1
|
Kieffer C, Primas N, Hutter S, Merckx A, Reininger L, Bach S, Ruchaud S, Gaillard F, Laget M, Amrane D, Hervé L, Castera-Ducros C, Renault J, Dumètre A, Rault S, Doerig C, Rathelot P, Vanelle P, Azas N, Verhaeghe P. Target fishing reveals PfPYK-1 and PfRab6 as potential targets of an antiplasmodial 4-anilino-2-trichloromethylquinazoline hit compound. Bioorg Med Chem 2024; 102:117654. [PMID: 38452406 DOI: 10.1016/j.bmc.2024.117654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2023] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024]
Abstract
We present investigations about the mechanism of action of a previously reported 4-anilino-2-trichloromethylquinazoline antiplasmodial hit-compound (Hit A), which did not share a common mechanism of action with established commercial antimalarials and presented a stage-specific effect on the erythrocytic cycle of P. falciparum at 8 < t < 16 h. The target of Hit A was searched by immobilising the molecule on a solid support via a linker and performing affinity chromatography on a plasmodial lysate. Several anchoring positions of the linker (6,7 and 3') and PEG-type linkers were assessed, to obtain a linked-hit molecule displaying in vitro antiplasmodial activity similar to that of unmodified Hit A. This allowed us to identify the PfPYK-1 kinase and the PfRab6 GTP-ase as potential targets of Hit A.
Collapse
Affiliation(s)
- C Kieffer
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France
| | - N Primas
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, Marseille, France; AP-HM, Service Central de la Qualité et de l'Information Pharmaceutiques, Hôpital Conception, Marseille 13005, France
| | - S Hutter
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, IRD, SSA, Mycology & Tropical Eucaryotic Pathogens, Marseille, France
| | - A Merckx
- Université Paris Cité, MERIT, IRD, Paris, France
| | - L Reininger
- Sorbonne Université, CNRS, UMR8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680 Roscoff, France; Sorbonne Université, CNRS, FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, 29680 Roscoff, France
| | - S Bach
- Sorbonne Université, CNRS, UMR8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680 Roscoff, France; Sorbonne Université, CNRS, FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, 29680 Roscoff, France
| | - S Ruchaud
- Sorbonne Université, CNRS, UMR8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680 Roscoff, France; Sorbonne Université, CNRS, FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, 29680 Roscoff, France
| | - F Gaillard
- Sorbonne Université, CNRS, UMR8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680 Roscoff, France; Sorbonne Université, CNRS, FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, 29680 Roscoff, France
| | - M Laget
- Aix Marseille Univ, INSERMN, SSA, MCT, Marseille, France
| | - D Amrane
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, Marseille, France
| | - L Hervé
- Université Paris Cité, MERIT, IRD, Paris, France
| | - C Castera-Ducros
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, Marseille, France; AP-HM, Service Central de la Qualité et de l'Information Pharmaceutiques, Hôpital Conception, Marseille 13005, France
| | - J Renault
- Université de Rennes - Faculté de Pharmacie, ISCR UMR CNRS 6226, Equipe CORINT, Rennes, France
| | - A Dumètre
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, IRD, SSA, Mycology & Tropical Eucaryotic Pathogens, Marseille, France
| | - S Rault
- Normandie Univ, UNICAEN, CERMN, 14000 Caen, France
| | - C Doerig
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - P Rathelot
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, Marseille, France; AP-HM, Service Central de la Qualité et de l'Information Pharmaceutiques, Hôpital Conception, Marseille 13005, France
| | - P Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, Marseille, France; AP-HM, Service Central de la Qualité et de l'Information Pharmaceutiques, Hôpital Conception, Marseille 13005, France
| | - N Azas
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, IRD, SSA, Mycology & Tropical Eucaryotic Pathogens, Marseille, France.
| | - P Verhaeghe
- Univ. Grenoble Alpes, CNRS, DPM UMR 5063, F-38041 Grenoble, France; LCC-CNRS Université de Toulouse, CNRS, UPS, Toulouse, France; Service de Pharmacie, CHU de Nîmes, Place R. Debré, Nîmes, France.
| |
Collapse
|
2
|
Delehouzé C, Comte A, Leon-Icaza SA, Cougoule C, Hauteville M, Goekjian P, Bulinski JC, Dimanche-Boitrel MT, Meunier E, Rousselot M, Bach S. Nigratine as dual inhibitor of necroptosis and ferroptosis regulated cell death. Sci Rep 2022; 12:5118. [PMID: 35332201 PMCID: PMC8944179 DOI: 10.1038/s41598-022-09019-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/23/2021] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
Nigratine (also known as 6E11), a flavanone derivative of a plant natural product, was characterized as highly specific non-ATP competitive inhibitor of RIPK1 kinase, one of the key components of necroptotic cell death signaling. We show here that nigratine inhibited both necroptosis (induced by Tumor Necrosis Factor-α) and ferroptosis (induced by the small molecules glutamate, erastin, RSL3 or cumene hydroperoxide) with EC50 in the µM range. Taken together, our data showed that nigratine is a dual inhibitor of necroptosis and ferroptosis cell death pathways. These findings open potential new therapeutic avenues for treating complex necrosis-related diseases.
