic, a leukorrhea treatment and as a birth control herb. C. oblongifolius has been used to increase breast milk production, for post‑partum care (where it is used as a hot bath herb), and as a treatment for flat worms and dysmenorrhea. However, there is little research investigating the anticancer properties of these herbs. The present study aimed to investigate the anticancer properties of crude ethyl acetate extracts of B. ovata (BEA) and C. oblongifolius (CEA) in order to explore their underlying mechanisms in breast cancer cell death. The phytoconstituents of the crude extracts of BEA and CEA were studied using gas chromatography‑mass spectrometry (GC‑MS). GC‑MS analysis showed that the primary compound in BEA is friedelan‑3‑one, and kaur‑16‑en‑18‑oic acid in CEA. Cytotoxicity was investigated using an MTT assay, both BEA and CEA showed greater toxicity against MDA‑MB‑231 breast cancer cells compared with their effect on MCF10A normal epithelial mammary cells. BEA and CEA exerted various effects, including inducing apoptotic cell death, reducing mitochondrial transmembrane potential, increasing the levels of intracellular ROS, activating caspases, upregulating pro‑apoptotic and downregulating anti‑apoptotic genes and proteins. BEA and CEA were shown to have anticancer activity against breast cancer cells and induce apoptosis in these cells via a mitochondrial pathway and oxidative stress.
Collapse
Affiliation(s)
- Juthathip Poofery
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Bungorn Sripanidkulchai
- Center for Research and Development of Herbal Health Products, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ratana Banjerdpongchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
22
|
Vallese F, Barazzuol L, Maso L, Brini M, Calì T. ER-Mitochondria Calcium Transfer, Organelle Contacts and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:719-746. [PMID: 31646532 DOI: 10.1007/978-3-030-12457-1_29] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is generally accepted that interorganellar contacts are central to the control of cellular physiology. Virtually, any intracellular organelle can come into proximity with each other and, by establishing physical protein-mediated contacts within a selected fraction of the membrane surface, novel specific functions are acquired. Endoplasmic reticulum (ER) contacts with mitochondria are among the best studied and have a major role in Ca2+ and lipid transfer, signaling, and membrane dynamics.Their functional (and structural) diversity, their dynamic nature as well as the growing number of new players involved in the tethering concurred to make their monitoring difficult especially in living cells. This review focuses on the most established examples of tethers/modulators of the ER-mitochondria interface and on the roles of these contacts in health and disease by specifically dissecting how Ca2+ transfer occurs and how mishandling eventually leads to disease. Additional functions of the ER-mitochondria interface and an overview of the currently available methods to measure/quantify the ER-mitochondria interface will also be discussed.
Collapse
Affiliation(s)
- Francesca Vallese
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Lucia Barazzuol
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Lorenzo Maso
- Department of Biology, University of Padua, Padua, Italy
| | - Marisa Brini
- Department of Biology, University of Padua, Padua, Italy.
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy. .,Padua Neuroscience Center (PNC), Padua, Italy.
| |
Collapse
|
23
|
Silencing lncRNA LOC101928963 Inhibits Proliferation and Promotes Apoptosis in Spinal Cord Glioma Cells by Binding to PMAIP1. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:485-495. [PMID: 31670198 PMCID: PMC6838552 DOI: 10.1016/j.omtn.2019.07.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/24/2019] [Indexed: 12/16/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been widely highlighted due to their involvement in various types of cancers, including glioma; however, the exact mechanism and function by which they operate in regard to spinal cord glioma remain poorly understood. LOC101928963 was screened out for its differential expression in spinal cord glioma by microarray analysis. Therefore, this study was conducted to investigate the modulatory effects of LOC101928963 on spinal cord glioma by binding to phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1). The expression of LOC101928963 and LOC101928963 was characterized in spinal cord glioma tissues, and their interaction was examined by dual-luciferase reporter gene assay. Cells with LOC101928963 that exhibited elevated or suppressed levels of PMAIP1 were established to substantiate the mechanism between LOC101928963 and PMAIP1. qRT-PCR and western blot methods were subsequently applied to determine the expression of cell-proliferation- and apoptosis-related genes in response to the alterations of LOC101928963 and PMAIP1. Glioma cell proliferation and apoptosis were assessed by MTT assay and flow cytometry. Decreased cell apoptosis and PMAIP1 expression, as well as overexpressed LOC101928963, were exhibited among spinal cord glioma tissues. LOC101928963 overexpression was observed to promote cell proliferation and cell-cycle entry and inhibit the process of apoptosis. PMAIP1, a target of LOC101928963, displayed a downregulated level following the elevation of LOC101928963. The present results strongly emphasize the neutralization effect of PMAIP1 overexpression on spinal cord glioma progression induced by the overexpression of LOC101928963. The data obtained during the study highlighted the inhibitory role of LOC101928963 silencing in spinal cord glioma through the increase in PMAIP1, which suggests a potential target in the treatment of spinal cord glioma.
Collapse
|
24
|
Jeong S, Yun HK, Jeong YA, Jo MJ, Kang SH, Kim JL, Kim DY, Park SH, Kim BR, Na YJ, Lee SI, Kim HD, Kim DH, Oh SC, Lee DH. Cannabidiol-induced apoptosis is mediated by activation of Noxa in human colorectal cancer cells. Cancer Lett 2019; 447:12-23. [PMID: 30660647 DOI: 10.1016/j.canlet.2019.01.011] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/21/2018] [Accepted: 01/11/2019] [Indexed: 12/21/2022]
Abstract
Cannabidiol (CBD), one of the compounds present in the marijuana plant, has anti-tumor properties, but its mechanism is not well known. This study aimed to evaluate the apoptotic action of CBD in colorectal cancer (CRC) cells, and focused on its effects on the novel pro-apoptotic Noxa-reactive oxygen species (ROS) signaling pathway. CBD experiments were performed using the CRC cell lines HCT116 and DLD-1. CBD induced apoptosis by regulating many pro- and anti-apoptotic proteins, of which Noxa showed significantly higher expression. To understand the relationship between Noxa and CBD-induced apoptosis, Noxa levels were downregulated using siRNA, and the expression of apoptosis markers decreased. After ROS production was blocked, the level of Noxa also decreased, suggesting that ROS is involved in the regulation of Noxa, which along with ROS is a well-known pro-apoptotic signaling agents. As a result, CBD induced apoptosis in a Noxa-and-ROS-dependent manner. Taken together, the results obtained in this study re-demonstrated the effects of CBD treatment in vivo, thus confirming its role as a novel, reliable anticancer drug.
