51
|
Wang T, Wang L, Zhang Y, Sun J, Xie Y, Yuan Y, Gu J, Bian J, Liu Z, Zou H. Puerarin Restores Autophagosome-Lysosome Fusion to Alleviate Cadmium-Induced Autophagy Blockade via Restoring the Expression of Rab7 in Hepatocytes. Front Pharmacol 2021; 12:632825. [PMID: 33935722 PMCID: PMC8079953 DOI: 10.3389/fphar.2021.632825] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/01/2021] [Indexed: 01/02/2023] Open
Abstract
Autophagic dysfunction is one of the main mechanisms by which the environmental pollutant cadmium (Cd) induces cell injury. Puerarin (Pue, a monomeric Chinese herbal medicine extract) has been reported to alleviate Cd-induced cell injury by regulating autophagy pathways; however, its detailed mechanisms are unclear. In the present study, to investigate the detailed mechanisms by which Pue targets autophagy to alleviate Cd hepatotoxicity, alpha mouse liver 12 (AML12) cells were used to construct a model of Cd-induced hepatocyte injury in vitro. First, the protective effect of Pue on Cd-induced cell injury was confirmed by changes in cell proliferation, cell morphology, and cell ultrastructure. Next, we found that Pue activated autophagy and mitigated Cd-induced autophagy blockade. In this process, the lysosome was further activated and the lysosomal degradation capacity was strengthened. We also found that Pue restored the autophagosome-lysosome fusion and the expression of Rab7 in Cd-exposed hepatocytes. However, the fusion of autophagosomes with lysosomes and autophagic flux were inhibited after knocking down Rab7, and were further inhibited after combined treatment with Cd. In addition, after knocking down Rab7, the protective effects of Pue on restoring autophagosome-lysosome fusion and alleviating autophagy blockade in Cd-exposed cells were inhibited. In conclusion, Pue-mediated alleviation of Cd-induced hepatocyte injury was related to the activation of autophagy and the alleviation of autophagy blockade. Pue also restored the fusion of autophagosomes and lysosomes by restoring the protein expression of Rab7, thereby alleviating Cd-induced autophagy blockade in hepatocytes.
Collapse
Affiliation(s)
- Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Li Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yi Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yilin Xie
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
52
|
Guiney SJ, Adlard PA, Lei P, Mawal CH, Bush AI, Finkelstein DI, Ayton S. Fibrillar α-synuclein toxicity depends on functional lysosomes. J Biol Chem 2021; 295:17497-17513. [PMID: 33453994 DOI: 10.1074/jbc.ra120.013428] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 10/01/2020] [Indexed: 02/05/2023] Open
Abstract
Neurodegeneration in Parkinson's disease (PD) can be recapitulated in animals by administration of α-synuclein preformed fibrils (PFFs) into the brain. However, the mechanism by which these PFFs induce toxicity is unknown. Iron is implicated in PD pathophysiology, so we investigated whether α-synuclein PFFs induce ferroptosis, an iron-dependent cell death pathway. A range of ferroptosis inhibitors were added to a striatal neuron-derived cell line (STHdhQ7/7 cells), a dopaminergic neuron-derived cell line (SN4741 cells), and WT primary cortical neurons, all of which had been intoxicated with α-synuclein PFFs. Viability was not recovered by these inhibitors except for liproxstatin-1, a best-in-class ferroptosis inhibitor, when used at high doses. High-dose liproxstatin-1 visibly enlarged the area of a cell that contained acidic vesicles and elevated the expression of several proteins associated with the autophagy-lysosomal pathway similarly to the known lysosomal inhibitors, chloroquine and bafilomycin A1. Consistent with high-dose liproxstatin-1 protecting via a lysosomal mechanism, we further de-monstrated that loss of viability induced by α-synuclein PFFs was attenuated by chloroquine and bafilomycin A1 as well as the lysosomal cysteine protease inhibitors, leupeptin, E-64D, and Ca-074-Me, but not other autophagy or lysosomal enzyme inhibitors. We confirmed using immunofluorescence microscopy that heparin prevented uptake of α-synuclein PFFs into cells but that chloroquine did not stop α-synuclein uptake into lysosomes despite impairing lysosomal function and inhibiting α-synuclein toxicity. Together, these data suggested that α-synuclein PFFs are toxic in functional lysosomes in vitro. Therapeutic strategies that prevent α-synuclein fibril uptake into lysosomes may be of benefit in PD.
Collapse
Affiliation(s)
- Stephanie J Guiney
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - Paul A Adlard
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - Peng Lei
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia; Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University/Collaborative Center for Biotherapy, Chengdu, China
| | - Celeste H Mawal
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - David I Finkelstein
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria Australia.
| |
Collapse
|
53
|
Ferreira PMP, Sousa RWRD, Ferreira JRDO, Militão GCG, Bezerra DP. Chloroquine and hydroxychloroquine in antitumor therapies based on autophagy-related mechanisms. Pharmacol Res 2021; 168:105582. [PMID: 33775862 DOI: 10.1016/j.phrs.2021.105582] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 02/09/2023]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are the most common drugs used to relieve acute and chronic inflammatory diseases. In this article, we present a review about the use of CQ and HCQ in antitumor therapies based on autophagy mechanisms. These molecules break/discontinue autophagosome-lysosome fusions in initial phases and enhance antiproliferative action of chemotherapeutics. Their sensitizing effects of chemotherapy when used as an adjuvant option in clinical trials against cancer. However, human related-MDR genes are also under risk to develop chemo or radioresistance because cancer cells have ability to throw 4-aminoquinolines out from digestive vacuoles well. Additionally, they also have antitumor mechanism unrelated to autophagy, including cell death from apoptosis and necroptosis and immunomodulatory/anti-inflammatory properties. However, the link between some anticancer mechanisms, clinical efficacy and pharmacological safety has not yet been fully defined.
Collapse
Affiliation(s)
- Paulo Michel Pinheiro Ferreira
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, 64049-550 Teresina, Brazil.
| | - Rayran Walter Ramos de Sousa
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, 64049-550 Teresina, Brazil
| | | | | | - Daniel Pereira Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ-BA), 40296-710 Salvador, Brazil
| |
Collapse
|
54
|
Puhl AC, Fritch EJ, Lane TR, Tse LV, Yount BL, Sacramento CQ, Fintelman-Rodrigues N, Tavella TA, Maranhão Costa FT, Weston S, Logue J, Frieman M, Premkumar L, Pearce KH, Hurst BL, Andrade CH, Levi JA, Johnson NJ, Kisthardt SC, Scholle F, Souza TML, Moorman NJ, Baric RS, Madrid PB, Ekins S. Repurposing the Ebola and Marburg Virus Inhibitors Tilorone, Quinacrine, and Pyronaridine: In Vitro Activity against SARS-CoV-2 and Potential Mechanisms. ACS OMEGA 2021; 6:7454-7468. [PMID: 33778258 PMCID: PMC7992063 DOI: 10.1021/acsomega.0c05996] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/02/2021] [Indexed: 05/11/2023]
Abstract
Severe acute respiratory coronavirus 2 (SARS-CoV-2) is a newly identified virus that has resulted in over 2.5 million deaths globally and over 116 million cases globally in March, 2021. Small-molecule inhibitors that reverse disease severity have proven difficult to discover. One of the key approaches that has been widely applied in an effort to speed up the translation of drugs is drug repurposing. A few drugs have shown in vitro activity against Ebola viruses and demonstrated activity against SARS-CoV-2 in vivo. Most notably, the RNA polymerase targeting remdesivir demonstrated activity in vitro and efficacy in the early stage of the disease in humans. Testing other small-molecule drugs that are active against Ebola viruses (EBOVs) would appear a reasonable strategy to evaluate their potential for SARS-CoV-2. We have previously repurposed pyronaridine, tilorone, and quinacrine (from malaria, influenza, and antiprotozoal uses, respectively) as inhibitors of Ebola and Marburg viruses in vitro in HeLa cells and mouse-adapted EBOV in mice in vivo. We have now tested these three drugs in various cell lines (VeroE6, Vero76, Caco-2, Calu-3, A549-ACE2, HUH-7, and monocytes) infected with SARS-CoV-2 as well as other viruses (including MHV and HCoV 229E). The compilation of these results indicated considerable variability in antiviral activity observed across cell lines. We found that tilorone and pyronaridine inhibited the virus replication in A549-ACE2 cells with IC50 values of 180 nM and IC50 198 nM, respectively. We used microscale thermophoresis to test the binding of these molecules to the spike protein, and tilorone and pyronaridine bind to the spike receptor binding domain protein with K d values of 339 and 647 nM, respectively. Human Cmax for pyronaridine and quinacrine is greater than the IC50 observed in A549-ACE2 cells. We also provide novel insights into the mechanism of these compounds which is likely lysosomotropic.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ethan J. Fritch
- Department
of Microbiology and Immunology, University
of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Thomas R. Lane
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Longping V. Tse
- Department
of Epidemiology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Boyd L. Yount
- Department
of Epidemiology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Carolina Q. Sacramento
- Laboratório
de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ 21040-900, Brazil
- Centro
De Desenvolvimento Tecnológico Em Saúde (CDTS), Fiocruz, Rio de
Janeiro 21040-900, Brazil
| | - Natalia Fintelman-Rodrigues
- Laboratório
de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ 21040-900, Brazil
- Centro
De Desenvolvimento Tecnológico Em Saúde (CDTS), Fiocruz, Rio de
Janeiro 21040-900, Brazil
| | - Tatyana Almeida Tavella
- Laboratory
of Tropical Diseases—Prof. Dr. Luiz Jacinto da Silva, Department
of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo 13083-970, Brazil
| | - Fabio Trindade Maranhão Costa
- Laboratory
of Tropical Diseases—Prof. Dr. Luiz Jacinto da Silva, Department
of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo 13083-970, Brazil
| | - Stuart Weston
- Department
of Microbiology and Immunology, University
of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - James Logue
- Department
of Microbiology and Immunology, University
of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Matthew Frieman
- Department
of Microbiology and Immunology, University
of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Lakshmanane Premkumar
- Department
of Microbiology and Immunology, University
of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Kenneth H. Pearce
- Center
for Integrative Chemical Biology and Drug Discovery, Chemical Biology
and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- UNC
Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina 27599, United States
| | - Brett L. Hurst
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322, United States
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah 84322, United States
| | - Carolina Horta Andrade
- Laboratory
of Tropical Diseases—Prof. Dr. Luiz Jacinto da Silva, Department
of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo 13083-970, Brazil
- LabMol—Laboratory of Molecular Modeling
and Drug Design, Faculdade
de Farmácia, Universidade Federal
de Goiás, Goiânia,
GO 74605-170, Brazil
| | - James A. Levi
- Department of Biological Sciences, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - Nicole J. Johnson
- Department of Biological Sciences, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - Samantha C. Kisthardt
- Department of Biological Sciences, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - Frank Scholle
- Department of Biological Sciences, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - Thiago Moreno L. Souza
- Laboratório
de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ 21040-900, Brazil
- Centro
De Desenvolvimento Tecnológico Em Saúde (CDTS), Fiocruz, Rio de
Janeiro 21040-900, Brazil
| | - Nathaniel John Moorman
- Department
of Microbiology and Immunology, University
of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
- Center
for Integrative Chemical Biology and Drug Discovery, Chemical Biology
and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Rapidly Emerging Antiviral Drug Discovery
Initiative, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Ralph S. Baric
- Department
of Microbiology and Immunology, University
of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
- Department
of Epidemiology, University of North Carolina
School of Medicine, Chapel Hill, North Carolina 27599, United States
- Rapidly Emerging Antiviral Drug Discovery
Initiative, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Peter B. Madrid
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
55
|
Bik E, Mateuszuk L, Orleanska J, Baranska M, Chlopicki S, Majzner K. Chloroquine-Induced Accumulation of Autophagosomes and Lipids in the Endothelium. Int J Mol Sci 2021; 22:ijms22052401. [PMID: 33673688 PMCID: PMC7957661 DOI: 10.3390/ijms22052401] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/14/2023] Open
Abstract
Chloroquine (CQ) is an antimalarial drug known to inhibit autophagy flux by impairing autophagosome–lysosome fusion. We hypothesized that autophagy flux altered by CQ has a considerable influence on the lipid composition of endothelial cells. Thus, we investigated endothelial responses induced by CQ on human microvascular endothelial cells (HMEC-1). HMEC-1 cells after CQ exposure were measured using a combined methodology based on label-free Raman and fluorescence imaging. Raman spectroscopy was applied to characterize subtle chemical changes in lipid contents and their distribution in the cells, while the fluorescence staining (LipidTox, LysoTracker and LC3) was used as a reference method. The results showed that CQ was not toxic to endothelial cells and did not result in the endothelial inflammation at concentrations of 1–30 µM. Notwithstanding, it yielded an increased intensity of LipidTox, LysoTracker, and LC3 staining, suggesting changes in the content of neutral lipids, lysosomotropism, and autophagy inhibition, respectively. The CQ-induced endothelial response was associated with lipid accumulation and was characterized by Raman spectroscopy. CQ-induced autophagosome accumulation in the endothelium is featured by a pronounced alteration in the lipid profile, but not in the endothelial inflammation. Raman-based assessment of CQ-induced biochemical changes offers a better understanding of the autophagy mechanism in the endothelial cells.
Collapse
Affiliation(s)
- Ewelina Bik
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
| | - Jagoda Orleanska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
| | - Malgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Chair of Pharmacology, Jagiellonian University, 16 Grzegorzecka Str., 31-531 Krakow, Poland
| | - Katarzyna Majzner
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland; (E.B.); (L.M.); (J.O.); (M.B.); (S.C.)