Collapse
Affiliation(s)
- Claire Delehouzé
- SeaBeLife Biotech, Place Georges Teissier, 29680, Roscoff, France
- Sorbonne Université, CNRS, UMR 8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680, Roscoff, France
| | - Arnaud Comte
- Université de Lyon, CNRS UMR 5246, ICBMS, Chimiothèque, Université Claude Bernard Lyon 1, 69622, Villeurbanne, France
| | - Stephen Adonai Leon-Icaza
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Céline Cougoule
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Marcelle Hauteville
- Laboratoire de Biochimie Analytique et Synthèse Bioorganique, Université de Lyon, Université Claude Bernard Lyon 1, 69622, Villeurbanne, France
| | - Peter Goekjian
- Université de Lyon, CNRS UMR 5246, ICBMS, Laboratoire Chimie Organique 2-Glycosciences, Université Claude Bernard Lyon 1, 69622, Villeurbanne, France
| | - Jeannette Chloë Bulinski
- Sorbonne Université, CNRS, UMR 8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680, Roscoff, France
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Marie-Thérèse Dimanche-Boitrel
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Etienne Meunier
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Stéphane Bach
- Sorbonne Université, CNRS, UMR 8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, 29680, Roscoff, France.
- Sorbonne Université, CNRS, FR 2424, Plateforme de Criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, 29680, Roscoff Cedex, France.
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| |
Collapse
|
3
|
Bach S, Colas P, Blondel M. [Budding yeast, a model and a tool… also for biomedical research]. Med Sci (Paris) 2020; 36:504-514. [PMID: 32452373 DOI: 10.1051/medsci/2020077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022] Open
Abstract
Yeast has been used for thousands of years as a leavening agent and for alcoholic fermentation, but it is only in 1857 that Louis Pasteur described the microorganism at the basis of these two tremendously important economic activities. From there, yeast strains could be selected and modified on a rational basis to optimize these uses, thereby also allowing the development of yeast as a popular eukaryotic model system. This model led to a cornucopia of seminal discoveries in cell biology. For about two decades yeast has also been used as a model and a tool for therapeutic research, from the production of therapeutics and the development of diagnostic tools to the identification of new therapeutic targets, drug candidates and chemical probes. These diverse chemobiological applications of yeast are presented and discussed in the present review article.
Collapse
Affiliation(s)
- Stéphane Bach
- Sorbonne Université, CNRS, UMR8227, Laboratoire de Biologie Intégrative des Modèles Marins, Station Biologique de Roscoff, place Georges Teissier, 29680 Roscoff, France - Sorbonne Université, CNRS, FR2424, Plateforme de criblage KISSf, Station Biologique de Roscoff, place Georges Teissier, 29680 Roscoff, France
| | - Pierre Colas
- Sorbonne Université, CNRS, UMR8227, Laboratoire de Biologie Intégrative des Modèles Marins, Station Biologique de Roscoff, place Georges Teissier, 29680 Roscoff, France
| | - Marc Blondel
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200 Brest, France - CHRU Brest, service de génétique clinique et de biologie de la reproduction, F-29200 Brest, France
| |
Collapse
|
4
|
Tian Y, Wang S, Shang H, Wang WQ, Wang BQ, Zhang X, Xu XD, Sun GB, Sun XB. The clickable activity-based probe of anti-apoptotic calenduloside E. PHARMACEUTICAL BIOLOGY 2019; 57:133-139. [PMID: 30843752 PMCID: PMC6407588 DOI: 10.1080/13880209.2018.1557699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/31/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 05/27/2023]
Abstract
CONTEXT Calenduloside E (CE), one of the primary natural products found in Aralia elata (Miq.) Seem. (Araliaceae), possesses prominent anti-apoptotic potential. A previous study found that one of the anti-apoptotic CE targets is heat shock protein 90 AB1 (Hsp90AB1) by probe CE-P, while the other targets of CE still need to be identified with more efficient probes. OBJECTIVE This study investigates CE analogue (CEA) as one clickable activity-based probe for use in exploring anti-apoptotic CE targets. MATERIALS AND METHODS Pretreatment of HUVECs with CEA (1.25 μM) for 8 hr, followed by ox-LDL stimulation for 24 h. Flow cytometry analysis and JC-1 staining assays were performed The kinetic constant measurements were tested by the Biacore T200, CM5 Sensor Chip which was activated by using sulpho-NHS/EDC. Ligands were dissolved and injected with a concentration of 12.5, 6.25, 3.125, 1.56, 0.78 and 0 μM. RESULTS CEA was confirmed to possess an anti-apoptotic effect. The probable targets of CE/CEA were calculated, and as one of the higher scores proteins (Fit values: 0.88/0.86), Hsp90 properly got our attention. Molecular modelling study showed that both CE and CEA could bind to Hsp90 with the similar interaction, and the docking scores (S value) were -7.61 and -7.33. SPR assay provided more evidence to prove that CEA can interact with Hsp90 with the KD value 11.7 µM. DISCUSSION AND CONCLUSIONS Our results suggest that clickable probe CEA could alleviate ox-LDL induced apoptosis by a similar mechanism of anti-apoptotic CE, and afforded the possibility of identifying additional anti-apoptotic targets of CE.