Collapse
Affiliation(s)
- Soyeon Jeong
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hye Kyeong Yun
- Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea
| | - Yoon A Jeong
- Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea
| | - Min Jee Jo
- Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea
| | - Sang Hee Kang
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jung Lim Kim
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Dae Yeong Kim
- Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea
| | - Seong Hye Park
- Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea
| | - Bo Ram Kim
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Yoo Jin Na
- Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea
| | - Sun Il Lee
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Han Do Kim
- Kaiyon Bio Tech Co., Ltd, 226 Gamasan-Ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Dae Hyun Kim
- Kaiyon Bio Tech Co., Ltd, 226 Gamasan-Ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Sang Cheul Oh
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea; Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea.
| | - Dae-Hee Lee
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea; Graduate School of Medicine, College of Medicine, Korea University, Seoul, 08308, Republic of Korea.
| |
Collapse
|
25
|
Pinto BAS, França LM, Laurindo FRM, Paes AMDA. Unfolded Protein Response: Cause or Consequence of Lipid and Lipoprotein Metabolism Disturbances? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1127:67-82. [DOI: 10.1007/978-3-030-11488-6_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
26
|
Fedorova O, Petukhov A, Daks A, Shuvalov O, Leonova T, Vasileva E, Aksenov N, Melino G, Barlev NA. Orphan receptor NR4A3 is a novel target of p53 that contributes to apoptosis. Oncogene 2018; 38:2108-2122. [PMID: 30455429 DOI: 10.1038/s41388-018-0566-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 10/02/2018] [Accepted: 10/06/2018] [Indexed: 11/09/2022]
Abstract
Major tumor suppressor and transcription factor p53 coordinates expression of many genes hence affecting critical cellular functions including cell cycle, senescence, and apoptosis. The NR4A family of orphan receptors (NR4A1-3) belongs to the superfamily of nuclear receptors. They regulate genes involved in proliferation, cell migration, and apoptosis. In this study, we report an identification of NR4A3 as a direct transcriptional target of p53. Using various techniques, we showed that p53 directly bound the promoter of NR4A3 gene and induced its transcription. Functionally, over-expression of NR4A3 attenuated proliferation of cancer cells and promoted apoptosis by augmenting the expression of pro-apoptotic genes, PUMA and Bax. Knockdown of NR4A3 reversed these phenotypes. Importantly, NR4A3 exhibited tumor suppressive functions both in p53-dependent and independent manner. In addition, NR4A3 physically interacted with an anti-apoptotic Bcl-2 protein hence sequestering it from blunting apoptosis. These observations were corroborated by the bioinformatics analysis, which demonstrated a correlation between high levels of NR4A3 expression and better survival of breast and lung cancer patients. Collectively, our studies revealed a novel transcriptional target of p53, NR4A3, which triggers apoptosis and thus likely has a tumor suppressive role in breast and lung cancers.
Collapse
Affiliation(s)
- Olga Fedorova
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064
| | - Alexey Petukhov
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064.,Almazov National Medical Research Centre, St. Petersburg, Russia, 197341
| | - Alexandra Daks
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064
| | - Oleg Shuvalov
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064
| | - Tatyana Leonova
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064
| | - Elena Vasileva
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064
| | - Nikolai Aksenov
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064
| | | | - Nikolai A Barlev
- Gene Expression Program, Institute of Cytology, Saint-Petersburg, Russia, 194064. .,Moscow Institute of Technology and Physics, Dolgoprudny, Moscow Region, Russia, 141700.
| |
Collapse
|
27
|
Hlosrichok A, Sumkhemthong S, Sritularak B, Chanvorachote P, Chaotham C. A bibenzyl from Dendrobium ellipsophyllum induces apoptosis in human lung cancer cells. J Nat Med 2018; 72:615-625. [PMID: 29488156 DOI: 10.1007/s11418-018-1186-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/31/2018] [Indexed: 12/22/2022]
Abstract
Failure of current chemotherapeutic drugs leads to the recurrence of tumor pathology and mortality in lung cancer patients. This study aimed to evaluate the anticancer activity and related mechanisms of 4,5,4'-trihydroxy-3,3'-dimethoxybibenzyl (TDB), a bibenzyl extracted from Dendrobium ellipsophyllum Tang and Wang, in human lung cancer cells. Cytotoxicity of TDB (0-300 µM) in different types of human lung cancer cells (H460, H292 and H23) and human dermal papilla cells (DPCs) was evaluated via MTT viability assay. Selective anticancer activity of TDB against human lung cancer cells was demonstrated with a high IC50 (approximately > 300 µM) in DPCs, while IC50 in human lung cancer H460, H292 and H23 cells was approximately 100 ± 5.18, 100 ± 8.73 and 188.89 ± 8.30 µM, respectively. After treatment with 50 µM of TDB for 24 h, flow cytometry analysis revealed the significant increase of early and late apoptosis with absence of necrosis cell death in human lung cancer cells. The up-regulation of p53, a tumor-suppressor protein, was elucidated in human lung cancer cells treated with 10-50 µM of TDB. Alteration to down-stream signaling of p53 including activation of pro-apoptosis protein (Bcl-2-associated X protein; Bax), reduction of anti-apoptosis (B cell lymphoma 2; Bcl-2 and myeloid cell leukemia 1; Mcl-1) and suppression on protein kinase B (Akt) survival pathway were notified in TDB-treated lung cancer cells. The information obtained from this study strengthens the potential development of TDB as an anticancer compound with a favorable human safety profile and high efficacy.