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
- Correspondence:
| |
Collapse
|
56
|
Wilkaniec A, Lenkiewicz AM, Babiec L, Murawska E, Jęśko HM, Cieślik M, Culmsee C, Adamczyk A. Exogenous Alpha-Synuclein Evoked Parkin Downregulation Promotes Mitochondrial Dysfunction in Neuronal Cells. Implications for Parkinson's Disease Pathology. Front Aging Neurosci 2021; 13:591475. [PMID: 33716707 PMCID: PMC7943853 DOI: 10.3389/fnagi.2021.591475] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Aberrant secretion and accumulation of α-synuclein (α-Syn) as well as the loss of parkin function are associated with the pathogenesis of Parkinson's disease (PD). Our previous study suggested a functional interaction between those two proteins, showing that the extracellular α-Syn evoked post-translational modifications of parkin, leading to its autoubiquitination and degradation. While parkin plays an important role in mitochondrial biogenesis and turnover, including mitochondrial fission/fusion as well as mitophagy, the involvement of parkin deregulation in α-Syn-induced mitochondrial damage is largely unknown. In the present study, we demonstrated that treatment with exogenous α-Syn triggers mitochondrial dysfunction, reflected by the depolarization of the mitochondrial membrane, elevated synthesis of the mitochondrial superoxide anion, and a decrease in cellular ATP level. At the same time, we observed a protective effect of parkin overexpression on α-Syn-induced mitochondrial dysfunction. α-Syn-dependent disturbances of mitophagy were also shown to be directly related to reduced parkin levels in mitochondria and decreased ubiquitination of mitochondrial proteins. Also, α-Syn impaired mitochondrial biosynthesis due to the parkin-dependent reduction of PGC-1α protein levels. Finally, loss of parkin function as a result of α-Syn treatment induced an overall breakdown of mitochondrial homeostasis that led to the accumulation of abnormal mitochondria. These findings may thus provide the first compelling evidence for the direct association of α-Syn-mediated parkin depletion to impaired mitochondrial function in PD. We suggest that improvement of parkin function may serve as a novel therapeutic strategy to prevent mitochondrial impairment and neurodegeneration in PD (thereby slowing the progression of the disease).
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| | - Anna M Lenkiewicz
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| | - Lidia Babiec
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| | - Emilia Murawska
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| | - Henryk M Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Philipps-University of Marburg, Marburg, Germany
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre (PAN), Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
57
|
Saha J, Kim JH, Amaya CN, Witcher C, Khammanivong A, Korpela DM, Brown DR, Taylor J, Bryan BA, Dickerson EB. Propranolol Sensitizes Vascular Sarcoma Cells to Doxorubicin by Altering Lysosomal Drug Sequestration and Drug Efflux. Front Oncol 2021; 10:614288. [PMID: 33598432 PMCID: PMC7882688 DOI: 10.3389/fonc.2020.614288] [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: 10/05/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023] Open
Abstract
Angiosarcoma is a rare cancer of blood vessel-forming cells with a high patient mortality and few treatment options. Although chemotherapy often produces initial clinical responses, outcomes remain poor, largely due to the development of drug resistance. We previously identified a subset of doxorubicin-resistant cells in human angiosarcoma and canine hemangiosarcoma cell lines that exhibit high lysosomal accumulation of doxorubicin. Hydrophobic, weak base chemotherapeutics, like doxorubicin, are known to sequester within lysosomes, promoting resistance by limiting drug accessibility to cellular targets. Drug synergy between the beta adrenergic receptor (β-AR) antagonist, propranolol, and multiple chemotherapeutics has been documented in vitro, and clinical data have corroborated the increased therapeutic potential of propranolol with chemotherapy in angiosarcoma patients. Because propranolol is also a weak base and accumulates in lysosomes, we sought to determine whether propranolol enhanced doxorubicin cytotoxicity via antagonism of β-ARs or by preventing the lysosomal accumulation of doxorubicin. β-AR-like immunoreactivities were confirmed in primary tumor tissues and cell lines; receptor function was verified by monitoring downstream signaling pathways of β-ARs in response to receptor agonists and antagonists. Mechanistically, propranolol increased cytoplasmic doxorubicin concentrations in sarcoma cells by decreasing the lysosomal accumulation and cellular efflux of this chemotherapeutic agent. Equivalent concentrations of the receptor-active S-(-) and -inactive R-(+) enantiomers of propranolol produced similar effects, supporting a β-AR-independent mechanism. Long-term exposure of hemangiosarcoma cells to propranolol expanded both lysosomal size and number, yet cells remained sensitive to doxorubicin in the presence of propranolol. In contrast, removal of propranolol increased cellular resistance to doxorubicin, underscoring lysosomal doxorubicin sequestration as a key mechanism of resistance. Our results support the repurposing of the R-(+) enantiomer of propranolol with weak base chemotherapeutics to increase cytotoxicity and reduce the development of drug-resistant cell populations without the cardiovascular and other side effects associated with antagonism of β-ARs.
Collapse
Affiliation(s)
- Jhuma Saha
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Jong Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States.,Animal Cancer Care and Research Program, College of Veterinary Medicine University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Clarissa N Amaya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, United States.,Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Caleb Witcher
- Department of Biology, Stephen F. Austin State University, Nacogdoches, TX, United States
| | - Ali Khammanivong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Derek M Korpela
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - David R Brown
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Josephine Taylor
- Department of Biology, Stephen F. Austin State University, Nacogdoches, TX, United States
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, United States.,Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Erin B Dickerson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States.,Animal Cancer Care and Research Program, College of Veterinary Medicine University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
58
|
Collins KP, Witta S, Coy JW, Pang Y, Gustafson DL. Lysosomal Biogenesis and Implications for Hydroxychloroquine Disposition. J Pharmacol Exp Ther 2021; 376:294-305. [PMID: 33172973 PMCID: PMC7841421 DOI: 10.1124/jpet.120.000309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022] Open
Abstract
Lysosomes act as a cellular drug sink for weakly basic, lipophilic (lysosomotropic) xenobiotics, with many instances of lysosomal trapping associated with multiple drug resistance. Lysosomotropic agents have also been shown to activate master lysosomal biogenesis transcription factor EB (TFEB) and ultimately lysosomal biogenesis. We investigated the role of lysosomal biogenesis in the disposition of hydroxychloroquine (HCQ), a hallmark lysosomotropic agent, and observed that modulating the lysosomal volume of human breast cancer cell lines can account for differences in disposition of HCQ. Through use of an in vitro pharmacokinetic (PK) model, we characterized total cellular uptake of HCQ within the duration of static equilibrium (1 hour), as well as extended exposure to HCQ that is subject to dynamic equilibrium (>1 hour), wherein HCQ increases the size of the lysosomal compartment through swelling and TFEB-induced lysosomal biogenesis. In addition, we observe that pretreatment of cell lines with TFEB-activating agent Torin1 contributed to an increase of whole-cell HCQ concentrations by 1.4- to 1.6-fold, which were also characterized by the in vitro PK model. This investigation into the role of lysosomal volume dynamics in lysosomotropic drug disposition, including the ability of HCQ to modify its own disposition, advances our understanding of how chemically similar agents may distribute on the cellular level and examines a key area of lysosomal-mediated multiple drug resistance and drug-drug interaction. SIGNIFICANCE STATEMENT: Hydroxychloroquine is able to modulate its own cellular pharmacokinetic uptake by increasing the cellular lysosomal volume fraction through activation of lysosomal biogenesis master transcription factor EB and through lysosomal swelling. This concept can be applied to many other lysosomotropic drugs that activate transcription factor EB, such as doxorubicin and other tyrosine kinase inhibitor drugs, as these drugs may actively increase their own sequestration within the lysosome to further exacerbate multiple drug resistance and lead to potential acquired resistance.
Collapse
Affiliation(s)
- Keagan P Collins
- Colorado State University, School of Biomedical Engineering (K.P.C., S.W., D.L.G.) and Department of Clinical Sciences (D.L.G., J.W.C.), Colorado State University, Fort Collins, Colorado; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado (D.L.G.); and University of Akron, Department of Chemistry, Akron, Ohio (Y.P.)
| | - Sandra Witta
- Colorado State University, School of Biomedical Engineering (K.P.C., S.W., D.L.G.) and Department of Clinical Sciences (D.L.G., J.W.C.), Colorado State University, Fort Collins, Colorado; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado (D.L.G.); and University of Akron, Department of Chemistry, Akron, Ohio (Y.P.)
| | - Jonathan W Coy
- Colorado State University, School of Biomedical Engineering (K.P.C., S.W., D.L.G.) and Department of Clinical Sciences (D.L.G., J.W.C.), Colorado State University, Fort Collins, Colorado; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado (D.L.G.); and University of Akron, Department of Chemistry, Akron, Ohio (Y.P.)
| | - Yi Pang
- Colorado State University, School of Biomedical Engineering (K.P.C., S.W., D.L.G.) and Department of Clinical Sciences (D.L.G., J.W.C.), Colorado State University, Fort Collins, Colorado; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado (D.L.G.); and University of Akron, Department of Chemistry, Akron, Ohio (Y.P.)
| | - Daniel L Gustafson
- Colorado State University, School of Biomedical Engineering (K.P.C., S.W., D.L.G.) and Department of Clinical Sciences (D.L.G., J.W.C.), Colorado State University, Fort Collins, Colorado; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado (D.L.G.); and University of Akron, Department of Chemistry, Akron, Ohio (Y.P.)
| |
Collapse
|
59
|
Udupa A, Leverenz D, Balevic SJ, Sadun RE, Tarrant TK, Rogers JL. Hydroxychloroquine and COVID-19: a Rheumatologist's Take on the Lessons Learned. Curr Allergy Asthma Rep 2021; 21:5. [PMID: 33475900 PMCID: PMC7818062 DOI: 10.1007/s11882-020-00983-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Told from the viewpoint of rheumatologists, this review tells the story of hydroxychloroquine and its swift ascent to become a household name as a therapeutic strategy against the novel SARS-CoV-2 virus. This review describes the history, mechanisms, pharmacokinetics, therapeutic applications, and safety profile of hydroxychloroquine as an immunomodulatory and antiviral agent. It also summarizes the major studies that launched and assessed the use of hydroxychloroquine against COVID-19 infection. RECENT FINDINGS More recent literature calls into question the long-held dogma that endolysosomal alkalinization is the primary mode of action of hydroxychloroquine. Ongoing uncertainty about the multiple potential mechanisms contributing to the therapeutic effect of hydroxychloroquine in rheumatic and viral disease led to a natural avenue for exploration in the treatment of COVID-19. Taken as a whole, the literature does not support utilizing hydroxychloroquine to treat or prevent infection from the SARS-CoV-2 virus. This is, at least in part, due to the wide variability in hydroxychloroquine pharmacokinetics between patients and difficulty achieving adequate target tissue concentrations of hydroxychloroquine without encountering unacceptable toxicities. Hydroxychloroquine continues to be a routinely prescribed, well-tolerated, effective, and low-cost treatment for rheumatic disease. Its therapeutic versatility has led to frequent repurposing for other conditions, most recently as an investigative treatment against the SARS-CoV-2 virus. Despite overall negative findings, the intense study of hydroxychloroquine against COVID-19 infection has enhanced our overall understanding of how hydroxychloroquine operates in autoimmune disease and beyond.
Collapse
Affiliation(s)
- Akrithi Udupa
- Duke University Medical Center, Box 2978, Durham, NC, 27710, USA.
| | - David Leverenz
- Duke University Medical Center, Box 2978, Durham, NC, 27710, USA
| | | | - Rebecca E Sadun
- Duke University Medical Center, Box 2978, Durham, NC, 27710, USA
| | - Teresa K Tarrant
- Duke University Medical Center, Box 2978, Durham, NC, 27710, USA
| | | |
Collapse
|
60
|
Zhai X, El Hiani Y. Getting Lost in the Cell-Lysosomal Entrapment of Chemotherapeutics. Cancers (Basel) 2020; 12:E3669. [PMID: 33297435 PMCID: PMC7762281 DOI: 10.3390/cancers12123669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Despite extensive research, resistance to chemotherapy still poses a major obstacle in clinical oncology. An exciting strategy to circumvent chemoresistance involves the identification and subsequent disruption of cellular processes that are aberrantly altered in oncogenic states. Upon chemotherapeutic challenges, lysosomes are deemed to be essential mediators that enable cellular adaptation to stress conditions. Therefore, lysosomes potentially hold the key to disarming the fundamental mechanisms of chemoresistance. This review explores modes of action of classical chemotherapeutic agents, adaptive response of the lysosomes to cell stress, and presents physiological and pharmacological insights pertaining to drug compartmentalization, sequestration, and extracellular clearance through the lens of lysosomes.