Collapse
Affiliation(s)
- Yu Tian
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Bioactive Substances and Resources, Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Zhong guan cun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of new drug discovery based on Classic Chinese Academy of Medical Sciences
| | - Shan Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Bioactive Substances and Resources, Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Zhong guan cun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of new drug discovery based on Classic Chinese Academy of Medical Sciences
| | - Hai Shang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Bioactive Substances and Resources, Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Zhong guan cun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of new drug discovery based on Classic Chinese Academy of Medical Sciences
| | - Wen-Qian Wang
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Bao-Qi Wang
- Center of Research and Development on Life Sciences and Environment Sciences, Harbin University of Commerce, Harbin, China
| | - Xi Zhang
- Center of Research and Development on Life Sciences and Environment Sciences, Harbin University of Commerce, Harbin, China
| | - Xu-Dong Xu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Bioactive Substances and Resources, Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Zhong guan cun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of new drug discovery based on Classic Chinese Academy of Medical Sciences
| | - Gui-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Bioactive Substances and Resources, Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Zhong guan cun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of new drug discovery based on Classic Chinese Academy of Medical Sciences
| | - Xiao-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Bioactive Substances and Resources, Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Zhong guan cun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of new drug discovery based on Classic Chinese Academy of Medical Sciences
| |
Collapse
|
5
|
Wan YJ, Xu L, Song WT, Liu YQ, Wang LC, Zhao MB, Jiang Y, Liu LY, Zeng KW, Tu PF. The Ethanolic Extract of Caesalpinia sappan Heartwood Inhibits Cerebral Ischemia/Reperfusion Injury in a Rat Model Through a Multi-Targeted Pharmacological Mechanism. Front Pharmacol 2019; 10:29. [PMID: 30804781 PMCID: PMC6370896 DOI: 10.3389/fphar.2019.00029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2018] [Accepted: 01/11/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Caesalpinia sappan L. (C. sappan) is a traditional Chinese medicinal plant. The dried heartwood of C. sappan (also known as Sappan wood) has been widely used for the folkloric medical treatment of ischemic cerebral stroke in China. However, the detailed underlying pharmacological mechanism still remains largely unexplored. Methods: In this study, a middle cerebral artery occlusion (MCAO) rat model was employed to elucidate the mechanism of the anti-cerebral ischemic effects of C. sappan ethanolic extract (CEE). Moreover, systemic multi-target identification coupled with gene ontology biological process (GO BP) and reactome pathway analysis was used to investigate the potential neuroprotective mechanism. Furthermore, the presumed mechanism was confirmed through biological analysis by determining the effects of CEE on the identified signaling pathways in PC12 cells model-induced by oxygen-glucose deprivation/reperfusion (OGD/R). Results: Our study demonstrates that CEE (both through in vivo administration at a dosage of 300 mg/kg and through in vitro incubation at a dosage of 2.4 μg/mL) is a neuroprotective agent that can effectively inhibit neuronal damage, promote synaptic generation, and suppress the activation of neutrophils, microglia, and astrocytes. Moreover, the neuroprotective mechanism of CEE is mediated via regulating 150 potential target proteins, which are associated with 6 biological processes and 10 pathways, including JAK-STAT, HSP90 and DNA damage/telomere stress. Conclusion: CEE can exert neuroprotective effect through multi-target pharmacological mechanisms to prevent ischemia/reperfusion-induced cerebral injury.