Collapse
Affiliation(s)
- Anirut Hlosrichok
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Somruethai Sumkhemthong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Boonchoo Sritularak
- Departments of Pharmacognosy and Pharmaceutical Botany, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand. .,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
28
|
Davaadelger B, Duan L, Perez RE, Gitelis S, Maki CG. Crosstalk between the IGF-1R/AKT/mTORC1 pathway and the tumor suppressors p53 and p27 determines cisplatin sensitivity and limits the effectiveness of an IGF-1R pathway inhibitor. Oncotarget 2018; 7:27511-26. [PMID: 27050276 PMCID: PMC5053668 DOI: 10.18632/oncotarget.8484] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/18/2016] [Indexed: 12/21/2022] Open
Abstract
The insulin-like growth factor-1 receptor (IGF-1R) signaling pathway is aberrantly activated in multiple cancers and can promote proliferation and chemotherapy resistance. Multiple IGF-1R inhibitors have been developed as potential therapeutics. However, these inhibitors have failed to increase patient survival when given alone or in combination with chemotherapy agents. The reason(s) for the disappointing clinical effect of these inhibitors is not fully understood. Cisplatin (CP) activated the IGF-1R/AKT/mTORC1 pathway and stabilized p53 in osteosarcoma (OS) cells. p53 knockdown reduced IGF-1R/AKT/mTORC1 activation by CP, and IGF-1R inhibition reduced the accumulation of p53. These data demonstrate positive crosstalk between p53 and the IGF-1R/AKT/mTORC1 pathway in response to CP. Further studies showed the effect of IGF-1R inhibition on CP response is dependent on p53 status. In p53 wild-type cells treated with CP, IGF-1R inhibition increased p53s apoptotic function but reduced p53-dependent senescence, and had no effect on long term survival. In contrast, in p53-null/knockdown cells, IGF-1R inhibition reduced apoptosis in response to CP and increased long term survival. These effects were due to p27 since IGF-1R inhibition stabilized p27 in CP-treated cells, and p27 depletion restored apoptosis and reduced long term survival. Together, the results demonstrate 1) p53 expression determines the effect of IGF-1R inhibition on cancer cell CP response, and 2) crosstalk between the IGF-1R/AKT/mTORC1 pathway and p53 and p27 can reduce cancer cell responsiveness to chemotherapy and may ultimately limit the effectiveness of IGF-1R pathway inhibitors in the clinic.
Collapse
Affiliation(s)
- Batzaya Davaadelger
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Lei Duan
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Ricardo E Perez
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Steven Gitelis
- Section of Orthopedic Oncology, Department of Orthopedic Surgery, Rush University, Medical Center, Chicago, IL, USA
| | - Carl G Maki
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
29
|
Facchini G, Rossetti S, Cavaliere C, D'Aniello C, Di Franco R, Iovane G, Grimaldi G, Piscitelli R, Muto P, Botti G, Perdonà S, Veneziani BM, Berretta M, Montanari M. Exploring the molecular aspects associated with testicular germ cell tumors: a review. Oncotarget 2017; 9:1365-1379. [PMID: 29416701 PMCID: PMC5787445 DOI: 10.18632/oncotarget.22373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/05/2017] [Indexed: 11/25/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) represent the most common solid tumors affecting young men. They constitute a distinct entity because of their embryonic origin and their unique biological behavior. Recent preclinical data regarding biological signaling machinery as well as genetic and epigenetic mechanisms associated with molecular patterns of tumors have contribute to explain the pathogenesis and the differentiation of TGCTs and to understand the mechanisms responsible for the development of resistance to treatment. In this review, we discuss the main genetic and epigenetic events associated with TGCTs development in order to better define their role in the pathogenesis of these tumors and in cisplatin-acquired resistance.
Collapse
Affiliation(s)
- Gaetano Facchini
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Sabrina Rossetti
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Carla Cavaliere
- Medical Oncology Unit, ASL NA 3 SUD, Ospedali Riuniti Area Nolana, Nola, Italy
| | - Carmine D'Aniello
- Division of Medical Oncology, A.O.R.N. dei COLLI "Ospedali Monaldi-Cotugno-CTO", Naples, Italy
| | - Rossella Di Franco
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale", IRCCS, Naples, Italy
| | - Gelsomina Iovane
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Giovanni Grimaldi
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Raffaele Piscitelli
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Paolo Muto
- Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale", IRCCS, Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy.,Scientific Management, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Sisto Perdonà
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, Aviano, Italy
| | - Micaela Montanari
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy.,Department of Biology, College of Science and Technology, Temple University, Philadelphia, USA
| |
Collapse
|
30
|
Guikema JE, Amiot M, Eldering E. Exploiting the pro-apoptotic function of NOXA as a therapeutic modality in cancer. Expert Opin Ther Targets 2017; 21:767-779. [PMID: 28670929 DOI: 10.1080/14728222.2017.1349754] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jeroen E Guikema
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), The Netherlands
| | - Martine Amiot
- CRCINA, INSERM, CNRS, Université d’Angers, Université de Nantes, Nantes, France
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), The Netherlands
| |
Collapse
|
31
|
Pihán P, Carreras-Sureda A, Hetz C. BCL-2 family: integrating stress responses at the ER to control cell demise. Cell Death Differ 2017. [PMID: 28622296 DOI: 10.1038/cdd.2017.82] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the last decade, the endoplasmic reticulum (ER) has emerged as a central organelle regulating the core mitochondrial apoptosis pathway. At the ER membrane, a variety of stress signals are integrated toward determining cell fate, involving a complex cross talk between key homeostatic pathways including the unfolded protein response, autophagy, calcium signaling and mitochondrial bioenergetics. In this context, key regulators of cell death of the BCL-2 and TMBIM/BI-1 family of proteins have relevant functions as stress rheostats mediated by the formation of distinct protein complexes that regulate the switch between adaptive and proapoptotic phases under stress. Here, we overview recent advances on our molecular understanding of how the apoptotic machinery integrates stress signals toward cell fate decisions upstream of the mitochondrial gateway of death.