Collapse
Affiliation(s)
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
61
|
Puhl AC, Fritch EJ, Lane TR, Tse LV, Yount BL, Sacramento CQ, Tavella TA, Costa FTM, Weston S, Logue J, Frieman M, Premkumar L, Pearce KH, Hurst BL, Andrade CH, Levi JA, Johnson NJ, Kisthardt SC, Scholle F, Souza TML, Moorman NJ, Baric RS, Madrid P, Ekins S. Repurposing the Ebola and Marburg Virus Inhibitors Tilorone, Quinacrine and Pyronaridine: In vitro Activity Against SARS-CoV-2 and Potential Mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.12.01.407361. [PMID: 33299990 PMCID: PMC7724658 DOI: 10.1101/2020.12.01.407361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SARS-CoV-2 is a newly identified virus that has resulted in over 1.3 M deaths globally and over 59 M cases globally to date. Small molecule inhibitors that reverse disease severity have proven difficult to discover. One of the key approaches that has been widely applied in an effort to speed up the translation of drugs is drug repurposing. A few drugs have shown in vitro activity against Ebola virus and demonstrated activity against SARS-CoV-2 in vivo . Most notably the RNA polymerase targeting remdesivir demonstrated activity in vitro and efficacy in the early stage of the disease in humans. Testing other small molecule drugs that are active against Ebola virus would seem a reasonable strategy to evaluate their potential for SARS-CoV-2. We have previously repurposed pyronaridine, tilorone and quinacrine (from malaria, influenza, and antiprotozoal uses, respectively) as inhibitors of Ebola and Marburg virus in vitro in HeLa cells and of mouse adapted Ebola virus in mouse in vivo . We have now tested these three drugs in various cell lines (VeroE6, Vero76, Caco-2, Calu-3, A549-ACE2, HUH-7 and monocytes) infected with SARS-CoV-2 as well as other viruses (including MHV and HCoV 229E). The compilation of these results indicated considerable variability in antiviral activity observed across cell lines. We found that tilorone and pyronaridine inhibited the virus replication in A549-ACE2 cells with IC 50 values of 180 nM and IC 50 198 nM, respectively. We have also tested them in a pseudovirus assay and used microscale thermophoresis to test the binding of these molecules to the spike protein. They bind to spike RBD protein with K d values of 339 nM and 647 nM, respectively. Human C max for pyronaridine and quinacrine is greater than the IC 50 hence justifying in vivo evaluation. We also provide novel insights into their mechanism which is likely lysosomotropic.
Collapse
Affiliation(s)
- Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Ethan James Fritch
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
| | - Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Longping V. Tse
- Department of Epidemiology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
| | - Boyd L. Yount
- Department of Epidemiology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
| | - Carol Queiroz Sacramento
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Centro De Desenvolvimento Tecnológico Em Saúde (CDTS), Fiocruz, Rio de Janeiro, Brasil
| | - Tatyana Almeida Tavella
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Stuart Weston
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James Logue
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
| | - Kenneth H. Pearce
- Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina 27599, USA
| | - Brett L. Hurst
- Institute for Antiviral Research, Utah State University, Logan, UT, USA
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Carolina Horta Andrade
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - James A. Levi
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Nicole J. Johnson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Samantha C. Kisthardt
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Frank Scholle
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Thiago Moreno L. Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Centro De Desenvolvimento Tecnológico Em Saúde (CDTS), Fiocruz, Rio de Janeiro, Brasil
| | - Nathaniel John Moorman
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
- Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
- Department of Epidemiology, University of North Carolina School of Medicine, Chapel Hill NC 27599, USA
- Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peter Madrid
- SRI International, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
62
|
Krishnan Y, Zou J, Jani MS. Quantitative Imaging of Biochemistry in Situ and at the Nanoscale. ACS CENTRAL SCIENCE 2020; 6:1938-1954. [PMID: 33274271 PMCID: PMC7706076 DOI: 10.1021/acscentsci.0c01076] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Indexed: 05/12/2023]
Abstract
Biochemical reactions in eukaryotic cells occur in subcellular, membrane-bound compartments called organelles. Each kind of organelle is characterized by a unique lumenal chemical composition whose stringent regulation is vital to proper organelle function. Disruption of the lumenal ionic content of organelles is inextricably linked to disease. Despite their vital roles in cellular homeostasis, there are large gaps in our knowledge of organellar chemical composition largely from a lack of suitable probes. In this Outlook, we describe how, using organelle-targeted ratiometric probes, one can quantitatively image the lumenal chemical composition and biochemical activity inside organelles. We discuss how excellent fluorescent detection chemistries applied largely to the cytosol may be expanded to study organelles by chemical imaging at subcellular resolution in live cells. DNA-based reporters are a new and versatile platform to enable such approaches because the resultant probes have precise ratiometry and accurate subcellular targeting and are able to map multiple chemicals simultaneously. Quantitatively mapping lumenal ions and biochemical activity can drive the discovery of new biology and biomedical applications.
Collapse
Affiliation(s)
| | - Junyi Zou
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
- Grossman Institute of Neuroscience,
Quantitative Biology and Human Behavior, University of Chicago, Chicago, Illinois 60637, United States
| | - Maulik S. Jani
- Department of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
- Grossman Institute of Neuroscience,
Quantitative Biology and Human Behavior, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
63
|
Li Y, Xu Y, Xie J, Chen W. Malvidin-3-O-arabinoside ameliorates ethyl carbamate-induced oxidative damage by stimulating AMPK-mediated autophagy. Food Funct 2020; 11:10317-10328. [PMID: 33215619 DOI: 10.1039/d0fo01562h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ethyl carbamate (EC) is an environmental toxin, commonly present in various fermented foods and beverages, as well as tobacco and polluted ambient air. However, studies on the effects of EC-induced toxicity on the intestines and potential protection methods are limited. In this study, we show that EC could cause severe toxicity in intestinal epithelial cells (IECs) triggering the induction of decreased cell viability, ROS accumulation and glutathione (GSH) depletion in a dose-dependent manner. Based on these results, we established an EC-treated IEC model to screen the potential protective effects of 12 kinds of anthocyanins extracted from blueberry. Interestingly, we found that malvidin-3-O-arabinoside (M3A) significantly reversed the oxidative damage caused by EC exposure by stimulating autophagy flux, which was determined by the LC3-II level and GFP-RFP-LC3 transfection experiment. Enhancement of autophagy was mainly ascribed to the regulation of lysosomes. M3A pretreatment remarkably upregulated LAMP-1 expression, which indicated elevated lysosomal mass. Besides, M3A also successfully restored lysosomal acidity and subsequently strengthened lysosomal functions. Furthermore, M3A stimulated phosphorylation of AMP-activated protein kinase (AMPK), a master regulator of autophagy. Furthermore, our study indicated the possibility of EC-caused oxidative damage to the intestines and unveiled the remarkably protective benefits of M3A-induced AMPK-mediated autophagy against this toxicity.
Collapse
Affiliation(s)
- Yuting Li
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China.
| | | | | | | |
Collapse
|
64
|
Marinho EM, Batista de Andrade Neto J, Silva J, Rocha da Silva C, Cavalcanti BC, Marinho ES, Nobre Júnior HV. Virtual screening based on molecular docking of possible inhibitors of Covid-19 main protease. Microb Pathog 2020; 148:104365. [PMID: 32619669 PMCID: PMC7834391 DOI: 10.1016/j.micpath.2020.104365] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus (COVID-19) is an enveloped RNA virus that is diversely found in humans and that has now been declared a global pandemic by the World Health Organization. Thus, there is an urgent need to develop effective therapies and vaccines against this disease. In this context, this study aimed to evaluate in silico the molecular interactions of drugs with therapeutic indications for treatment of COVID-19 (Azithromycin, Baricitinib and Hydroxychloroquine) and drugs with similar structures (Chloroquine, Quinacrine and Ruxolitinib) in docking models from the SARS-CoV-2 main protease (M-pro) protein. The results showed that all inhibitors bound to the same enzyme site, more specifically in domain III of the SARS-CoV-2 main protease. Therefore, this study allows proposing the use of baricitinib and quinacrine, in combination with azithromycin; however, these computer simulations are just an initial step for conceiving new projects for the development of antiviral molecules.
Collapse
Affiliation(s)
- Emanuelle Machado Marinho
- Department of Analytical Chemistry and Physical Chemistry, Group of Theoretical Chemistry (GQT), Science Center, Federal University of Ceará, Fortaleza, CE, 60.455-760, Brazil
| | - João Batista de Andrade Neto
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceara, Fortaleza, CE, Brazil; Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jacilene Silva
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Cecília Rocha da Silva
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceara, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Emmanuel Silva Marinho
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Hélio Vitoriano Nobre Júnior
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceara, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
65
|
Korolenko TA, Johnston TP, Vetvicka V. Lysosomotropic Features and Autophagy Modulators among Medical Drugs: Evaluation of Their Role in Pathologies. Molecules 2020; 25:molecules25215052. [PMID: 33143272 PMCID: PMC7662698 DOI: 10.3390/molecules25215052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/25/2022] Open
Abstract
The concept of lysosomotropic agents significantly changed numerous aspects of cellular biochemistry, biochemical pharmacology, and clinical medicine. In the present review, we focused on numerous low-molecular and high-molecular lipophilic basic compounds and on the role of lipophagy and autophagy in experimental and clinical medicine. Attention was primarily focused on the most promising agents acting as autophagy inducers, which offer a new window for treatment and/or prophylaxis of various diseases, including type 2 diabetes mellitus, Parkinson's disease, and atherosclerosis. The present review summarizes current knowledge on the lysosomotropic features of medical drugs, as well as autophagy inducers, and their role in pathological processes.
Collapse
Affiliation(s)
- Tatiana A. Korolenko
- Federal State Budgetary Scientific Institution Scientific Research Institute of Physiology and Basic Medicine, Timakova Str. 4, 630117 Novosibirsk, Russia;
| | - Thomas P. Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO 64108, USA;
| | - Vaclav Vetvicka
- Department of Pathology, University of Louisville, Louisville, KY 40292, USA
- Correspondence:
| |
Collapse
|
66
|
Varalda M, Antona A, Bettio V, Roy K, Vachamaram A, Yellenki V, Massarotti A, Baldanzi G, Capello D. Psychotropic Drugs Show Anticancer Activity by Disrupting Mitochondrial and Lysosomal Function. Front Oncol 2020; 10:562196. [PMID: 33194631 PMCID: PMC7604408 DOI: 10.3389/fonc.2020.562196] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
Background and Purpose: Drug repositioning is a promising strategy for discovering new therapeutic strategies for cancer therapy. We investigated psychotropic drugs for their antitumor activity because of several epidemiological studies reporting lower cancer incidence in individuals receiving long term drug treatment. Experimental Approach: We investigated 27 psychotropic drugs for their cytotoxic activity in colorectal carcinoma, glioblastoma and breast cancer cell lines. Consistent with the cationic amphiphilic structure of the most cytotoxic compounds, we investigated their effect on mitochondrial and lysosomal compartments. Results: Penfluridol, ebastine, pimozide and fluoxetine, fluspirilene and nefazodone showed significant cytotoxicity, in the low micromolar range, in all cell lines tested. In MCF7 cells these drugs caused mitochondrial membrane depolarization, increased the acidic vesicular compartments and induced phospholipidosis. Both penfluridol and spiperone induced AMPK activation and autophagy. Neither caspase nor autophagy inhibitors rescued cells from death induced by ebastine, fluoxetine, fluspirilene and nefazodone. Treatment with 3-methyladenine partially rescued cell death induced by pimozide and spiperone, whereas enhanced the cytotoxic activity of penfluridol. Conversely, inhibition of lysosomal cathepsins significantly reduced cell death induced by ebastin, penfluridol, pimozide, spiperone and mildly in fluoxetine treated cells. Lastly, Spiperone cytotoxicity was restricted to colorectal cancer and breast cancer and caused apoptotic cell death in MCF7 cells. Conclusions: The cytotoxicity of psychotropic drugs with cationic amphiphilic structures relied on simultaneous mitochondrial and lysosomal disruption and induction of cell death that not necessarily requires apoptosis. Since dual targeting of lysosomes and mitochondria constitutes a new promising therapeutic approach for cancer, particularly those in which the apoptotic machinery is defective, these data further support their clinical development for cancer therapy.
Collapse
Affiliation(s)
- Marco Varalda
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,UPO Biobank, University of Piemonte Orientale, Novara, Italy
| | - Annamaria Antona
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valentina Bettio
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,UPO Biobank, University of Piemonte Orientale, Novara, Italy
| | - Konkonika Roy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Ajay Vachamaram
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Vaibhav Yellenki
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Alberto Massarotti
- Department Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Gianluca Baldanzi
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Daniela Capello
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,UPO Biobank, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
67
|
Mondal B, Pandey B, Parekh N, Panda S, Dutta T, Padhy A, Sen Gupta S. Amphiphilic mannose-6-phosphate glycopolypeptide-based bioactive and responsive self-assembled nanostructures for controlled and targeted lysosomal cargo delivery. Biomater Sci 2020; 8:6322-6336. [PMID: 33025968 DOI: 10.1039/d0bm01469a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Receptors of carbohydrate mannose-6-phosphate (M6P) are overexpressed in specific cancer cells (such as breast cancer) and are also involved in the trafficking of mannose-6-phosphate labeled proteins exclusively onto lysosomes via cell surface M6P receptor (CI-MPR) mediated endocytosis. Herein, for the first time, mannose-6-phosphate glycopolypeptide (M6PGP)-based bioactive and stimuli-responsive nanocarriers are reported. They are selectively taken up via receptor-mediated endocytosis, and trafficked to lysosomes where they are subsequently degraded by pH or enzymes, leading to the release of the cargo inside the lysosomes. Two different amphiphilic M6P block copolymers M6PGP15-APPO44 and M6PGP15-(PCL25)2 were synthesized by click reaction of the alkyne end-functionalized M6PGP15 with pH-responsive biocompatible azide end-functionalized acetal PPO and azide end-functionalized branched PCL, respectively. In water, the amphiphilic M6P-glycopolypeptide block copolymers self-assembled into micellar nanostructures, as was evidenced by DLS, TEM, AFM, and fluorescence spectroscopy techniques. These micellar systems were competent to encapsulate the hydrophobic dye rhodamine-B-octadecyl ester, which was used as the model drug. They were stable at physiological pH but were found to disassemble at acidic pH (for M6PGP15-APPO44) or in the presence of esterase (for M6PGP15-(PCL25)2). These M6PGP based micellar nanoparticles can selectively target lysosomes in cancerous cells such as MCF-7 and MDA-MB-231. Finally, we demonstrate the clathrin-mediated endocytic pathway of the native FL-M6PGP polymer and RBOE loaded M6PGP micellar-nanocarriers, and selective trafficking of MCF-7 and MDA-MB-231 breast cancer cell lysosomes, demonstrating their potential applicability toward receptor-mediated lysosomal cargo delivery.