Collapse
Affiliation(s)
- Yan-Jun Wan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Li Xu
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wen-Ting Song
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu-Qi Liu
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Li-Chao Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ming-Bo Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Lian-Ying Liu
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
6
|
Woodland JG, Hunter R, Smith PJ, Egan TJ. Chemical Proteomics and Super-resolution Imaging Reveal That Chloroquine Interacts with Plasmodium falciparum Multidrug Resistance-Associated Protein and Lipids. ACS Chem Biol 2018; 13:2939-2948. [PMID: 30208272 DOI: 10.1021/acschembio.8b00583] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
It is well established that chloroquine, a quinoline antimalarial, inhibits hemozoin formation in the malaria parasite. Counterintuitively, this archetypal antimalarial is also used in the treatment of diseases in which hemozoin biocrystallization does not play a role. Hence, we decided to investigate whether chloroquine possesses binding targets other than Fe(III) protoporphyrin IX in blood stage Plasmodium falciparum parasites and whether these are related to sites of accumulation within the parasite other than the digestive vacuole. A 7-nitrobenz-2-oxa-1,3-diazole (NBD)-labeled fluorescent derivative of chloroquine, especially sensitive to regions outside the digestive vacuole and retaining the antiplasmodial pharmacophore, was synthesized to investigate subcellular localization in the parasite. Super-resolution microscopy revealed association with membranes including the parasite plasma membrane, the endoplasmic reticulum, and possibly also the mitochondrion. A drug-labeled affinity matrix was then prepared to capture protein binding targets of chloroquine. SDS-PAGE revealed a single prominent band between 200 and 250 kDa from the membrane-associated fraction. Subsequent proteomic analysis revealed that this band corresponded to P. falciparum multidrug resistance-associated protein (PfMRP1). Intrigued by this finding, we demonstrated pull-down of PfMRP1 by matrices labeled with Cinchona alkaloids quinine and quinidine. While PfMRP1 has been implicated in resistance to quinolines and other antimalarials, this is the first time that these drugs have been found to bind directly to this protein. Based on previous reports, PfMRP1, the only prominent protein found to bind to quinolines in this work, is likely to modulate the activity of these antimalarials in P. falciparum rather than act as a drug target.
Collapse
Affiliation(s)
- John G. Woodland
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch, Cape Town 7701, South Africa
| | - Roger Hunter
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch, Cape Town 7701, South Africa
| | | | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch, Cape Town 7701, South Africa
| |
Collapse
|
7
|
Wang J, Wu J, Li X, Liu H, Qin J, Bai Z, Chi B, Chen X. Identification and validation nucleolin as a target of curcumol in nasopharyngeal carcinoma cells. J Proteomics 2018; 182:1-11. [PMID: 29684682 DOI: 10.1016/j.jprot.2018.04.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/17/2017] [Revised: 03/22/2018] [Accepted: 04/19/2018] [Indexed: 11/16/2022]
Abstract
Identification of the specific protein target(s) of a drug is a critical step in unraveling its mechanisms of action (MOA) in many natural products. Curcumol, isolated from well known Chinese medicinal plant Curcuma zedoary, has been shown to possess multiple biological activities. It can inhibit nasopharyngeal carcinoma (NPC) proliferation and induce apoptosis, but its target protein(s) in NPC cells remains unclear. In this study, we employed a mass spectrometry-based chemical proteomics approach reveal the possible protein targets of curcumol in NPC cells. Cellular thermal shift assay (CETSA), molecular docking and cell-based assay was used to validate the binding interactions. Chemical proteomics capturing uncovered that NCL is a target of curcumol in NPC cells, Molecular docking showed that curcumol bound to NCL with an -7.8 kcal/mol binding free energy. Cell function analysis found that curcumol's treatment leads to a degradation of NCL in NPC cells, and it showed slight effects on NP69 cells. In conclusion, our results providing evidences that NCL is a target protein of curcumol. We revealed that the anti-cancer effects of curcumol in NPC cells are mediated, at least in part, by NCL inhibition. SIGNIFICANCE Many natural products showed high bioactivity, while their mechanisms of action (MOA) are very poor or completely missed. Understanding the MOA of natural drugs can thoroughly exploit their therapeutic potential and minimize their adverse side effects. Identification of the specific protein target(s) of a drug is a critical step in unraveling its MOA. Compound-centric chemical proteomics is a classic chemical proteomics approach which integrates chemical synthesis with cell biology and mass spectrometry (MS) to identify protein targets of natural products determine the drug mechanism of action, describe its toxicity, and figure out the possible cause of off-target. It is an affinity-based chemical proteomics method to identify small molecule-protein interactions through affinity chromatography approach coupled with mass spectrometry, has been conventionally used to identify target proteins and has yielded good results. Curcumol, has shown effective inhibition on Nasopharyngeal Carcinoma (NPC) Cells, interacted with NCL and then initiated the anti-tumor biological effect. This research demonstrated the effectiveness of chemical proteomics approaches in natural drugs molecular target identification, revealing and understanding of the novel mechanism of actions of curcumol is crucial for cancer prevention and treatment in nasopharynx cancer.