Collapse
Affiliation(s)
- Philippe Pihán
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Faculty of Medicine, Center for Geroscience, Brain Health and Metabolism, University of Chile, Santiago, Chile
| | - Amado Carreras-Sureda
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Faculty of Medicine, Center for Geroscience, Brain Health and Metabolism, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Faculty of Medicine, Center for Geroscience, Brain Health and Metabolism, University of Chile, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA 94945, USA.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston MA 02115, USA
| |
Collapse
|
32
|
Abstract
p53 that is activated in response to DNA-damaging stress can induce apoptosis or either transient or permanent cell cycle arrests. Apoptosis and permanent cell cycle arrest (senescence) are bona-fide tumor suppressor mechanisms through which p53 inhibits cancer cell survival. In contrast, transient cell cycle arrests induced by p53 can increase survival by allowing cells time to repair their DNA before proceeding with cell division. Mechanisms that control the choice of response to p53 (apoptosis vs arrest) are not fully understood. There is abundant crosstalk between p53 and the IGF-1R/AKT/mTORC1 signaling pathway. Recent studies indicate this crosstalk can determine the choice of response to p53. These findings have clear implications for the potential use of IGF-1R pathway inhibitors against p53 wild-type or p53-null or mutant cancers.
Collapse
Affiliation(s)
- Lei Duan
- Rush University Medical Center, Department of Anatomy and Cell Biology, 600 S Paulina Ave., AcFac 507, Chicago, IL 60612
| | - Carl G Maki
- Rush University Medical Center, Department of Anatomy and Cell Biology, 600 S Paulina Ave., AcFac 507, Chicago, IL 60612
| |
Collapse
|
33
|
Luo R, Fang D, Chu P, Wu H, Zhang Z, Tang Z. Multiple molecular targets in breast cancer therapy by betulinic acid. Biomed Pharmacother 2016; 84:1321-1330. [DOI: 10.1016/j.biopha.2016.10.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 01/11/2023] Open
|
34
|
Moncunill-Massaguer C, Saura-Esteller J, Pérez-Perarnau A, Palmeri CM, Núñez-Vázquez S, Cosialls AM, González-Gironès DM, Pomares H, Korwitz A, Preciado S, Albericio F, Lavilla R, Pons G, Langer T, Iglesias-Serret D, Gil J. A novel prohibitin-binding compound induces the mitochondrial apoptotic pathway through NOXA and BIM upregulation. Oncotarget 2016; 6:41750-65. [PMID: 26497683 PMCID: PMC4747186 DOI: 10.18632/oncotarget.6154] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/30/2015] [Indexed: 01/08/2023] Open
Abstract
We previously described diaryl trifluorothiazoline compound 1a (hereafter referred to as fluorizoline) as a first-in-class small molecule that induces p53-independent apoptosis in a wide range of tumor cell lines. Fluorizoline directly binds to prohibitin 1 and 2 (PHBs), two proteins involved in the regulation of several cellular processes, including apoptosis. Here we demonstrate that fluorizoline-induced apoptosis is mediated by PHBs, as cells depleted of these proteins are highly resistant to fluorizoline treatment. In addition, BAX and BAK are necessary for fluorizoline-induced cytotoxic effects, thereby proving that apoptosis occurs through the intrinsic pathway. Expression analysis revealed that fluorizoline induced the upregulation of Noxa and Bim mRNA levels, which was not observed in PHB-depleted MEFs. Finally, Noxa−/−/Bim−/− MEFs and NOXA-downregulated HeLa cells were resistant to fluorizoline-induced apoptosis. All together, these findings show that fluorizoline requires PHBs to execute the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Cristina Moncunill-Massaguer
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - José Saura-Esteller
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Alba Pérez-Perarnau
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Claudia Mariela Palmeri
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Ana M Cosialls
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Diana M González-Gironès
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Helena Pomares
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Anne Korwitz
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sara Preciado
- Barcelona Science Park and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain
| | - Fernando Albericio
- Barcelona Science Park and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain.,Institute for Research in Biomedicine Barcelona, Barcelona, Spain.,Department of Organic Chemistry, University of Barcelona, Barcelona, Spain
| | - Rodolfo Lavilla
- Barcelona Science Park and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain.,Laboratory of Organic Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Thomas Langer
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Daniel Iglesias-Serret
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| |
Collapse
|
35
|
El Halabi L, Ghez D, Ribrag V. Novel targeted therapeutics for mantle cell lymphoma – What’s on the horizon? Expert Rev Hematol 2016; 9:271-81. [DOI: 10.1586/17474086.2016.1134309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Activation of the DNA Damage Response by RNA Viruses. Biomolecules 2016; 6:2. [PMID: 26751489 PMCID: PMC4808796 DOI: 10.3390/biom6010002] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022] Open
Abstract
RNA viruses are a genetically diverse group of pathogens that are responsible for some of the most prevalent and lethal human diseases. Numerous viruses introduce DNA damage and genetic instability in host cells during their lifecycles and some species also manipulate components of the DNA damage response (DDR), a complex and sophisticated series of cellular pathways that have evolved to detect and repair DNA lesions. Activation and manipulation of the DDR by DNA viruses has been extensively studied. It is apparent, however, that many RNA viruses can also induce significant DNA damage, even in cases where viral replication takes place exclusively in the cytoplasm. DNA damage can contribute to the pathogenesis of RNA viruses through the triggering of apoptosis, stimulation of inflammatory immune responses and the introduction of deleterious mutations that can increase the risk of tumorigenesis. In addition, activation of DDR pathways can contribute positively to replication of viral RNA genomes. Elucidation of the interactions between RNA viruses and the DDR has provided important insights into modulation of host cell functions by these pathogens. This review summarises the current literature regarding activation and manipulation of the DDR by several medically important RNA viruses.