Collapse
Affiliation(s)
- Basudeb Mondal
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata-741246, India.
| | | | | | | | | | | | | |
Collapse
|
68
|
Drechsel J, Kyrousi C, Cappello S, Sieber SA. Tranylcypromine specificity for monoamine oxidase is limited by promiscuous protein labelling and lysosomal trapping. RSC Chem Biol 2020; 1:209-213. [PMID: 34458760 PMCID: PMC8341850 DOI: 10.1039/d0cb00048e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/24/2020] [Indexed: 11/21/2022] Open
Abstract
Monoamine oxidases MAOA and MAOB catalyze important cellular functions such as the deamination of neurotransmitters. Correspondingly, MAO inhibitors are used for the treatment of severe neuropsychiatric disorders such as depression. A commonly prescribed drug against refractory depression is tranylcypromine, however, the side effects are poorly understood. In order to decipher putative off-targets, we synthesized two tranylcypromine probes equipped with either an alkyne moiety or an alkyne-diazirine minimal photocrosslinker for in situ proteome profiling. Surprisingly, LC–MS/MS analysis revealed low enrichment of MAOA and relatively promiscuous labeling of proteins. Photoprobe labeling paired with fluorescent imaging studies revealed lysosomal trapping which could be largely reverted by the addition of lysosomotropic drugs. Chemical proteomics and cellular imaging reveal lysosomal trapping of tranylcypromine which can be largely reverted by the addition of lysosomotropic drugs.![]()
Collapse
Affiliation(s)
- Jonas Drechsel
- Department of Chemistry, Technical University of Munich Lichtenbergstraße 4 85748 Garching Germany
| | - Christina Kyrousi
- Max Planck Institute of Psychiatry Kraepelinstraße 2 80804 Munich Germany
| | - Silvia Cappello
- Max Planck Institute of Psychiatry Kraepelinstraße 2 80804 Munich Germany
| | - Stephan A Sieber
- Department of Chemistry, Technical University of Munich Lichtenbergstraße 4 85748 Garching Germany
| |
Collapse
|
69
|
Verma DK, Ghosh A, Ruggiero L, Cartier E, Janezic E, Williams D, Jung EG, Moore M, Seo JB, Kim YH. The SUMO Conjugase Ubc9 Protects Dopaminergic Cells from Cytotoxicity and Enhances the Stability of α-Synuclein in Parkinson's Disease Models. eNeuro 2020; 7:ENEURO.0134-20.2020. [PMID: 32887693 PMCID: PMC7519168 DOI: 10.1523/eneuro.0134-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/23/2022] Open
Abstract
Small ubiquitin-like modifier (SUMO) is a widespread regulatory mechanism of post-translational modification (PTM) that induces rapid and reversible changes in protein function and stability. Using SUMO conjugase Ubc9-overexpressing or knock-down cells in Parkinson's disease (PD) models, we demonstrate that SUMOylation protects dopaminergic cells against MPP+ or preformed fibrils (PFFs) of α-synuclein (α-syn)-induced toxicities in cell viability and cytotoxicity assays. In the mechanism of protection, Ubc9 overexpression significantly suppressed the MPP+ or PFF-induced reactive oxygen species (ROS) generation, while Ubc9-RNAi enhanced the toxicity-induced ROS production. Further, PFF-mediated protein aggregation was exacerbated by Ubc9-RNAi in thioflavin T staining, compared with NC1 controls. In cycloheximide (Chx)-based protein stability assays, higher protein level of α-syn was identified in Ubc9-enhanced green fluorescent protein (EGFP) than in EGFP cells. Since there was no difference in endogenous mRNA levels of α-syn between Ubc9 and EGFP cells in quantitative real-time PCR (qRT-PCR), we assessed the mechanisms of SUMO-mediated delayed α-syn degradation via MG132, proteasomal inhibitor, and PMA, lysosomal degradation inducer. Ubc9-mediated SUMOylated α-syn avoided PMA-induced lysosomal degradation because of its high solubility. Our results suggest that Ubc9 enhances the levels of SUMO1 and ubiquitin on α-syn and interrupts SUMO1 removal from α-syn. In immunohistochemistry, dopaminergic axon tips in the striatum and cell bodies in the substantia nigra from Ubc9-overexpressing transgenic mice were protected from MPTP toxicities compared with wild-type (WT) siblings. Our results support that SUMOylation can be a regulatory target to protect dopaminergic neurons from oxidative stress and protein aggregation, with the implication that high levels of SUMOylation in dopaminergic neurons can prevent the pathologic progression of PD.
Collapse
Affiliation(s)
- Dinesh Kumar Verma
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| | - Anurupa Ghosh
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| | - Lindsey Ruggiero
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| | - Etienne Cartier
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| | - Eric Janezic
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| | - Dionne Williams
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| | - Eui-Gil Jung
- Seoul Center, Korea Basic Science Institute, Seoul 02841, Republic of Korea
| | - Michael Moore
- Imaging Core, Delaware State University, Dover, DE 19901
| | - Jong Bok Seo
- Seoul Center, Korea Basic Science Institute, Seoul 02841, Republic of Korea
| | - Yong-Hwan Kim
- Department of Biological Sciences/Neuroscience program, Delaware State University, Dover, DE 19901
| |
Collapse
|
70
|
Klouda CB, Stone WL. Oxidative Stress, Proton Fluxes, and Chloroquine/Hydroxychloroquine Treatment for COVID-19. Antioxidants (Basel) 2020; 9:E894. [PMID: 32967165 PMCID: PMC7555760 DOI: 10.3390/antiox9090894] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) have been proposed as treatments for COVID-19. These drugs have been studied for many decades, primarily in the context of their use as antimalarials, where they induce oxidative stress-killing of the malarial parasite. Less appreciated, however, is evidence showing that CQ/HCQ causes systemic oxidative stress. In vitro and observational data suggest that CQ/HCQ can be repurposed as potential antiviral medications. This review focuses on the potential health concerns of CQ/HCQ induced by oxidative stress, particularly in the hyperinflammatory stage of COVID-19 disease. The pathophysiological role of oxidative stress in acute respiratory distress syndrome (ARDS) has been well-documented. Additional oxidative stress caused by CQ/HCQ during ARDS could be problematic. In vitro data showing that CQ forms a complex with free-heme that promotes lipid peroxidation of phospholipid bilayers are also relevant to COVID-19. Free-heme induced oxidative stress is implicated as a systemic activator of coagulation, which is increasingly recognized as a contributor to COVID-19 morbidity. This review will also provide a brief overview of CQ/HCQ pharmacology with an emphasis on how these drugs alter proton fluxes in subcellular organelles. CQ/HCQ-induced alterations in proton fluxes influence the type and chemical reactivity of reactive oxygen species (ROS).
Collapse
Affiliation(s)
| | - William L. Stone
- Department of Pediatrics, East Tennessee State University, Johnson City, TN 37614, USA;
| |
Collapse
|
71
|
Li Y, Hu D, Qi J, Cui S, Chen W. Lysosomal Reacidification Ameliorates Vinyl Carbamate-Induced Toxicity and Disruption on Lysosomal pH. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8951-8961. [PMID: 32806125 DOI: 10.1021/acs.jafc.0c00534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Ethyl carbamate (EC) is a carcinogen toxicant, commonly found in fermented foods and beverages. The carcinogenic and toxic possibility of EC is thought to be related to its metabolite vinyl carbamate (VC). However, we found interesting mechanisms underlying VC-induced toxicity in this study, which were greatly different from EC. We first conducted a simple synthesis procedure for VC and found that VC possessed higher toxicity but failed to regulate levels of reactive oxygen species, glutathione, and autophagy. Notably, VC treatment resulted in upregulation of lysosomal pH, which was responsible for its cytotoxicity. Cyclic adenosine monophosphate (cAMP) pretreatment could enhance restoration of lysosomal acidity and ameliorate VC-induced damage. Inhibition of protein kinase A and cystic fibrosis transmembrane conductance regulator can block cAMP-induced cytoprotection. Together, our results provided the evidence for novel mechanisms of toxicity and possible protection method under VC exposure, which might give new perspectives on the study of EC-induced toxicity.
Collapse
Affiliation(s)
- Yuting Li
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Dongwen Hu
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Jifeng Qi
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Chen
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| |
Collapse
|
72
|
Borggaard XG, Pirapaharan DC, Delaissé JM, Søe K. Osteoclasts' Ability to Generate Trenches Rather Than Pits Depends on High Levels of Active Cathepsin K and Efficient Clearance of Resorption Products. Int J Mol Sci 2020; 21:ijms21165924. [PMID: 32824687 PMCID: PMC7460581 DOI: 10.3390/ijms21165924] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022] Open
Abstract
Until recently, it was well-accepted that osteoclasts resorb bone according to the resorption cycle model. This model is based on the assumption that osteoclasts are immobile during bone erosion, allowing the actin ring to be firmly attached and thereby provide an effective seal encircling the resorptive compartment. However, through time-lapse, it was recently documented that osteoclasts making elongated resorption cavities and trenches move across the bone surface while efficiently resorbing bone. However, it was also shown that osteoclasts making rounded cavities and pits indeed resorb bone while they are immobile. Only little is known about what distinguishes these two different resorption modes. This is of both basic and clinical interest because these resorption modes are differently sensitive to drugs and are affected by the gender as well as age of the donor. In the present manuscript we show that: 1. levels of active cathepsin K determine the switch from pit to trench mode; 2. pit and trench mode depend on clathrin-mediated endocytosis; and 3. a mechanism integrating release of resorption products and membrane/integrin recycling is required for prolongation of trench mode. Our study therefore contributes to an improved understanding of the molecular and cellular determinants for the two osteoclastic bone resorption modes.
Collapse
Affiliation(s)
- Xenia G. Borggaard
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
- Correspondence: (X.G.B.); (K.S.); Tel.: +45-65413190 (K.S.)
| | - Dinisha C. Pirapaharan
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Jean-Marie Delaissé
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Kent Søe
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
- Correspondence: (X.G.B.); (K.S.); Tel.: +45-65413190 (K.S.)
| |
Collapse
|
73
|
Abstract
Lysosomes are acidic, dynamic organelles that supervise catabolism, integrate signaling cascades, and tune cellular trafficking. Moreover, the loss of their integrity may jeopardize cell viability. In cancer cells, lysosomes are qualitatively and quantitatively modified for the tumor's own benefit. For all these reasons, these organelles emerge as appealing intracellular targets to manipulate non-oncogene addiction. This is of particular interest for brain diseases, including neurodegenerative disorders and cancer, in which stem cells are exhausted and transformed, respectively. Recent publications had demonstrated that stem cells displayed disarmed lysosomes in terms of number and functions during aging and oncogenic progression. Likewise, our laboratory identified that the arginine protease MALT1, normally dedicated to the assembly of proper NF-kB activation and processing a number of substrates, arbitrates lysosome biogenesis and mTOR signaling in glioblastoma stem-like cells. Indeed, blocking either the expression or the activity of this enzyme leads to an aberrant increase of lysosomes, alongside of the down-regulation of the mTOR signaling. This surge of lysosomes eradicates glioblastoma stem-like cells. Targeting lysosomes might thus inspire the design of new strategies to face this devastating human cancer. Here, we provide an overview of the functions of the lysosome as well as its role as a cell death initiator, to highlight the potential of lysosomal drugs for glioblastoma therapy.
Collapse
Affiliation(s)
- Kathryn A Jacobs
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France
| | - Clément Maghe
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France
| | - Julie Gavard
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France.,Integrated Center for Oncology, ICO , St. Herblain, France
| |
Collapse
|
74
|
Homolak J, Kodvanj I. Widely available lysosome targeting agents should be considered as potential therapy for COVID-19. Int J Antimicrob Agents 2020; 56:106044. [PMID: 32522674 PMCID: PMC7275137 DOI: 10.1016/j.ijantimicag.2020.106044] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/21/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023]
Abstract
Lysosome targeting agents can disrupt endolysosomal maturation and inhibit viral replication Lysosome targeting agents and lysosomotropic drugs should be explored as antiviral drugs for severe acute respiratory syndrome coronavirus-2 Some lysosomotropic drugs are commonly used pharmacological agents Particular attention should be directed towards macrolides (e.g. azithromycin), non-steroidal anti-inflammatory drugs (e.g. indomethacin) and other lysosomotoropic agents
While the coronavirus disease 2019 (COVID-19) pandemic advances, the scientific community continues to struggle in the search for treatments. Several improvements have been made, including discovery of the clinical efficacy of chloroquine (CQ) in patients with COVID-19, but effective treatment protocols remain elusive. In the search for novel treatment options, many scientists have used the in-silico approach to identify compounds that could interfere with the key molecules involved in entrance, replication or dissemination of severe acute respiratory syndrome coronavirus-2. However, most of the identified molecules are not available as pharmacological agents at present, and assessment of their safety and efficacy could take many months. This review took a different approach based on the proposed pharmacodynamic model of CQ in COVID-19. The main mechanism of action responsible for the favourable outcome of patients with COVID-19 treated with CQ seems to be related to a pH-modulation-mediated effect on endolysosomal trafficking, a characteristic of chemical compounds often called ‘lysosomotropic agents’ because of the physico-chemical properties that enable them to diffuse passively through the endosomal membrane and undergo protonation-based trapping in the lumen of the acidic vesicles. This review discusses lysosomotropic and lysosome targeting drugs that are already in clinical use and are characterized by good safety profiles, low cost and wide availability. Some of these drugs –particularly azithromycin and other macrolides, indomethacin and some other non-steroidal anti-inflammatory drugs, proton pump inhibitors and fluoxetine – could provide additional therapeutic benefits in addition to the potential antiviral effect that is still to be confirmed by well-controlled clinical trials. As some of these drugs have probably been used empirically in the treatment of COVID-19, it is hoped that colleagues worldwide will publish patient data to enable evaluation of the potential efficacy of these agents in the clinical context, and rapid implementation in therapeutic protocols if they are shown to have a beneficial effect on clinical outcome.