Collapse
Affiliation(s)
- Juan Wang
- Xiangya Hospital, Central South University, Changsha 410008, China; College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Jiacai Wu
- Research Center for Science, Guilin Medical University, Guilin 541004, China
| | - Xumei Li
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Haowei Liu
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Jianli Qin
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Zhun Bai
- Intensive Care Unit, The Affiliated Zhuzhou Hospital XiangYa Medical College CSU, Zhuzhou 412007, China
| | - Bixia Chi
- Department of Gastroenterology, The First People's Hospital of Yueyang, Yueyang 414000, China
| | - Xu Chen
- College of Pharmacy, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
8
|
Cui Q, Hou Y, Wang Y, Li X, Liu Y, Ma X, Wang Z, Wang W, Tao J, Wang Q, Jiang M, Chen D, Feng X, Bai G. Biodistribution of arctigenin-loaded nanoparticles designed for multimodal imaging. J Nanobiotechnology 2017; 15:27. [PMID: 28388905 PMCID: PMC5383946 DOI: 10.1186/s12951-017-0263-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/04/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Tracking targets of natural products is one of the most challenging issues in fields ranging from pharmacognosy to biomedicine. It is widely recognized that the biocompatible nanoparticle (NP) could function as a "key" that opens the target "lock". RESULTS We report a functionalized poly-lysine NP technique that can monitor the target protein of arctigenin (ATG) in vivo non-invasively. The NPs were synthesized, and their morphologies and surface chemical properties were characterized by transmission electron microscopy (TEM), laser particle size analysis and atomic force microscopy (AFM). In addition, we studied the localization of ATG at the level of the cell and the whole animal (zebrafish and mice). We demonstrated that fluorescent NPs could be ideal carriers in the development of a feasible method for target identification. The distributions of the target proteins were found to be consistent with the pharmacological action of ATG at the cellular and whole-organism levels. CONCLUSIONS The results indicated that functionalized poly-lysine NPs could be valuable in the multimodal imaging of arctigenin.
Collapse
Affiliation(s)
- Qingxin Cui
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yuanyuan Hou
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yanan Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071 China
| | - Xu Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Physiology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Yang Liu
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Xiaoyao Ma
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Zengyong Wang
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Weiya Wang
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Jin Tao
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Qian Wang
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Min Jiang
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Dongyan Chen
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Physiology, School of Medicine, Nankai University, Tianjin, 300071 China
| | - Xizeng Feng
- State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071 China
| | - Gang Bai
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071 People’s Republic of China
| |
Collapse
|
9
|
Tian Y, Wang S, Shang H, Wang M, Sun G, Xu X, Sun X. The proteomic profiling of calenduloside E targets in HUVEC: design, synthesis and application of biotinylated probe BCEA. RSC Adv 2017. [DOI: 10.1039/c6ra25572h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022] Open
Abstract
The proteomic profiling of calenduloside E targets was researched by employing the biotinylated probe BCEA of natural product calenduloside E.
Collapse
Affiliation(s)
- Yu Tian
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Shan Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Hai Shang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Xudong Xu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| |
Collapse
|
10
|
Abstract
Cumulative scientific and technological advances over the past two centuries have transformed drug discovery from a largely serendipitous process into the high tech pipelines of today.