Collapse
|
37
|
RANJBAR S, HASHEMZADEH MS, KHOSHTINAT NIKKHOI S, FARASAT A, TAT M, GHALAVAND M, DOROSTKAR R. Selective suppression of tumor cells by a tumor-specific bicistronic lentiviral vector. Turk J Biol 2016. [DOI: 10.3906/biy-1512-53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
38
|
Duan L, Perez RE, Hansen M, Gitelis S, Maki CG. Increasing cisplatin sensitivity by schedule-dependent inhibition of AKT and Chk1. Cancer Biol Ther 2015; 15:1600-12. [PMID: 25482935 DOI: 10.4161/15384047.2014.961876] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The effectiveness of DNA damaging chemotherapy drugs can be limited by activation of survival signaling pathways and cell cycle checkpoints that allow DNA repair. Targeting survival pathways and inhibiting cell cycle checkpoints may increase chemotherapy-induced cancer cell killing. AKT and Chk1 are survival and cell cycle checkpoint kinases, respectively, that can be activated by DNA damage. Cisplatin (CP) is a standard chemotherapy agent for osteosarcoma (OS). CP induced apoptosis to varying extents and activated AKT and Chk1 in multiple p53 wild-type and p53-null OS cell lines. A Chk1 inhibitor increased CP-induced apoptosis in all OS cell lines regardless of p53 status. In contrast, an AKT inhibitor increased CP-induced apoptosis only in p53 wild-type OS cells, but not p53 nulll cells. The increased apoptosis in p53 wild-type cells was coincident with decreased p53 protein levels, but increased expression of p53-responsive apoptotic genes Noxa and PUMA. Further studies revealed the inability of AKT inhibitor to CP-sensitize p53-null OS cells resulted from 2 things: 1) AKT inhibition stabilized/maintained p27 levels in CP-treated cells, which then mediated a protective G1-phase cell cycle arrest, 2) AKT inhibition increased the levels of activated Chk1. Finally, schedule dependent inhibition of AKT and Chk1 evaded the protective G1 arrest mediated by p27 and maximized CP-induced OS cell killing. These data demonstrate AKT and Chk1 activation promote survival in CP-treated OS cells, and that strategic, scheduled targeting of AKT and Chk1 can maximize OS cell killing by CP.
Collapse
Affiliation(s)
- Lei Duan
- a Department of Anatomy and Cell Biology ; Rush University Medical Center ; Chicago , IL USA
| | | | | | | | | |
Collapse
|
39
|
The triterpenoids of Hibiscus syriacus induce apoptosis and inhibit cell migration in breast cancer cells. Altern Ther Health Med 2015; 15:65. [PMID: 25885960 PMCID: PMC4410586 DOI: 10.1186/s12906-015-0592-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 02/26/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Breast cancer-related mortality increases annually. The efficacy of current breast cancer treatments is limited, and they have numerous side effects and permit high recurrence. Patients with estrogen receptor (ER)-negative or triple-negative breast cancer are particularly difficult to treat. Treatment for this type of cancer is lacking, and its prognosis is poor, necessitating the search for alternative treatments. METHODS This study screened Chinese herb Hibiscus syriacus extracts and identified a novel anti-cancer drug for patients with ER-negative breast cancer. The inhibitory effects on cell viability and migration were evaluated for each compound, and the molecular regulatory effects were evaluated on both mRNA and protein levels. RESULT We found several triterpenoids including betulin (K02) and its derivatives (K03, K04, and K06) from H. syriacus inhibited human triple-negative breast cancer cell viability and migration but revealed smaller cytotoxic effects on normal mammalian epithelial cells. Betulin and its derivatives induced apoptosis by activating apoptosis-related genes. In addition, they activated p21 expression, which induced cell cycle arrest in breast cancer cells. Betulin (K02) and betulinic acid (K06) had stronger inhibitory effects on cell viability and migration than K03 and K04. CONCLUSIONS H. syriacus extracts might inhibit breast cancer cell viability and induce apoptosis by activating p53 family regulated pathways and inhibiting AKT activation. H. syriacus extracts may provide important insight into the development of novel alternative therapies for breast cancer.
Collapse
|
40
|
p53 negatively regulates Pin1 expression under ER stress. Biochem Biophys Res Commun 2014; 454:518-23. [PMID: 25451271 DOI: 10.1016/j.bbrc.2014.10.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 10/20/2014] [Indexed: 11/20/2022]
Abstract
Accumulating evidence suggests that endoplasmic reticulum (ER) stress plays a major role in the development of many diseases. A previous study indicated that the apoptotic regulator p53 is significantly increased in response to ER stress and participates in ER stress-induced apoptosis. However, the regulators of p53 expression during ER stress are still not fully understood. Here, we investigated whether p53 contributes to the impairment of Pin1 signaling under ER stress. We found that treatment with thapsigargin, a stimulator of p53 expression and an inducer of ER stress, decreased Pin1 expression in HCT116 cells. Also, we identified functional p53 response elements (p53REs) in the Pin1 promoter. Overexpression of p53 significantly decreased Pin1 expression in HCT116 cells while abolition of p53 gene expression induced Pin1 expression. Pin1 expression was significantly increased by treatment with the p53 inhibitor pifithrin-α or down-regulation of p53 expression. Taken together, ER stress decreased Pin1 expression through p53 activation, and this mechanism may be associated with ER stress-induced cell death. These data reported here support the importance of Pin1 as a potential target molecule mediating tumor development.
Collapse
|
41
|
Analysis of BH3-only proteins upregulated in response to oxygen/glucose deprivation in cortical neurons identifies Bmf but not Noxa as potential mediator of neuronal injury. Cell Death Dis 2014; 5:e1456. [PMID: 25299781 PMCID: PMC4237251 DOI: 10.1038/cddis.2014.426] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 08/28/2014] [Accepted: 09/04/2014] [Indexed: 11/30/2022]
Abstract
Stress signaling in response to oxygen/glucose deprivation (OGD) and ischemic injury activates a group of pro-apoptotic genes, the Bcl-2 homology domain 3 (BH3)-only proteins, which are capable of activating the mitochondrial apoptosis pathway. Targeted studies previously identified the BH3-only proteins Puma, Bim and Bid to have a role in ischemic/hypoxic neuronal injury. We here investigated the transcriptional activation of pro-apoptotic BH3-only proteins after OGD-induced injury in murine neocortical neurons. We observed a potent and early upregulation of noxa at mRNA and protein level, and a significant increase in Bmf protein levels during OGD in neocortical neurons and in the ipsilateral cortex of mice subjected to transient middle cerebral artery occlusion (tMCAO). Surprisingly, gene deficiency in noxa reduced neither OGD- nor glutamate-induced neuronal injury in cortical neurons and failed to influence infarct size or neurological deficits after tMCAO. In contrast, bmf deficiency induced significant protection against OGD- or glutamate-induced injury in cultured neurons, and bmf-deficient mice showed reduced neurological deficits after tMCAO in vivo. Collectively, our data not only point to a role of Bmf as a BH3-only protein contributing to excitotoxic and ischemic neuronal injury but also demonstrate that the early and potent induction of noxa does not influence ischemic neuronal injury.