Collapse
Affiliation(s)
- J Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb 10000, Croatia.
| | - I Kodvanj
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb 10000, Croatia
| |
Collapse
|
75
|
Naser E, Kadow S, Schumacher F, Mohamed ZH, Kappe C, Hessler G, Pollmeier B, Kleuser B, Arenz C, Becker KA, Gulbins E, Carpinteiro A. Characterization of the small molecule ARC39, a direct and specific inhibitor of acid sphingomyelinase in vitro. J Lipid Res 2020; 61:896-910. [PMID: 32156719 PMCID: PMC7269768 DOI: 10.1194/jlr.ra120000682] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/01/2020] [Indexed: 01/03/2023] Open
Abstract
Inhibition of acid sphingomyelinase (ASM), a lysosomal enzyme that catalyzes the hydrolysis of sphingomyelin into ceramide and phosphorylcholine, may serve as an investigational tool or a therapeutic intervention to control many diseases. Specific ASM inhibitors are currently not sufficiently characterized. Here, we found that 1-aminodecylidene bis-phosphonic acid (ARC39) specifically and efficiently (>90%) inhibits both lysosomal and secretory ASM in vitro. Results from investigating sphingomyelin phosphodiesterase 1 (SMPD1/Smpd1) mRNA and ASM protein levels suggested that ARC39 directly inhibits ASM's catalytic activity in cultured cells, a mechanism that differs from that of functional inhibitors of ASM. We further provide evidence that ARC39 dose- and time-dependently inhibits lysosomal ASM in intact cells, and we show that ARC39 also reduces platelet- and ASM-promoted adhesion of tumor cells. The observed toxicity of ARC39 is low at concentrations relevant for ASM inhibition in vitro, and it does not strongly alter the lysosomal compartment or induce phospholipidosis in vitro. When applied intraperitoneally in vivo, even subtoxic high doses administered short-term induced sphingomyelin accumulation only locally in the peritoneal lavage without significant accumulation in plasma, liver, spleen, or brain. These findings require further investigation with other possible chemical modifications. In conclusion, our results indicate that ARC39 potently and selectively inhibits ASM in vitro and highlight the need for developing compounds that can reach tissue concentrations sufficient for ASM inhibition in vivo.
Collapse
Affiliation(s)
- Eyad Naser
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Fabian Schumacher
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany; Department of Toxicology, Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Zainelabdeen H Mohamed
- Institute of Chemistry, Humboldt University of Berlin, 12489 Berlin, Germany; Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Christian Kappe
- Institute of Chemistry, Humboldt University of Berlin, 12489 Berlin, Germany
| | - Gabriele Hessler
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Barbara Pollmeier
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Christoph Arenz
- Institute of Chemistry, Humboldt University of Berlin, 12489 Berlin, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany; Department of Surgery, University of Cincinnati, Cincinnati, OH 45229
| | - Alexander Carpinteiro
- Department of Molecular Biology, University of Duisburg-Essen, 45147 Essen, Germany; Department of Hematology, University Hospital Essen, 45147 Essen, Germany. mailto:
| |
Collapse
|
76
|
Lesire L, Chaput L, Cruz De Casas P, Rousseau F, Piveteau C, Dumont J, Pointu D, Déprez B, Leroux F. High-Throughput Image-Based Aggresome Quantification. SLAS DISCOVERY 2020; 25:783-791. [PMID: 32449635 DOI: 10.1177/2472555220919708] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aggresomes are subcellular perinuclear structures where misfolded proteins accumulate by retrograde transport on microtubules. Different methods are available to monitor aggresome formation, but they are often laborious, time-consuming, and not quantitative. Proteostat is a red fluorescent molecular rotor dye, which becomes brightly fluorescent when it binds to protein aggregates. As this reagent was previously validated to detect aggresomes, we have miniaturized its use in 384-well plates and developed a method for high-throughput imaging and quantification of aggresomes. Two different image analysis methods, including one with machine learning, were evaluated. They lead to similar robust data to quantify cells having aggresome, with satisfactory Z' factor values and reproducible EC50 values for compounds known to induce aggresome formation, like proteasome inhibitors. We demonstrated the relevance of this phenotypic assay by screening a chemical library of 1280 compounds to find aggresome modulators. We obtained hits that present similarities in their structural and physicochemical properties. Interestingly, some of them were previously described to modulate autophagy, which could explain their effect on aggresome structures. In summary, we have optimized and validated the Proteostat detection reagent to easily measure aggresome formation in a miniaturized, automated, quantitative, and high-content assay. This assay can be used at low, middle, or high throughput to quantify changes in aggresome formation that could help in the understanding of chemical compound activity in pathologies such as protein misfolding disorders or cancer.
Collapse
Affiliation(s)
- Laetitia Lesire
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - Ludovic Chaput
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - Paulina Cruz De Casas
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - Fanny Rousseau
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - Catherine Piveteau
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - Julie Dumont
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - David Pointu
- Succursale France, GE Healthcare Europe GmbH, Vélizy-Villacoublay, France
| | - Benoît Déprez
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| | - Florence Leroux
- Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, University of Lille, Lille, France
| |
Collapse
|
77
|
Subramanian K, Hutt DM, Scott SM, Gupta V, Mao S, Balch WE. Correction of Niemann-Pick type C1 trafficking and activity with the histone deacetylase inhibitor valproic acid. J Biol Chem 2020; 295:8017-8035. [PMID: 32354745 DOI: 10.1074/jbc.ra119.010524] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Niemann-Pick type C (NPC) disease is primarily caused by mutations in the NPC1 gene and is characterized by the accumulation of unesterified cholesterol and lipids in the late endosomal (LE) and lysosomal (Ly) compartments. The most prevalent disease-linked mutation is the I1061T variant of NPC1, which exhibits defective folding and trafficking from the endoplasmic reticulum to the LE/Ly compartments. We now show that the FDA-approved histone deacetylase inhibitor (HDACi) valproic acid (VPA) corrects the folding and trafficking defect associated with I1061T-NPC1 leading to restoration of cholesterol homeostasis, an effect that is largely driven by a reduction in HDAC7 expression. The VPA-mediated trafficking correction is in part associated with an increase in the acetylation of lysine residues in the cysteine-rich domain of NPC1. The HDACi-mediated correction is synergistically improved by combining it with the FDA-approved anti-malarial, chloroquine, a known lysosomotropic compound, which improved the stability of the LE/Ly-localized fraction of the I1061T variant. We posit that combining the activity of VPA, to modulate epigenetically the cellular acetylome, with chloroquine, to alter the lysosomal environment to favor stability of the trafficked I1061T variant protein can have a significant therapeutic benefit in patients carrying at least one copy of the I1061T variant of NPC1, the most common disease-associated mutation leading to NPC disease. Given its ability to cross the blood-brain barrier, we posit VPA provides a potential mechanism to improve the response to 2-hydroxypropyl-β-cyclodextrin, by restoring a functional NPC1 to the cholesterol managing compartment as an adjunct therapy.
Collapse
Affiliation(s)
| | - Darren M Hutt
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Samantha M Scott
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Vijay Gupta
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Shu Mao
- Department of Biochemistry, Weill Cornell Medical College, New York, New York, USA
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
78
|
Auguste M, Balbi T, Ciacci C, Canonico B, Papa S, Borello A, Vezzulli L, Canesi L. Shift in Immune Parameters After Repeated Exposure to Nanoplastics in the Marine Bivalve Mytilus. Front Immunol 2020; 11:426. [PMID: 32351496 PMCID: PMC7174705 DOI: 10.3389/fimmu.2020.00426] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/25/2020] [Indexed: 12/14/2022] Open
Abstract
Bivalves are widespread in coastal environments subjected to a wide range of environmental fluctuations: however, the rapidly occurring changes due to several anthropogenic factors can represent a significant threat to bivalve immunity. The mussel Mytilus spp. has extremely powerful immune defenses toward different potential pathogens and contaminant stressors. In particular, the mussel immune system represents a significant target for different types of nanoparticles (NPs), including amino-modified nanopolystyrene (PS-NH2) as a model of nanoplastics. In this work, the effects of repeated exposure to PS-NH2 on immune responses of Mytilus galloprovincialis were investigated after a first exposure (10 μg/L; 24 h), followed by a resting period (72-h depuration) and a second exposure (10 μg/L; 24 h). Functional parameters were measured in hemocytes, serum, and whole hemolymph samples. In hemocytes, transcription of selected genes involved in proliferation/apoptosis and immune response was evaluated by qPCR. First exposure to PS-NH2 significantly affected hemocyte mitochondrial and lysosomal parameters, serum lysozyme activity, and transcription of proliferation/apoptosis markers; significant upregulation of extrapallial protein precursor (EPp) and downregulation of lysozyme and mytilin B were observed. The results of functional hemocyte parameters indicate the occurrence of stress conditions that did not however result in changes in the overall bactericidal activity. After the second exposure, a shift in hemocyte subpopulations, together with reestablishment of basal functional parameters and of proliferation/apoptotic markers, was observed. Moreover, hemolymph bactericidal activity, as well as transcription of five out of six immune-related genes, all codifying for secreted proteins, was significantly increased. The results indicate an overall shift in immune parameters that may act as compensatory mechanisms to maintain immune homeostasis after a second encounter with PS-NH2.
Collapse
Affiliation(s)
- Manon Auguste
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Teresa Balbi
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Caterina Ciacci
- Department of Biomolecular Sciences (DIBS), University of Urbino, Urbino, Italy
| | - Barbara Canonico
- Department of Biomolecular Sciences (DIBS), University of Urbino, Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences (DIBS), University of Urbino, Urbino, Italy
| | - Alessio Borello
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Luigi Vezzulli
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Laura Canesi
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| |
Collapse
|
79
|
Karim MR, Fisher CR, Kapphahn RJ, Polanco JR, Ferrington DA. Investigating AKT activation and autophagy in immunoproteasome-deficient retinal cells. PLoS One 2020; 15:e0231212. [PMID: 32275682 PMCID: PMC7147741 DOI: 10.1371/journal.pone.0231212] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/18/2020] [Indexed: 12/04/2022] Open
Abstract
Two major proteolytic systems, the proteasome and the autophagy pathway, are key components of the proteostasis network. The immunoproteasome, a proteasome subtype, and autophagy are upregulated under stress conditions, forming a coordinated unit designed to minimize the effect of cell stress. We investigated how genetic ablation of the LMP2 immunoproteasome subunit affects autophagy in retinal pigment epithelium (RPE) from WT and LMP2 knockout mice. We monitored autophagy regulation by measuring LC3, phosphorylation of AKT (S473), and phosphorylation of S6, a downstream readout of AKT (mTOR) pathway activation. We also evaluated transcription factor EB (TFEB) nuclear translocation, a transcription factor that controls expression of autophagy and lysosome genes. WT and LMP2 KO cells were monitored after treatment with EBSS to stimulate autophagy, insulin to stimulate AKT, or an AKT inhibitor (trehalose or MK-2206). Under basal conditions, we observed hyper-phosphorylation of AKT and S6, as well as lower nuclear-TFEB content in LMP2 KO RPE compared with WT. AKT inhibitors MK-2206 and trehalose significantly inhibited AKT phosphorylation and stimulated nuclear translocation of TFEB. Starvation and AKT inhibition upregulated autophagy, albeit to a lesser extent in LMP2 KO RPE. These data support the idea that AKT hyper-activation is an underlying cause of defective autophagy regulation in LMP2 KO RPE, revealing a unique link between two proteolytic systems and a previously unknown function in autophagy regulation by the immunoproteasome.
Collapse
Affiliation(s)
- Md. Razaul Karim
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cody R. Fisher
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
- Graduate Program in Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rebecca J. Kapphahn
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jorge R. Polanco
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Deborah A. Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
- Graduate Program in Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
80
|
Zhao B, Dierichs L, Gu JN, Trajkovic-Arsic M, Axel Hilger R, Savvatakis K, Vega-Rubin-de-Celis S, Liffers ST, Peña-Llopis S, Behrens D, Hahn S, Siveke JT, Lueong SS. TFEB-mediated lysosomal biogenesis and lysosomal drug sequestration confer resistance to MEK inhibition in pancreatic cancer. Cell Death Discov 2020; 6:12. [PMID: 32194992 PMCID: PMC7066197 DOI: 10.1038/s41420-020-0246-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 01/06/2023] Open
Abstract
Oncogenic KRAS mutations are encountered in more than 90% of pancreatic ductal adenocarcinomas. MEK inhibition has failed to procure any clinical benefits in mutant RAS-driven cancers including pancreatic ductal adenocarcinoma (PDAC). To identify potential resistance mechanisms underlying MEK inhibitor (MEKi) resistance in PDAC, we investigated lysosomal drug accumulation in PDAC models both in vitro and in vivo. Mouse PDAC models and human PDAC cell lines as well as human PDAC xenografts treated with the MEK inhibitor trametinib or refametinib led to an enhanced expression of lysosomal markers and enrichment of lysosomal gene sets. A time-dependent, increase in lysosomal content was observed upon MEK inhibition. Strikingly, there was a strong activation of lysosomal biogenesis in cell lines of the classical compared to the basal-like molecular subtype. Increase in lysosomal content was associated with nuclear translocation of the Transcription Factor EB (TFEB) and upregulation of TFEB target genes. siRNA-mediated depletion of TFEB led to a decreased lysosomal biogenesis upon MEK inhibition and potentiated sensitivity. Using LC-MS, we show accumulation of MEKi in the lysosomes of treated cells. Therefore, MEK inhibition triggers lysosomal biogenesis and subsequent drug sequestration. Combined targeting of MEK and lysosomal function may improve sensitivity to MEK inhibition in PDAC.