Collapse
Affiliation(s)
- Hassan Al-Ali
- Miami Project to Cure Paralysis
- University of Miami Miller School of Medicine
- Miami FL 33136
- USA
| |
Collapse
|
11
|
Fang R, Cui Q, Sun J, Duan X, Ma X, Wang W, Cheng B, Liu Y, Hou Y, Bai G. PDK1/Akt/PDE4D axis identified as a target for asthma remedy synergistic with β2 AR agonists by a natural agent arctigenin. Allergy 2015; 70:1622-32. [PMID: 26335809 DOI: 10.1111/all.12763] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 08/31/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Asthma is a heterogenetic disorder characterized by chronic inflammation with variable airflow obstruction and airway hyper-responsiveness. As the most potent and popular bronchodilators, β2 adrenergic receptor (β2 AR) agonists bind to the β2 ARs that are coupled via a stimulatory G protein to adenylyl cyclase, thereby improving cAMP accumulation and resulting in airway smooth muscle relaxation. We previously demonstrated arctigenin had a synergistic function with the β2 AR agonist, but the target for this remained elusive. METHOD Chemical proteomics capturing was used to enrich and uncover the target of arctigenin in human bronchial smooth muscle cells, and reverse docking and molecular dynamic stimulation were performed to evaluate the binding of arctigenin and its target. In vitro enzyme activities and protein levels were demonstrated with special kits and Western blotting. Finally, guinea pig tracheal muscle segregation and ex vivo function were analysed. RESULTS Arctigenin bound to PDK1 with an ideal binding free energy -25.45 kcal/mol and inhibited PDK1 kinase activity without changing its protein level. Additionally, arctigenin reduced PKB/Akt-induced phosphorylation of PDE4D, which was first identified in this study. Attenuation of PDE4D resulted in cAMP accumulation in human bronchial smooth muscle. The inhibition of PDK1 showed a synergistic function with β2 AR agonists and relaxed the constriction of segregated guinea pig tracheal muscle. CONCLUSIONS The PDK1/Akt/PDE4D axis serves as a novel asthma target, which may benefit airflow obstruction.
Collapse
Affiliation(s)
- R. Fang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
- State Key Laboratory of Natural and Biomimetic Drugs; Peking University; Beijing China
- State Key Laboratory of Medicinal Chemical Biology; Department of Biochemistry; College of Life Sciences; Nankai University; Tianjin China
| | - Q. Cui
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - J. Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - X. Duan
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - X. Ma
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - W. Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - B. Cheng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - Y. Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - Y. Hou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
- State Key Laboratory of Natural and Biomimetic Drugs; Peking University; Beijing China
| | - G. Bai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy; Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| |
Collapse
|
12
|
Baratte B, Serive B, Bach S. [Screening marine resources to find novel chemical inhibitors of disease-relevant protein kinases]. Med Sci (Paris) 2015; 31:538-45. [PMID: 26059305 DOI: 10.1051/medsci/20153105016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022] Open
Abstract
Since the early 1970's, investigators at Station Biologique de Roscoff (SBR), France, have been using marine organisms as models to describe molecular pathways conserved through evolution in mammalian cells (e.g. the cyclin-dependent kinases involved in the control of the cell division cycle). Some kinases are misregulated in various human pathologies, including cancers. Using a specialized screening approach, chemical libraries were analysed, using on-site facilities at Roscoff, in order to identify small chemical inhibitors of protein kinases. Eight chemical scaffolds produced by marine organisms were characterized as candidate drugs by our screening facility, some of which are being considered as chemical tools to pinpoint specific cellular functions of the targeted kinases. In this review, we describe our existing screening facilities and we discuss new perspectives related to marine bioprospecting.
Collapse
Affiliation(s)
- Blandine Baratte
- CNRS/UPMC USR3151, plate-forme de criblage KISSf (kinase inhibitor specialized screening facility), station biologique de Roscoff, place Georges Teissier, CS 90074, 29688 Roscoff Cedex, France
| | - Benoît Serive
- CNRS/UPMC USR3151, plate-forme de criblage KISSf (kinase inhibitor specialized screening facility), station biologique de Roscoff, place Georges Teissier, CS 90074, 29688 Roscoff Cedex, France
| | - Stéphane Bach
- CNRS/UPMC USR3151, plate-forme de criblage KISSf (kinase inhibitor specialized screening facility), station biologique de Roscoff, place Georges Teissier, CS 90074, 29688 Roscoff Cedex, France
| |
Collapse
|
13
|
Corbel C, Zhang B, Le Parc A, Baratte B, Colas P, Couturier C, Kosik KS, Landrieu I, Le Tilly V, Bach S. Tamoxifen inhibits CDK5 kinase activity by interacting with p35/p25 and modulates the pattern of tau phosphorylation. ACTA ACUST UNITED AC 2015; 22:472-482. [PMID: 25865311 DOI: 10.1016/j.chembiol.2015.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2014] [Revised: 02/27/2015] [Accepted: 03/06/2015] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent kinase 5 (CDK5) is a multifunctional enzyme that plays numerous roles, notably in brain development. CDK5 is activated through its association with the activators, p35 and p39, rather than by cyclins. Proteolytic procession of the N-terminal part of its activators has been linked to Alzheimer's disease and various other neuropathies. The interaction with the proteolytic product p25 prolongs CDK5 activation and modifies the substrate specificity. In order to discover small-molecule inhibitors of the interaction between CDK5 and p25, we have used a bioluminescence resonance energy transfer (BRET)-based screening assay. Among the 1,760 compounds screened, the generic drug tamoxifen has been identified. The inhibition of the CDK5 activity by tamoxifen was notably validated by monitoring the phosphorylation state of tau protein. The study of the molecular mechanism of inhibition indicates that tamoxifen interacts with p25 to block the CDK5/p25 interaction and pave the way for new treatments of tauopathies.