Collapse
|
42
|
Wang S, Sun W, Zhao Y, McEachern D, Meaux I, Barrière C, Stuckey JA, Meagher JL, Bai L, Liu L, Hoffman-Luca CG, Lu J, Shangary S, Yu S, Bernard D, Aguilar A, Dos-Santos O, Besret L, Guerif S, Pannier P, Gorge-Bernat D, Debussche L. SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res 2014; 74:5855-65. [PMID: 25145672 DOI: 10.1158/0008-5472.can-14-0799] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Blocking the oncoprotein murine double minute 2 (MDM2)-p53 protein-protein interaction has long been considered to offer a broad cancer therapeutic strategy, despite the potential risks of selecting tumors harboring p53 mutations that escape MDM2 control. In this study, we report a novel small-molecule inhibitor of the MDM2-p53 interaction, SAR405838 (MI-77301), that has been advanced into phase I clinical trials. SAR405838 binds to MDM2 with K(i) = 0.88 nmol/L and has high specificity over other proteins. A cocrystal structure of the SAR405838:MDM2 complex shows that, in addition to mimicking three key p53 amino acid residues, the inhibitor captures additional interactions not observed in the p53-MDM2 complex and induces refolding of the short, unstructured MDM2 N-terminal region to achieve its high affinity. SAR405838 effectively activates wild-type p53 in vitro and in xenograft tumor tissue of leukemia and solid tumors, leading to p53-dependent cell-cycle arrest and/or apoptosis. At well-tolerated dose schedules, SAR405838 achieves either durable tumor regression or complete tumor growth inhibition in mouse xenograft models of SJSA-1 osteosarcoma, RS4;11 acute leukemia, LNCaP prostate cancer, and HCT-116 colon cancer. Remarkably, a single oral dose of SAR405838 is sufficient to achieve complete tumor regression in the SJSA-1 model. Mechanistically, robust transcriptional upregulation of PUMA induced by SAR405838 results in strong apoptosis in tumor tissue, leading to complete tumor regression. Our findings provide a preclinical basis upon which to evaluate SAR405838 as a therapeutic agent in patients whose tumors retain wild-type p53.
Collapse
Affiliation(s)
- Shaomeng Wang
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan. Department of Pharmacology, University of Michigan, Ann Arbor, Michigan. Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan.
| | - Wei Sun
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yujun Zhao
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Donna McEachern
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Jeanne A Stuckey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | | | - Longchuan Bai
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Liu Liu
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Cassandra Gianna Hoffman-Luca
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Jianfeng Lu
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sanjeev Shangary
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shanghai Yu
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Denzil Bernard
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Angelo Aguilar
- University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan. Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | | | |
Collapse
|
43
|
Kong B, Wang Q, Fung E, Xue K, Tsang BK. p53 is required for cisplatin-induced processing of the mitochondrial fusion protein L-Opa1 that is mediated by the mitochondrial metallopeptidase Oma1 in gynecologic cancers. J Biol Chem 2014; 289:27134-27145. [PMID: 25112877 DOI: 10.1074/jbc.m114.594812] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mitochondria are highly dynamic organelles, and mitochondrial fission is a crucial step of apoptosis. Although Oma1 is believed to be responsible for long form Opa1 (L-Opa1) processing during mitochondrial fragmentation, whether and how Oma1 is involved in L-Opa1 processing and participates in the regulation of chemoresistance is unknown. Chemosensitive and chemoresistant ovarian (OVCA) and cervical (CECA) cancer cells were treated with cisplatin (CDDP). Mitochondrial dynamics and protein contents were assessed by immunofluorescence and Western blot, respectively. The requirements of Oma1 and p53 for CDDP-induced L-Opa1 processing, mitochondrial fragmentation, and apoptosis were examined by siRNA or cDNA. CDDP induces L-Opa1 processing and mitochondrial fragmentation in chemosensitive but not in chemoresistant cells. CDDP induced Oma1 40-kDa form increases in OV2008 cells, not in C13* cells. Oma1 knockdown inhibited L-Opa1 processing, mitochondrial fragmentation, and apoptosis. Silencing p53 expression attenuated the effects of CDDP in Oma1 (40 kDa) increase, L-Opa1 processing, mitochondrial fragmentation, and apoptosis in chemosensitive OVCA cells, whereas reconstitution of p53 in p53 mutant or null chemoresistant OVCA cells induced Oma1 (40 kDa) increase, L-Opa1 processing, mitochondrial fragmentation, and apoptosis irrespective of the presence of CDDP. Prohibitin 1 (Phb1) dissociates from Opa1-Phb1 complex and binds phosphorylated p53 (serine 15) in response to CDDP in chemosensitive but not chemoresistant CECA cells. These findings demonstrate that (a) p53 and Oma1 mediate L-Opa1 processing, (b) mitochondrial fragmentation is involved in CDDP-induced apoptosis in OVCA and CECA cells, and (c) dysregulated mitochondrial dynamics may in part be involved in the pathophysiology of CDDP resistance.