Collapse
Affiliation(s)
- Ben Zhao
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| | - Laura Dierichs
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| | - Jiang-Ning Gu
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province China
| | - Marija Trajkovic-Arsic
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| | - Ralf Axel Hilger
- Dept Med Oncol, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Konstantinos Savvatakis
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| | | | - Sven-Thorsten Liffers
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| | - Samuel Peña-Llopis
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
- Translational Genomics in Solid Tumors, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Diana Behrens
- EPO – Experimental Pharmacology and Oncology GmbH Berlin-Buch, Berlin, Germany
| | - Stephan Hahn
- Department of Molecular GI-Oncology, Rurh University Bochum, Bochum, Germany
| | - Jens T. Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| | - Smiths S. Lueong
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany, Essen, Germany
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany, Heidelberg, Germany
| |
Collapse
|
81
|
Po WW, Thein W, Khin PP, Khing TM, Han KWW, Park CH, Sohn UD. Fluoxetine Simultaneously Induces Both Apoptosis and Autophagy in Human Gastric Adenocarcinoma Cells. Biomol Ther (Seoul) 2020; 28:202-210. [PMID: 31522488 PMCID: PMC7059812 DOI: 10.4062/biomolther.2019.103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 01/14/2023] Open
Abstract
Fluoxetine is used widely as an antidepressant for the treatment of cancer-related depression, but has been reported to also have anti-cancer activity. In this study, we investigated the cytotoxicity of fluoxetine to human gastric adenocarcinoma cells; as shown by the MTT assay, fluoxetine induced cell death. Subsequently, cells were treated with 10 or 20 µM fluoxetine for 24 h and analyzed. Apoptosis was confirmed by the increased number of early apoptotic cells, shown by Annexin V- propidium iodide staining. Nuclear condensation was visualized by DAPI staining. A significant increase in the expression of cleaved PARP was observed by western blotting. The pan-caspase inhibitor Z-VAD-FMK was used to detect the extent of caspase-dependent cell death. The induction of autophagy was determined by the formation of acidic vesicular organelles (AVOs), which was visualized by acridine orange staining, and the increased expression of autophagy markers, such as LC3B, Beclin 1, and p62/SQSTM 1, observed by western blotting. The expression of upstream proteins, such as p-Akt and p-mTOR, were decreased. Autophagic degradation was evaluated by using bafilomycin, an inhibitor of late-stage autophagy. Bafilomycin did not significantly enhance LC3B expression induced by fluoxetine, which suggested autophagic degradation was impaired. In addition, the co-administration of the autophagy inhibitor 3-methyladenine and fluoxetine significantly increased fluoxetine-induced apoptosis, with decreased p-Akt and markedly increased death receptor 4 and 5 expression. Our results suggested that fluoxetine simultaneously induced both protective autophagy and apoptosis and that the inhibition of autophagy enhanced fluoxetine-induced apoptosis through increased death receptor expression.
Collapse
Affiliation(s)
- Wah Wah Po
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Wynn Thein
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Phyu Phyu Khin
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Tin Myo Khing
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Khin Wah Wah Han
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Chan Hee Park
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.,Center for Metareceptome Research, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Uy Dong Sohn
- Laboratory of Signalling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
82
|
Laraia L, Garivet G, Foley DJ, Kaiser N, Müller S, Zinken S, Pinkert T, Wilke J, Corkery D, Pahl A, Sievers S, Janning P, Arenz C, Wu Y, Rodriguez R, Waldmann H. Image‐Based Morphological Profiling Identifies a Lysosomotropic, Iron‐Sequestering Autophagy Inhibitor. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201913712] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Luca Laraia
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- current address: Technical University of Denmark Department of Chemistry Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Guillaume Garivet
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Daniel J. Foley
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- current address: School of Physical and Chemical Sciences University of Canterbury Christchurch New Zealand
| | - Nadine Kaiser
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Sebastian Müller
- Institut Curie CNRS UMR 3666 INSERM U1143 PSL University Paris Chemical Cell Biology Group 26 Rue d'Ulm 75248 Paris Cedex 05 France
| | - Sarah Zinken
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Thomas Pinkert
- Institut für Chemie der Humboldt-Universität zu Berlin Brook-Taylor-Str. 2 (R 1'102) 12489 Berlin Germany
| | - Julian Wilke
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Dale Corkery
- Department of Chemistry Umeå Universitet KB.A4, Linnaeus väg 10 (rum: A4.35.07) 90187 Umeå Sweden
| | - Axel Pahl
- Compound Management and Screening Center, Dortmund Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Sonja Sievers
- Compound Management and Screening Center, Dortmund Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Petra Janning
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Christoph Arenz
- Institut für Chemie der Humboldt-Universität zu Berlin Brook-Taylor-Str. 2 (R 1'102) 12489 Berlin Germany
| | - Yaowen Wu
- Department of Chemistry Umeå Universitet KB.A4, Linnaeus väg 10 (rum: A4.35.07) 90187 Umeå Sweden
| | - Raphaël Rodriguez
- Institut Curie CNRS UMR 3666 INSERM U1143 PSL University Paris Chemical Cell Biology Group 26 Rue d'Ulm 75248 Paris Cedex 05 France
| | - Herbert Waldmann
- Department of Chemical Biology Max-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology TU Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| |
Collapse
|
83
|
Laraia L, Garivet G, Foley DJ, Kaiser N, Müller S, Zinken S, Pinkert T, Wilke J, Corkery D, Pahl A, Sievers S, Janning P, Arenz C, Wu Y, Rodriguez R, Waldmann H. Image-Based Morphological Profiling Identifies a Lysosomotropic, Iron-Sequestering Autophagy Inhibitor. Angew Chem Int Ed Engl 2020; 59:5721-5729. [PMID: 31769920 PMCID: PMC7154763 DOI: 10.1002/anie.201913712] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Indexed: 01/15/2023]
Abstract
Chemical proteomics is widely applied in small-molecule target identification. However, in general it does not identify non-protein small-molecule targets, and thus, alternative methods for target identification are in high demand. We report the discovery of the autophagy inhibitor autoquin and the identification of its molecular mode of action using image-based morphological profiling in the cell painting assay. A compound-induced fingerprint representing changes in 579 cellular parameters revealed that autoquin accumulates in lysosomes and inhibits their fusion with autophagosomes. In addition, autoquin sequesters Fe2+ in lysosomes, resulting in an increase of lysosomal reactive oxygen species and ultimately cell death. Such a mechanism of action would have been challenging to unravel by current methods. This work demonstrates the potential of the cell painting assay to deconvolute modes of action of small molecules, warranting wider application in chemical biology.
Collapse
Affiliation(s)
- Luca Laraia
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,current address: Technical University of Denmark, Department of Chemistry, Kemitorvet 207, 2800 Kgs., Lyngby, Denmark
| | - Guillaume Garivet
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Daniel J Foley
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,current address: School of Physical and Chemical Sciences, University of Canterbury, Christchurch, New Zealand
| | - Nadine Kaiser
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Sebastian Müller
- Institut Curie, CNRS UMR 3666, INSERM U1143, PSL University Paris, Chemical Cell Biology Group, 26 Rue d'Ulm, 75248, Paris Cedex 05, France
| | - Sarah Zinken
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Thomas Pinkert
- Institut für Chemie der, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2 (R 1'102), 12489, Berlin, Germany
| | - Julian Wilke
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Dale Corkery
- Department of Chemistry, Umeå Universitet, KB.A4, Linnaeus väg 10 (rum: A4.35.07), 90187, Umeå, Sweden
| | - Axel Pahl
- Compound Management and Screening Center, Dortmund, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Sonja Sievers
- Compound Management and Screening Center, Dortmund, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Petra Janning
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| | - Christoph Arenz
- Institut für Chemie der, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2 (R 1'102), 12489, Berlin, Germany
| | - Yaowen Wu
- Department of Chemistry, Umeå Universitet, KB.A4, Linnaeus väg 10 (rum: A4.35.07), 90187, Umeå, Sweden
| | - Raphaël Rodriguez
- Institut Curie, CNRS UMR 3666, INSERM U1143, PSL University Paris, Chemical Cell Biology Group, 26 Rue d'Ulm, 75248, Paris Cedex 05, France
| | - Herbert Waldmann
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227, Dortmund, Germany.,Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44227, Dortmund, Germany
| |
Collapse
|
84
|
Krempaska K, Barnowski S, Gavini J, Hobi N, Ebener S, Simillion C, Stokes A, Schliep R, Knudsen L, Geiser TK, Funke-Chambour M. Azithromycin has enhanced effects on lung fibroblasts from idiopathic pulmonary fibrosis (IPF) patients compared to controls [corrected]. Respir Res 2020; 21:25. [PMID: 31941499 PMCID: PMC6964061 DOI: 10.1186/s12931-020-1275-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/01/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic fatal lung disease without a cure and new drug strategies are urgently needed. Differences in behavior between diseased and healthy cells are well known and drug response can be different between cells isolated from IPF patients and controls. The macrolide Azithromycin (AZT) has anti-inflammatory and immunomodulatory properties. Recently anti-fibrotic effects have been described. However, the anti-fibrotic effects on primary IPF-fibroblasts (FB) directly compared to control-FB are unknown. We hypothesized that IPF-FB react differently to AZT in terms of anti-fibrotic effects. METHODS Primary normal human lung and IPF-FB were exposed to TGF-β (5 ng/ml), Azithromycin (50 μM) alone or in combination prior to gene expression analysis. Pro-collagen Iα1 secretion was assessed by ELISA and protein expression by western blot (αSMA, Fibronectin, ATP6V1B2, LC3 AB (II/I), p62, Bcl-xL). Microarray analysis was performed to screen involved genes and pathways after Azithromycin treatment in control-FB. Apoptosis and intraluminal lysosomal pH were analyzed by flow cytometry. RESULTS AZT significantly reduced collagen secretion in TGF-β treated IPF-FB compared to TGF-β treatment alone, but not in control-FB. Pro-fibrotic gene expression was similarly reduced after AZT treatment in IPF and control-FB. P62 and LC3II/I western blot revealed impaired autophagic flux after AZT in both control and IPF-FB with significant increase of LC3II/I after AZT in control and IPF-FB, indicating enhanced autophagy inhibition. Early apoptosis was significantly higher in TGF-β treated IPF-FB compared to controls after AZT. Microarray analysis of control-FB treated with AZT revealed impaired lysosomal pathways. The ATPase and lysosomal pH regulator ATP6V0D2 was significantly less increased after additional AZT in IPF-FB compared to controls. Lysosomal function was impaired in both IPF and control FB, but pH was significantly more increased in TGF-β treated IPF-FB. CONCLUSION We report different treatment responses after AZT with enhanced anti-fibrotic and pro-apoptotic effects in IPF compared to control-FB. Possibly impaired lysosomal function contributes towards these effects. In summary, different baseline cell phenotype and behavior of IPF and control cells contribute to enhanced anti-fibrotic and pro-apoptotic effects in IPF-FB after AZT treatment and strengthen its role as a new potential anti-fibrotic compound, that should further be evaluated in clinical studies.
Collapse
Affiliation(s)
- Kristina Krempaska
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sandra Barnowski
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Jacopo Gavini
- Department of Visceral Surgery and Medicine, Department for BioMedical Research, Inselspital, Bern University Hospital and University of Bern, 3010, Bern, Switzerland
| | - Nina Hobi
- AlveoliX AG, Murtenstrasse 50, 3008, Bern, Switzerland
- ARTORG Center for Biomedical Engineering Research, Organs-on-Chip Technologies, University of Bern, Bern, Switzerland
| | - Simone Ebener
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Cedric Simillion
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bioinformatics Unit and SIB Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Andrea Stokes
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ronja Schliep
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Thomas K Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
85
|
Li B, Clohisey SM, Chia BS, Wang B, Cui A, Eisenhaure T, Schweitzer LD, Hoover P, Parkinson NJ, Nachshon A, Smith N, Regan T, Farr D, Gutmann MU, Bukhari SI, Law A, Sangesland M, Gat-Viks I, Digard P, Vasudevan S, Lingwood D, Dockrell DH, Doench JG, Baillie JK, Hacohen N. Genome-wide CRISPR screen identifies host dependency factors for influenza A virus infection. Nat Commun 2020; 11:164. [PMID: 31919360 PMCID: PMC6952391 DOI: 10.1038/s41467-019-13965-x] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/06/2019] [Indexed: 12/26/2022] Open
Abstract
Host dependency factors that are required for influenza A virus infection may serve as therapeutic targets as the virus is less likely to bypass them under drug-mediated selection pressure. Previous attempts to identify host factors have produced largely divergent results, with few overlapping hits across different studies. Here, we perform a genome-wide CRISPR/Cas9 screen and devise a new approach, meta-analysis by information content (MAIC) to systematically combine our results with prior evidence for influenza host factors. MAIC out-performs other meta-analysis methods when using our CRISPR screen as validation data. We validate the host factors, WDR7, CCDC115 and TMEM199, demonstrating that these genes are essential for viral entry and regulation of V-type ATPase assembly. We also find that CMTR1, a human mRNA cap methyltransferase, is required for efficient viral cap snatching and regulation of a cell autonomous immune response, and provides synergistic protection with the influenza endonuclease inhibitor Xofluza. Here, Li et al. perform a genome-wide CRISPR screen to identify host dependency factors for influenza A virus infection and show that the host mRNA cap methyltransferase CMTR1 is important for viral cap snatching and that it affects expression of antiviral genes.