Collapse
Affiliation(s)
- Caroline Corbel
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France; EA4250-LIMATB-EG2B, Centre de Recherche et d'Enseignement Yves Coppens, Université de Bretagne Sud, 56017 Vannes, France; Kosik Laboratory, Neuroscience Research Institute, Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Bing Zhang
- School of Renewable Energy, North China Electric Power Electricity, 071003 Beijing, China
| | - Annabelle Le Parc
- EA4250-LIMATB-EG2B, Centre de Recherche et d'Enseignement Yves Coppens, Université de Bretagne Sud, 56017 Vannes, France
| | - Blandine Baratte
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France
| | - Pierre Colas
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France
| | - Cyril Couturier
- UMR761-INSERM Lille University, Biostructures and Drug Discovery, 59006 Lille, France
| | - Kenneth S Kosik
- Kosik Laboratory, Neuroscience Research Institute, Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Isabelle Landrieu
- UMR8576 CNRS-Lille North of France University, 59658 Villeneuve d'Ascq, France; Interdisciplinary Research Institute (IRI), 58658 Villeneuve d'Ascq, France
| | - Véronique Le Tilly
- EA4250-LIMATB-EG2B, Centre de Recherche et d'Enseignement Yves Coppens, Université de Bretagne Sud, 56017 Vannes, France
| | - Stéphane Bach
- USR3151-CNRS/UPMC, Protein Phosphorylation and Disease Laboratory, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, Bretagne, France.
| |
Collapse
|
14
|
Li S, Li Y, Yu H, Wu Z, Jiang J, Yu R, Wang Y. Fluorescence amplification detection via terminal protection of small molecule–protein interactions. RSC Adv 2015. [DOI: 10.1039/c5ra20240j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022] Open
Abstract
A novel fluorescence assay was developed for detecting small molecule–protein interactions on the basis of terminal protection.
Collapse
Affiliation(s)
- Sen Li
- College of Science
- The Naval University of Engineering
- Wuhan
- P. R. China
| | - Yu Li
- College of Science
- The Naval University of Engineering
- Wuhan
- P. R. China
| | - Hongwei Yu
- College of Science
- The Naval University of Engineering
- Wuhan
- P. R. China
| | - Zhan Wu
- State Key Laboratory for Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- P. R. China
| | - Jianhui Jiang
- State Key Laboratory for Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- P. R. China
| | - Ruqin Yu
- State Key Laboratory for Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- P. R. China
| | - Yuansheng Wang
- College of Science
- The Naval University of Engineering
- Wuhan
- P. R. China
| |
Collapse
|
15
|
Voisset C, Blondel M. [Chemobiology at happy hour: yeast as a model for pharmacological screening]. Med Sci (Paris) 2014; 30:1161-8. [PMID: 25537047 DOI: 10.1051/medsci/20143012020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022] Open
Abstract
Since its discovery and description by Louis Pasteur, the budding yeast Saccharomyces cerevisiae, which was used for thousands of years for alcoholic fermentation and as a leavening agent, has become a popular model system in biology. One of the reasons for this popularity is the strong conservation from yeast to human of most of the pathways controlling cell growth and fate. In addition, at least 30 % of human genes involved in diseases have a functional homolog in yeast. Hence, yeast is now widely used for modelling and deciphering physiopathological mechanisms as well as for developing pharmacological approaches like phenotype-based drug screening. Three examples of such yeast-based chemobiological studies are presented.