Collapse
Affiliation(s)
- Bao Kong
- Departments of Obstetrics and Gynecology and Cellular and Molecular Medicine, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada,; Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada, and
| | - Qi Wang
- Departments of Obstetrics and Gynecology and Cellular and Molecular Medicine, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada,; Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada, and
| | - Ella Fung
- Departments of Obstetrics and Gynecology and Cellular and Molecular Medicine, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada,; Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada, and
| | - Kai Xue
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada, and; State Key Laboratory of Reproductive Medicine, Clinical Reproductive Medicine Centre, Nanjing Medical University, Nanjing 210029, China
| | - Benjamin K Tsang
- Departments of Obstetrics and Gynecology and Cellular and Molecular Medicine, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario K1H 8L6, Canada,; Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada, and.
| |
Collapse
|
44
|
Sizova OS, Shinde VM, Lenox AR, Gorbatyuk MS. Modulation of cellular signaling pathways in P23H rhodopsin photoreceptors. Cell Signal 2013; 26:665-672. [PMID: 24378535 PMCID: PMC4083739 DOI: 10.1016/j.cellsig.2013.12.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 12/09/2013] [Accepted: 12/22/2013] [Indexed: 11/23/2022]
Abstract
We previously reported activation of the unfolded protein response (UPR) in P23H rhodopsin (RHO) retinas with autosomal dominant retinitis pigmentosa (ADRP). Knowing that the UPR can trigger Ca2+ release from the endoplasmic reticulum and regulate cellular signaling we examined the level of Ca2+-regulated proteins. We also looked for changes in the expression of Bcl2 family proteins, autophagy proteins and the mTOR/AKT pathways, as well as for the induction of mitochondria-associated apoptosis in the P23H RHO retina. Our data demonstrated that the elevation of calpain and caspase-12 activity was concomitantly observed with a decrease in the BCL2-XL/BAX ratio and an increase in mTor levels in the P23H-3 RHO retina suggesting a vulnerability of P23H RHO photoreceptors to apoptosis. The translocation of BAX to the mitochondria, as well as the release of cytochrome C and AIF into the cytosol supports this conclusion and indicates the involvement of mitochondria-induced apoptosis in the progression of ADRP. The level of autophagy proteins in general was found to be decreased in the P21–P30 P23H RHO retina. Injections of rapamycin, however, protected the P23H RHO rod photoreceptors from experiencing physiological decline. Despite this fact, the downregulation of mTOR did not alter the level of autophagy proteins. Our results imply that in addition to activation of the UPR during ADRP progression, photoreceptors also experience alterations in major proapoptotic pathways.
Collapse
Affiliation(s)
- Olga S Sizova
- University of North Texas Health Science Center, North Texas Eye Research Institute, Department of Cell Biology and Anatomy, United States
| | - Vishal M Shinde
- University of Alabama at Birmingham, Department of Visual Sciences, United States
| | - Austin R Lenox
- University of Alabama at Birmingham, Department of Visual Sciences, United States
| | - Marina S Gorbatyuk
- University of Alabama at Birmingham, Department of Visual Sciences, United States
| |
Collapse
|
45
|
Hu J, Van Valckenborgh E, Xu D, Menu E, De Raeve H, De Bruyne E, De Bryune E, Xu S, Van Camp B, Handisides D, Hart CP, Vanderkerken K. Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro. Mol Cancer Ther 2013; 12:1763-73. [PMID: 23832122 DOI: 10.1158/1535-7163.mct-13-0123] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recently, we showed that hypoxia is a critical microenvironmental factor in multiple myeloma, and that the hypoxia-activated prodrug TH-302 selectively targets hypoxic multiple myeloma cells and improves multiple disease parameters in vivo. To explore approaches for sensitizing multiple myeloma cells to TH-302, we evaluated in this study the antitumor effect of TH-302 in combination with the clinically used proteasome inhibitor bortezomib. First, we show that TH-302 and bortezomib synergistically induce apoptosis in multiple myeloma cell lines in vitro. Second, we confirm that this synergism is related to the activation of caspase cascades and is mediated by changes of Bcl-2 family proteins. The combination treatment induces enhanced cleavage of caspase-3/8/9 and PARP, and therefore triggers apoptosis and enhances the cleavage of proapoptotic BH3-only protein BAD and BID as well as the antiapoptotic protein Mcl-1. In particular, TH-302 can abrogate the accumulation of antiapoptotic Mcl-1 induced by bortezomib, and decreases the expression of the prosurvival proteins Bcl-2 and Bcl-xL. Furthermore, we found that the induction of the proapoptotic BH3-only proteins PUMA (p53-upregulated modulator of apoptosis) and NOXA is associated with this synergism. In response to the genotoxic and endoplasmic reticulum stresses by TH-302 and bortezomib, the expression of PUMA and NOXA were upregulated in p53-dependent and -independent manners. Finally, in the murine 5T33MMvv model, we showed that the combination of TH-302 and bortezomib can improve multiple disease parameters and significantly prolong the survival of diseased mice. In conclusion, our studies provide a rationale for clinical evaluation of the combination of TH-302 and bortezomib in patients with multiple myeloma.
Collapse
Affiliation(s)
- Jinsong Hu
- Corresponding Author: Karin Vanderkerken, Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Webster KA. Mitochondrial membrane permeabilization and cell death during myocardial infarction: roles of calcium and reactive oxygen species. Future Cardiol 2013; 8:863-84. [PMID: 23176689 DOI: 10.2217/fca.12.58] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Excess generation of reactive oxygen species (ROS) and cytosolic calcium accumulation play major roles in the initiation of programmed cell death during acute myocardial infarction. Cell death may include necrosis, apoptosis and autophagy, and combinations thereof. During ischemia, calcium handling between the sarcoplasmic reticulum and myofilament is disrupted and calcium is diverted to the mitochondria causing swelling. Reperfusion, while essential for survival, reactivates energy transduction and contractility and causes the release of ROS and additional ionic imbalance. During acute ischemia-reperfusion, the principal death pathways are programmed necrosis and apoptosis through the intrinsic pathway, initiated by the opening of the mitochondrial permeability transition pore and outer mitochondrial membrane permeabilization, respectively. Despite intense investigation, the mechanisms of action and modes of regulation of mitochondrial membrane permeabilization are incompletely understood. Extrinsic apoptosis, necroptosis and autophagy may also contribute to ischemia-reperfusion injury. In this review, the roles of dysregulated calcium and ROS and the contributions of Bcl-2 proteins, as well as mitochondrial morphology in promoting mitochondrial membrane permeability change and the ensuing cell death during myocardial infarction are discussed.