Collapse
Affiliation(s)
- Bo Li
- Harvard University Virology Program, Harvfvard Medical School, Boston, MA02142, USA.,Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Sara M Clohisey
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Bing Shao Chia
- Harvard University Virology Program, Harvfvard Medical School, Boston, MA02142, USA.,Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Bo Wang
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Ang Cui
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA.,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, 02115, USA
| | - Thomas Eisenhaure
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | | | - Paul Hoover
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | | | - Aharon Nachshon
- School of Molecular Cell Biology and Biotechnology, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nikki Smith
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Tim Regan
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - David Farr
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Michael U Gutmann
- School of informatics, University of Edinburgh, Edinburgh, EH8 9YL, UK
| | - Syed Irfan Bukhari
- Center for Cancer Research, Massachusetts General hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Law
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Maya Sangesland
- The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, Cambridge, MA, USA
| | - Irit Gat-Viks
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA.,School of Molecular Cell Biology and Biotechnology, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Paul Digard
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Shobha Vasudevan
- Center for Cancer Research, Massachusetts General hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, Cambridge, MA, USA
| | - David H Dockrell
- MRC Center for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - John G Doench
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - J Kenneth Baillie
- Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK. .,Intensive Care Unit, Royal Infirmary Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA. .,Massachusetts General Hospital Cancer Center, Boston, MA, 02129, USA.
| |
Collapse
|
86
|
Lysosomal Fusion: An Efficient Mechanism Increasing Their Sequestration Capacity for Weak Base Drugs without Apparent Lysosomal Biogenesis. Biomolecules 2020; 10:biom10010077. [PMID: 31947839 PMCID: PMC7022710 DOI: 10.3390/biom10010077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
Lysosomal sequestration of anticancer therapeutics lowers their cytotoxic potential, reduces drug availability at target sites, and contributes to cancer resistance. Only recently has it been shown that lysosomal sequestration of weak base drugs induces lysosomal biogenesis mediated by activation of transcription factor EB (TFEB) which, in turn, enhances their accumulation capacity, thereby increasing resistance to these drugs. Here, we addressed the question of whether lysosomal biogenesis is the only mechanism that increases lysosomal sequestration capacity. We found that lysosomal sequestration of some tyrosine kinase inhibitors (TKIs), gefitinib (GF) and imatinib (IM), induced expansion of the lysosomal compartment. However, an expression analysis of lysosomal genes, including lysosome-associated membrane proteins 1, 2 (LAMP1, LAMP2), vacuolar ATPase subunit B2 (ATP6V1B2), acid phosphatase (ACP), and galactosidase beta (GLB) controlled by TFEB, did not reveal increased expression. Instead, we found that both studied TKIs, GF and IM, induced lysosomal fusion which was dependent on nicotinic acid adenine dinucleotide phosphate (NAADP) mediated Ca2+signaling. A theoretical analysis revealed that lysosomal fusion is sufficient to explain the enlargement of lysosomal sequestration capacity. In conclusion, we demonstrated that extracellular TKIs, GF and IM, induced NAADP/Ca2+ mediated lysosomal fusion, leading to enlargement of the lysosomal compartment with significantly increased sequestration capacity for these drugs without apparent lysosomal biogenesis.
Collapse
|
87
|
A Potent Autophagy Inhibitor (Lys05) Enhances the Impact of Ionizing Radiation on Human Lung Cancer Cells H1299. Int J Mol Sci 2019; 20:ijms20235881. [PMID: 31771188 PMCID: PMC6928878 DOI: 10.3390/ijms20235881] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy inhibition through small-molecule inhibitors is one of the approaches to increase the efficiency of radiotherapy in oncological patients. A new inhibitor-Lys05-with the potential to accumulate within lysosomes and to block autophagy was discovered a few years ago. Several studies have addressed its chemosensitizing effects but nothing is known about its impact in the context of ionizing radiation (IR). To describe its role in radiosensitization, we employed radioresistant human non-small cell lung carcinoma cells (H1299, p53-negative). Combined treatment of H1299 cells by Lys05 together with IR decreased cell survival in the clonogenic assay and real-time monitoring of cell growth more than either Lys05 or IR alone. Immunodetection of LC3 and p62/SQSTM1 indicated that autophagy was inhibited, which correlated with increased SQSTM1 and decreased BNIP3 gene expression determined by qRT-PCR. Fluorescence microscopy and flow cytometry uncovered an accumulation of lysosomes. Similarly, transmission electron microscopy demonstrated the accumulation of autophagosomes confirming the ability of Lys05 to potentiate autophagy inhibition in H1299 cells. We report here for the first time that Lys05 could be utilized in combination with IR as a promising future strategy in the eradication of lung cancer cells.
Collapse
|
88
|
Merarchi M, Jung YY, Fan L, Sethi G, Ahn KS. A Brief Overview of the Antitumoral Actions of Leelamine. Biomedicines 2019; 7:biomedicines7030053. [PMID: 31330969 PMCID: PMC6783843 DOI: 10.3390/biomedicines7030053] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/27/2022] Open
Abstract
For the last couple of decades, natural products, either applied singly or in conjunction with other cancer therapies including chemotherapy and radiotherapy, have allowed us to combat different types of human cancers through the inhibition of their initiation and progression. The principal sources of these useful compounds are isolated from plants that were described in traditional medicines for their curative potential. Leelamine, derived from the bark of pine trees, was previously reported as having a weak agonistic effect on cannabinoid receptors and limited inhibitory effects on pyruvate dehydrogenase kinases (PDKs). It has been reported to possess a strong lysosomotropic property; this feature enables its assembly inside the acidic compartments within a cell, such as lysosomes, which may eventually hinder endocytosis. In this review, we briefly highlight the varied antineoplastic actions of leelamine that have found implications in pharmacological research, and the numerous intracellular targets affected by this agent that can effectively negate the oncogenic process.
Collapse
Affiliation(s)
- Myriam Merarchi
- Faculty of Pharmacy, University of Paris Descartes, 75006 Paris, France
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Young Yun Jung
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Lu Fan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
89
|
Lam Yi H, Than H, Sng C, Cheong MA, Chuah C, Xiang W. Lysosome Inhibition by Mefloquine Preferentially Enhances the Cytotoxic Effects of Tyrosine Kinase Inhibitors in Blast Phase Chronic Myeloid Leukemia. Transl Oncol 2019; 12:1221-1228. [PMID: 31276961 PMCID: PMC6611990 DOI: 10.1016/j.tranon.2019.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Despite the efficacy of BCR-ABL tyrosine kinase inhibitors (TKIs) in chronic phase-chronic myeloid leukemia, the management of blast phase-chronic myeloid leukemia (BP-CML) remains a challenge. Therefore, there is an urgent need to identify alternative agents that act synergistically with BCR-ABL TKIs in BP-CML. Our results show that the anti-malarial agent, mefloquine augments the efficacy of TKIs in CML cell lines and primary CML cells in vitro, including those with the T315I mutation. This effect is selective as mefloquine is more effective in inducing apoptosis, inhibiting colony formation and self-renewal capacity of CD34+ cells derived from TKI-resistant BP-CML patients than normal cord blood (CB) CD34+ stem/progenitor cells. Notably, the combination of mefloquine and TKIs at sublethal concentrations leads to synergistic effects in CML CD34+ cells, while sparing normal CB CD34+ cells. We further demonstrate that the initial action of mefloquine in CML cells is to increase lysosomal biogenesis and activation, followed by oxidative stress, lysosomal lipid damage and functional impairment. Taken together, our work elucidates that mefloquine selectively augments the effects of TKIs in CML stem/progenitor cells by inducing lysosomal dysfunction.
Collapse
Affiliation(s)
- Hui Lam Yi
- Department of Haematology, Singapore General Hospital, Singapore
| | - Hein Than
- Department of Haematology, Singapore General Hospital, Singapore
| | - Colin Sng
- Department of Haematology, Singapore General Hospital, Singapore
| | - May Anne Cheong
- Department of Haematology, Singapore General Hospital, Singapore
| | - Charles Chuah
- Department of Haematology, Singapore General Hospital, Singapore; Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore.
| | - Wei Xiang
- Department of Haematology, Singapore General Hospital, Singapore.
| |
Collapse
|
90
|
Selyunin AS, Hutchens S, McHardy SF, Mukhopadhyay S. Tamoxifen blocks retrograde trafficking of Shiga toxin 1 and 2 and protects against lethal toxicosis. Life Sci Alliance 2019; 2:2/3/e201900439. [PMID: 31243048 PMCID: PMC6599968 DOI: 10.26508/lsa.201900439] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/18/2022] Open
Abstract
This study reports an unexpected role of late endosome–lysosome fusion in early endosome-to-Golgi trafficking of Shiga toxins and identifies tamoxifen to be a potent inhibitor of Shiga toxicosis. Shiga toxin 1 (STx1) and 2 (STx2), produced by Shiga toxin–producing Escherichia coli, cause lethal untreatable disease. The toxins invade cells via retrograde trafficking. Direct early endosome-to-Golgi transport allows the toxins to evade degradative late endosomes. Blocking toxin trafficking, particularly at the early endosome-to-Golgi step, is appealing, but transport mechanisms of the more disease-relevant STx2 are unclear. Using data from a genome-wide siRNA screen, we discovered that disruption of the fusion of late endosomes, but not autophagosomes, with lysosomes blocked the early endosome-to-Golgi transport of STx2. A subsequent screen of clinically approved lysosome-targeting drugs identified tamoxifen (TAM) to be a potent inhibitor of the trafficking and toxicity of STx1 and STx2 in cells. The protective effect was independent of estrogen receptors but dependent on the weak base property of TAM, which allowed TAM to increase endolysosomal pH and alter endosomal dynamics. Importantly, TAM treatment enhanced survival of mice injected with a lethal dose of STx1 or STx2. Thus, it may be possible to repurpose TAM for treating Shiga toxin–producing E. coli infections.
Collapse
Affiliation(s)
- Andrey S Selyunin
- Division of Pharmacology and Toxicology, College of Pharmacy; Institute for Cellular and Molecular Biology; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Steven Hutchens
- Division of Pharmacology and Toxicology, College of Pharmacy; Institute for Cellular and Molecular Biology; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Stanton F McHardy
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology and Toxicology, College of Pharmacy; Institute for Cellular and Molecular Biology; and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
91
|
Pan HY, Alamri AH, Valapala M. Nutrient deprivation and lysosomal stress induce activation of TFEB in retinal pigment epithelial cells. Cell Mol Biol Lett 2019; 24:33. [PMID: 31160892 PMCID: PMC6537441 DOI: 10.1186/s11658-019-0159-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 05/15/2019] [Indexed: 12/03/2022] Open
Abstract
Background Induction of lysosomal function and autophagy is regarded as an adaptive mechanism in response to cellular stress. The transcription factor EB (TFEB) has been identified as a master regulator of lysosomal function and autophagy. TFEB is a member of the microphthalmia family of bHLH-LZ transcription factors that includes other members such as micropthalmia-associated transcription factor (MITF), TFE3, and TFEC. TFEB controls lysosome biogenesis and autophagy by upregulation of a family of genes belonging to the Coordinated Lysosomal Expression and Regulation (CLEAR) network. Here, we investigated the expression of TFEB in cells subjected to nutrient deprivation and lysosomal stress. We studied transcriptional induction of TFEB-regulated genes in response to nutrient deprivation and lysosomal stress in retinal pigment epithelial (RPE) cells. Furthermore, we also investigated the induction of autophagy and lysosomal genes upon overexpression of constitutively active form of TFEB. Methods Expression of TFEB and MITF protein levels were evaluated in cells subjected to prolonged periods of nutrient deprivation. mRNA levels of the CLEAR network genes was measured by quantitative real time PCR (qRT-PCR) analysis in cells deprived of nutrients, treated with ammonium chloride and upon overexpression of constitutively active TFEB. Immunostaining with LC3 antibody was used to measure autophagy flux. Labeling with lysoTracker dye was used to assess lysosomes. Results Our results show that nutrient deprivation increases protein levels of TFEB and MITF in ARPE-19 cells. Nutrient stress induces the expression of lysosomal (LAMP1, CTSD MCOLN1, SGSH) and autophagy (BECN1) genes. Lysosomal stress also increases the expression of lysosomal (ATP6V0A1 and LAMP1) and autophagy (p62 and BECN1) genes. Our results show that overexpression of constitutively active TFEB also induces the expression of CLEAR network genes. Conclusions Collectively, these observations suggest that nutrient stress induces the protein expression of both MITF and TFEB in ARPE-19 cells. TFEB-regulated transcriptional program plays an important role in adaptive response of cells during both nutrient and lysosomal stress.