Collapse
Affiliation(s)
- Keith A Webster
- Department of Molecular & Cellular Pharmacology, University of Miami Medical Center, FL 33101, USA.
| |
Collapse
|
47
|
Brinkmann K, Zigrino P, Witt A, Schell M, Ackermann L, Broxtermann P, Schüll S, Andree M, Coutelle O, Yazdanpanah B, Seeger J, Klubertz D, Drebber U, Hacker U, Krönke M, Mauch C, Hoppe T, Kashkar H. Ubiquitin C-Terminal Hydrolase-L1 Potentiates Cancer Chemosensitivity by Stabilizing NOXA. Cell Rep 2013; 3:881-91. [DOI: 10.1016/j.celrep.2013.02.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/18/2012] [Accepted: 02/11/2013] [Indexed: 01/09/2023] Open
|
48
|
Proliferative and survival effects of PUMA promote angiogenesis. Cell Rep 2012; 2:1272-85. [PMID: 23122957 DOI: 10.1016/j.celrep.2012.09.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 06/05/2012] [Accepted: 09/24/2012] [Indexed: 01/08/2023] Open
Abstract
The p53 upregulated modulator of apoptosis (PUMA) is known as an essential apoptosis inducer. Here, we report the seemingly paradoxical finding that PUMA is a proangiogenic factor critically required for the proliferation and survival of vascular and microglia cells. Strikingly, Puma deficiency by genetic deletion or small hairpin RNA knockdown inhibited developmental and pathological angiogenesis and reduced microglia numbers in vivo, whereas Puma gene delivery increased angiogenesis and cell survival. Mechanistically, we revealed that PUMA plays a critical role in regulating autophagy by modulating Erk activation and intracellular calcium level. Our findings revealed an unexpected function of PUMA in promoting angiogenesis and warrant more careful investigations into the therapeutic potential of PUMA in treating cancer and degenerative diseases.
Collapse
|
49
|
Yadunandam AK, Yoon JS, Seong YA, Oh CW, Kim GD. Prospective impact of 5-FU in the induction of endoplasmic reticulum stress, modulation of GRP78 expression and autophagy in Sk-Hep1 cells. Int J Oncol 2012; 41:1036-42. [PMID: 22684338 DOI: 10.3892/ijo.2012.1506] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/27/2012] [Indexed: 11/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive malignant diseases and is highly resistant to conventional chemotherapy. Therefore, HCC requires more effective prevention and treatment strategies. 5-fluorouracil (5-FU) remains the most widely used chemotherapeutic drug for the treatment of gastrointestinal, breast, head and neck, and ovarian cancers. In pursuit of a novel effective strategy, we have evaluated the potential of 5-FU to promote endoplasmic reticulum (ER) stress and autophagy in Sk-Hep1 HCC cells. We found that 5-FU profoundly induces ER stress in Sk-Hep1 cells and upregulates p53 and activates CHOP/GADD153 and caspase-12. Activation of CHOP/GADD153 and caspase-12 promotes mitochondrial cell death in Sk-Hep1 cells followed by ER stress. Changes in calcium homeostasis and the protein folding machinery cause stress in the ER, leading to apoptotic cell death. Stress in the ER activates autophagy to remove the misfolded protein aggregates and recover from the stress environment. Our study demonstrates that 5-FU-induced ER stress suppresses autophagy and also downregulates GRP78 expression. Activation of autophagy followed by ER stress facilitates the cell survival response. Therefore, the inhibition of protective autophagy may provide a useful pharmacological target. Taken together, these results indicate that 5-FU-induced ER stress activates the mitochondrial apoptotic cell death pathway by downregulating GRP78 and protective autophagy proteins in Sk-Hep1 cells, raising the possibility of using 5-FU as a therapeutic agent to target human HCC.
Collapse
Affiliation(s)
- Anandam Kasin Yadunandam
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 608-737, Republic of Korea
| | | | | | | | | |
Collapse
|
50
|
Ifeadi V, Garnett-Benson C. Sub-lethal irradiation of human colorectal tumor cells imparts enhanced and sustained susceptibility to multiple death receptor signaling pathways. PLoS One 2012; 7:e31762. [PMID: 22389673 PMCID: PMC3289623 DOI: 10.1371/journal.pone.0031762] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/18/2012] [Indexed: 01/01/2023] Open
Abstract
Background Death receptors (DR) of the TNF family function as anti-tumor immune effector molecules. Tumor cells, however, often exhibit DR-signaling resistance. Previous studies indicate that radiation can modify gene expression within tumor cells and increase tumor cell sensitivity to immune attack. The aim of this study is to investigate the synergistic effect of sub-lethal doses of ionizing radiation in sensitizing colorectal carcinoma cells to death receptor-mediated apoptosis. Methodology/Principal Findings The ability of radiation to modulate the expression of multiple death receptors (Fas/CD95, TRAILR1/DR4, TRAILR2/DR5, TNF-R1 and LTβR) was examined in colorectal tumor cells. The functional significance of sub-lethal doses of radiation in enhancing tumor cell susceptibility to DR-induced apoptosis was determined by in vitro functional sensitivity assays. The longevity of these changes and the underlying molecular mechanism of irradiation in sensitizing diverse colorectal carcinoma cells to death receptor-mediated apoptosis were also examined. We found that radiation increased surface expression of Fas, DR4 and DR5 but not LTβR or TNF-R1 in these cells. Increased expression of DRs was observed 2 days post-irradiation and remained elevated 7-days post irradiation. Sub-lethal tumor cell irradiation alone exhibited minimal cell death, but effectively sensitized three of three colorectal carcinoma cells to both TRAIL and Fas-induced apoptosis, but not LTβR-induced death. Furthermore, radiation-enhanced Fas and TRAIL-induced cell death lasted as long as 5-days post-irradiation. Specific analysis of intracellular sensitizers to apoptosis indicated that while radiation did reduce Bcl-XL and c-FLIP protein expression, this reduction did not correlate with the radiation-enhanced sensitivity to Fas and/or TRAIL mediated apoptosis among the three cell types. Conclusions/Significance Irradiation of tumor cells can overcome Fas and TRAIL resistance that is long lasting. Overall, results of these investigations suggest that non-lethal doses of radiation can be used to make human tumors more amenable to attack by anti-tumor effector molecules and cells.
Collapse
Affiliation(s)
| | - Charlie Garnett-Benson
- Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|