Collapse
Affiliation(s)
- Hsuan-Yeh Pan
- 1School of Optometry, Indiana University, Bloomington, IN 47405 USA
| | - Abdulla H Alamri
- 2State University of New York College of Optometry, 33 42nd St., New York, NY 10036 USA
| | - Mallika Valapala
- 1School of Optometry, Indiana University, Bloomington, IN 47405 USA
| |
Collapse
|
92
|
Brun S, Bassissi F, Serdjebi C, Novello M, Tracz J, Autelitano F, Guillemot M, Fabre P, Courcambeck J, Ansaldi C, Raymond E, Halfon P. GNS561, a new lysosomotropic small molecule, for the treatment of intrahepatic cholangiocarcinoma. Invest New Drugs 2019; 37:1135-1145. [DOI: 10.1007/s10637-019-00741-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/01/2019] [Indexed: 02/08/2023]
|
93
|
Englinger B, Kallus S, Senkiv J, Laemmerer A, Moser P, Gabler L, Groza D, Kowol CR, Heffeter P, Grusch M, Berger W. Lysosomal Sequestration Impairs the Activity of the Preclinical FGFR Inhibitor PD173074. Cells 2018; 7:E259. [PMID: 30544798 PMCID: PMC6315953 DOI: 10.3390/cells7120259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/29/2022] Open
Abstract
Knowledge of intracellular pharmacokinetics of anticancer agents is imperative for understanding drug efficacy as well as intrinsic and acquired cellular resistance mechanisms. However, the factors driving subcellular drug distribution are complex and poorly understood. Here, we describe for the first time the intrinsic fluorescence properties of the fibroblast growth factor receptor inhibitor PD1703074 as well as utilization of this physicochemical feature to investigate intracellular accumulation and compartmentalization of this compound in human lung cancer cells. Cell-free PD173074 fluorescence, intracellular accumulation and distribution were investigated using analytical chemistry and molecular biology approaches. Analyses on a subcellular scale revealed selective drug accumulation in lysosomes. Coincubation with inhibitors of lysosomal acidification strongly enhanced PD173074-mediated fibroblast growth factor receptor (FGFR) inhibition and cytotoxicity. In conclusion, intrinsic fluorescence enables analysis of molecular factors influencing intracellular pharmacokinetics of PD173074. Lysosome-alkalinizing agents might represent candidates for rational combination treatment, preventing cancer cell-intrinsic PD173074 resistance based on lysosomal trapping.
Collapse
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Sebastian Kallus
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, A-1090 Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", A-1090 Vienna, Austria.
| | - Julia Senkiv
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
- Institute of Cell Biology, National Academy of Sciences of Ukraine, 79005 Lviv, Ukraine.
| | - Anna Laemmerer
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Patrick Moser
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Lisa Gabler
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Diana Groza
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, A-1090 Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", A-1090 Vienna, Austria.
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", A-1090 Vienna, Austria.
| | - Michael Grusch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", A-1090 Vienna, Austria.
| |
Collapse
|
94
|
Samarani M, Loberto N, Soldà G, Straniero L, Asselta R, Duga S, Lunghi G, Zucca FA, Mauri L, Ciampa MG, Schiumarini D, Bassi R, Giussani P, Chiricozzi E, Prinetti A, Aureli M, Sonnino S. A lysosome-plasma membrane-sphingolipid axis linking lysosomal storage to cell growth arrest. FASEB J 2018; 32:5685-5702. [PMID: 29746165 PMCID: PMC6133699 DOI: 10.1096/fj.201701512rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/30/2018] [Indexed: 01/15/2023]
Abstract
Lysosomal accumulation of undegraded materials is a common feature of lysosomal storage diseases, neurodegenerative disorders, and the aging process. To better understand the role of lysosomal storage in the onset of cell damage, we used human fibroblasts loaded with sucrose as a model of lysosomal accumulation. Sucrose-loaded fibroblasts displayed increased lysosomal biogenesis followed by arrested cell proliferation. Notably, we found that reduced lysosomal catabolism and autophagy impairment led to an increase in sphingolipids ( i.e., sphingomyelin, glucosylceramide, ceramide, and the gangliosides GM3 and GD3), at both intracellular and plasma membrane (PM) levels. In addition, we observed an increase in the lysosomal membrane protein Lamp-1 on the PM of sucrose-loaded fibroblasts and a greater release of the soluble lysosomal protein cathepsin D in their extracellular medium compared with controls. These results indicate increased fusion between lysosomes and the PM, as also suggested by the increased activity of lysosomal glycosphingolipid hydrolases on the PM of sucrose-loaded fibroblasts. The inhibition of β-glucocerebrosidase and nonlysosomal glucosylceramidase, both involved in ceramide production resulting from glycosphingolipid catabolism on the PM, partially restored cell proliferation. Our findings indicate the existence of a new molecular mechanism underlying cell damage triggered by lysosomal impairment.-Samarani, M., Loberto, N., Soldà, G., Straniero, L., Asselta, R., Duga, S., Lunghi, G., Zucca, F. A., Mauri, L., Ciampa, M. G., Schiumarini, D., Bassi, R., Giussani, P., Chiricozzi, E., Prinetti, A., Aureli, M., Sonnino, S. A lysosome-plasma membrane-sphingolipid axis linking lysosomal storage to cell growth arrest.
Collapse
Affiliation(s)
- Maura Samarani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Nicoletta Loberto
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giulia Soldà
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Letizia Straniero
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Fabio A. Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Domitilla Schiumarini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Rosaria Bassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
95
|
Denzel MS, Lapierre LR, Mack HID. Emerging topics in C. elegans aging research: Transcriptional regulation, stress response and epigenetics. Mech Ageing Dev 2018; 177:4-21. [PMID: 30134144 PMCID: PMC6696993 DOI: 10.1016/j.mad.2018.08.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/13/2022]
Abstract
Key discoveries in aging research have been made possible with the use of model organisms. Caenorhabditis elegans is a short-lived nematode that has become a well-established system to study aging. The practicality and powerful genetic manipulations associated with this metazoan have revolutionized our ability to understand how organisms age. 25 years after the publication of the discovery of the daf-2 gene as a genetic modifier of lifespan, C. elegans remains as relevant as ever in the quest to understand the process of aging. Nematode aging research has proven useful in identifying transcriptional regulators, small molecule signals, cellular mechanisms, epigenetic modifications associated with stress resistance and longevity, and lifespan-extending compounds. Here, we review recent discoveries and selected topics that have emerged in aging research using this incredible little worm.
Collapse
Affiliation(s)
- Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
| | - Louis R Lapierre
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| | | |
Collapse
|
96
|
Papadopoulos C, Meyer H. Detection and Clearance of Damaged Lysosomes by the Endo-Lysosomal Damage Response and Lysophagy. Curr Biol 2018; 27:R1330-R1341. [PMID: 29257971 DOI: 10.1016/j.cub.2017.11.012] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lysosomal membrane permeabilization or lysosomal rupture is recognized as a common and severe stress condition relevant for infection, cellular degeneration and cancer. However, the cellular response mechanisms that protect cells from the consequences of lysosomal damage and ensure lysosomal quality control and homeostasis have only recently been explored. Key elements of this response involve the specific sensing of the damage followed by extensive modification of the organelles with ubiquitin to mark them for clearance by selective macroautophagy, termed lysophagy. Efficient lysophagy is ensured by additional layers of regulation, including modulation by the ubiquitin-directed AAA-ATPase VCP/p97. Lysophagy shares many features with mitophagy, the macroautophagic removal of damaged mitochondria. This review aims to gather available data from different fields and to define the key steps necessary for sensing and subsequent clearance of damaged lysosomes. We conclude with a discussion of disease implications with a focus on neurodegeneration.
Collapse
Affiliation(s)
- Chrisovalantis Papadopoulos
- Molecular Biology I, Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany.
| | - Hemmo Meyer
- Molecular Biology I, Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany.
| |
Collapse
|
97
|
Mauthe M, Orhon I, Rocchi C, Zhou X, Luhr M, Hijlkema KJ, Coppes RP, Engedal N, Mari M, Reggiori F. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy 2018; 14:1435-1455. [PMID: 29940786 PMCID: PMC6103682 DOI: 10.1080/15548627.2018.1474314] [Citation(s) in RCA: 1412] [Impact Index Per Article: 201.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 02/08/2023] Open
Abstract
Macroautophagy/autophagy is a conserved transport pathway where targeted structures are sequestered by phagophores, which mature into autophagosomes, and then delivered into lysosomes for degradation. Autophagy is involved in the pathophysiology of numerous diseases and its modulation is beneficial for the outcome of numerous specific diseases. Several lysosomal inhibitors such as bafilomycin A1 (BafA1), protease inhibitors and chloroquine (CQ), have been used interchangeably to block autophagy in in vitro experiments assuming that they all primarily block lysosomal degradation. Among them, only CQ and its derivate hydroxychloroquine (HCQ) are FDA-approved drugs and are thus currently the principal compounds used in clinical trials aimed to treat tumors through autophagy inhibition. However, the precise mechanism of how CQ blocks autophagy remains to be firmly demonstrated. In this study, we focus on how CQ inhibits autophagy and directly compare its effects to those of BafA1. We show that CQ mainly inhibits autophagy by impairing autophagosome fusion with lysosomes rather than by affecting the acidity and/or degradative activity of this organelle. Furthermore, CQ induces an autophagy-independent severe disorganization of the Golgi and endo-lysosomal systems, which might contribute to the fusion impairment. Strikingly, HCQ-treated mice also show a Golgi disorganization in kidney and intestinal tissues. Altogether, our data reveal that CQ and HCQ are not bona fide surrogates for other types of late stage lysosomal inhibitors for in vivo experiments. Moreover, the multiple cellular alterations caused by CQ and HCQ call for caution when interpreting results obtained by blocking autophagy with this drug.
Collapse
Affiliation(s)
- Mario Mauthe
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Cell Biology, University Medical Center Utrecht, Center for Molecular Medicine, Utrecht, The Netherlands
| | - Idil Orhon
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cecilia Rocchi
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Xingdong Zhou
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Morten Luhr
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership for Molecular Medicine, University of Oslo, Oslo, Norway
| | - Kerst-Jan Hijlkema
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert P. Coppes
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership for Molecular Medicine, University of Oslo, Oslo, Norway
| | - Muriel Mari
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Cell Biology, University Medical Center Utrecht, Center for Molecular Medicine, Utrecht, The Netherlands
| | - Fulvio Reggiori
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Cell Biology, University Medical Center Utrecht, Center for Molecular Medicine, Utrecht, The Netherlands
| |
Collapse
|
98
|
Collins KP, Jackson KM, Gustafson DL. Hydroxychloroquine: A Physiologically-Based Pharmacokinetic Model in the Context of Cancer-Related Autophagy Modulation. J Pharmacol Exp Ther 2018; 365:447-459. [PMID: 29438998 PMCID: PMC5931434 DOI: 10.1124/jpet.117.245639] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/06/2018] [Indexed: 12/14/2022] Open
Abstract
Hydroxychloroquine (HCQ) is a lysosomotropic autophagy inhibitor being used in over 50 clinical trials either alone or in combination with chemotherapy. Pharmacokinetic (PK) and pharmacodynamic (PD) studies with HCQ have shown that drug exposure in the blood does not correlate with autophagy inhibition in either peripheral blood mononuclear cells or tumor tissue. To better explain this PK/PD disconnect, a PBPK was developed for HCQ describing the tissue-specific absorption, distribution, metabolism, and excretion as well as lysosome-specific sequestration. Using physiologic and biochemical parameters derived from literature or obtained experimentally, the model was first developed and validated in mice, and then adapted to simulate human HCQ exposure in whole blood and urine through allometric scaling and species-specific parameter modification. The human model accurately simulated average steady-state concentrations (Css) of those observed in five different HCQ combination clinical trials across seven different doses, which was then expanded by comparison of the Css distribution in a virtual human population at this range of doses. Value of this model lies in its ability to simulate HCQ PK in patients while accounting for PK modification by combination treatment modalities, drug concentrations at the active site in the lysosome under varying pH conditions, and exposure in tissues where toxicity is observed.
Collapse
Affiliation(s)
- Keagan P Collins
- School of Biomedical Engineering (K.P.C., K.M.J., D.L.G.) and Department of Clinical Sciences (D.L.G.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G.)
| | - Kristen M Jackson
- School of Biomedical Engineering (K.P.C., K.M.J., D.L.G.) and Department of Clinical Sciences (D.L.G.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G.)
| | - Daniel L Gustafson
- School of Biomedical Engineering (K.P.C., K.M.J., D.L.G.) and Department of Clinical Sciences (D.L.G.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G.)
| |
Collapse
|
99
|
de Tezanos Pinto F, Adamo HP. The strategic function of the P5-ATPase ATP13A2 in toxic waste disposal. Neurochem Int 2018; 112:108-113. [DOI: 10.1016/j.neuint.2017.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/28/2022]
|
100
|
Zhang W, Meng X, Liu H, Xie L, Liu J, Xu H. Ratio of Polycation and Serum Is a Crucial Index for Determining the RNAi Efficiency of Polyplexes. ACS APPLIED MATERIALS & INTERFACES 2017; 9:43529-43537. [PMID: 29144122 DOI: 10.1021/acsami.7b15797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We report that the mass ratio of the polycation to serum in the medium determines the (RNA interference) RNAi efficiency in vitro by using spermine-modified pullulan (Ps) and spermine-modified dextran (Ds) as polycation models. The high ratio of Ps to serum protein (Ps/Pr) mediated the formation of larger polyplexes, which led to the promoted cellular uptake, enhanced lysosomal escape, and elevated RNAi efficiency. In addition, the supplementary of free Ps also enhanced small interfering RNA transfection because of the elevation of Ps/Pr. Similar results were obtained with Ds. Compared with the adjustment of the nitrogen to phosphate (N/P) ratio in the polyplex, these findings revealed a more applicable strategy to tune the polycation-mediated RNAi efficiency in the serum-containing culture medium.
Collapse
Affiliation(s)
- Weiqi Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100005, P. R. China
| | - Xianghui Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100005, P. R. China
| | - Huike Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100005, P. R. China
| | - Lifei Xie
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100005, P. R. China
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100005, P. R. China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100005, P. R. China
| |
Collapse
|