Collapse
Affiliation(s)
- Cécile Voisset
- Inserm UMR 1078 ; Université de Bretagne occidentale, Faculté de médecine et des sciences de la santé ; Établissement français du sang (EFS) ; CHRU Brest, hôpital Morvan, laboratoire de génétique moléculaire, 22, avenue Camille Desmoulins 29200 Brest, France
| | - Marc Blondel
- Inserm UMR 1078 ; Université de Bretagne occidentale, Faculté de médecine et des sciences de la santé ; Établissement français du sang (EFS) ; CHRU Brest, hôpital Morvan, laboratoire de génétique moléculaire, 22, avenue Camille Desmoulins 29200 Brest, France
| |
Collapse
|
16
|
Terminal protection of small molecule-linked DNA for small molecule-protein interaction assays. Int J Mol Sci 2014; 15:5221-32. [PMID: 24670475 PMCID: PMC4013559 DOI: 10.3390/ijms15045221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/25/2014] [Revised: 03/15/2014] [Accepted: 03/17/2014] [Indexed: 01/24/2023] Open
Abstract
Methods for the detection of specific interactions between diverse proteins and various small-molecule ligands are of significant importance in understanding the mechanisms of many critical physiological processes of organisms. The techniques also represent a major avenue to drug screening, molecular diagnostics, and public safety monitoring. Terminal protection assay of small molecule-linked DNA is a demonstrated novel methodology which has exhibited great potential for the development of simple, sensitive, specific and high-throughput methods for the detection of small molecule–protein interactions. Herein, we review the basic principle of terminal protection assay, the development of associated methods, and the signal amplification strategies adopted for performance improving in small molecule–protein interaction assay.
Collapse
|
17
|
Voisset C, Daskalogianni C, Contesse MA, Mazars A, Arbach H, Le Cann M, Soubigou F, Apcher S, Fåhraeus R, Blondel M. A yeast-based assay identifies drugs that interfere with immune evasion of the Epstein-Barr virus. Dis Model Mech 2014; 7:435-44. [PMID: 24558096 PMCID: PMC3974454 DOI: 10.1242/dmm.014308] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023] Open
Abstract
Epstein-Barr virus (EBV) is tightly associated with certain human cancers, but there is as yet no specific treatment against EBV-related diseases. The EBV-encoded EBNA1 protein is essential to maintain viral episomes and for viral persistence. As such, EBNA1 is expressed in all EBV-infected cells, and is highly antigenic. All infected individuals, including individuals with cancer, have CD8(+) T cells directed towards EBNA1 epitopes, yet the immune system fails to detect and destroy cells harboring the virus. EBV immune evasion depends on the capacity of the Gly-Ala repeat (GAr) domain of EBNA1 to inhibit the translation of its own mRNA in cis, thereby limiting the production of EBNA1-derived antigenic peptides presented by the major histocompatibility complex (MHC) class I pathway. Here we establish a yeast-based assay for monitoring GAr-dependent inhibition of translation. Using this assay we identify doxorubicin (DXR) as a compound that specifically interferes with the GAr effect on translation in yeast. DXR targets the topoisomerase-II-DNA complexes and thereby causes genomic damage. We show, however, that the genotoxic effect of DXR and various analogs thereof is uncoupled from the effect on GAr-mediated translation control. This is further supported by the observation that etoposide and teniposide, representing another class of topoisomerase-II-DNA targeting drugs, have no effect on GAr-mediated translation control. DXR and active analogs stimulate, in a GAr-dependent manner, EBNA1 expression in mammalian cells and overcome GAr-dependent restriction of MHC class I antigen presentation. These results validate our approach as an effective high-throughput screening assay to identify drugs that interfere with EBV immune evasion and, thus, constitute candidates for treating EBV-related diseases, in particular EBV-associated cancers.
Collapse
Affiliation(s)
- Cécile Voisset
- Institut National de la Santé et de la Recherche Médicale UMR 1078; Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé; Etablissement Français du Sang (EFS) Bretagne; CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest F-29200, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Margarucci L, Monti MC, Esposito R, Tosco A, Hamel E, Riccio R, Casapullo A. N-Formyl-7-amino-11-cycloamphilectene, a marine sponge metabolite, binds to tubulin and modulates microtubule depolymerization. MOLECULAR BIOSYSTEMS 2014; 10:862-7. [PMID: 24493364 DOI: 10.1039/c3mb70315k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/28/2023]
Abstract
The importance of protein-small molecule interaction in drug discovery, medicinal chemistry and biology has driven the development of new analytical methods to disclose the whole interactome of bioactive compounds. To accelerate targets discovery of N-formyl-7-amino-11-cycloamphilectene (CALe), a marine bioactive diterpene isolated from the Vanuatu sponge Axinella sp., a chemoproteomic-based approach has been successfully developed. CALe is a potent anti-inflammatory agent, modulating NO and prostaglandin E2 overproduction by dual inhibition of the enhanced inducible NO synthase expression and cyclo-oxygenase-2 activity, without any evidence of cytotoxic effects. In this paper, several isoforms of tubulin have been identified as CALe off-targets by chemical proteomics combined with bio-physical orthogonal approaches. In the following biological analysis of its cellular effect, CALe was found to protect microtubules against the colcemid depolymerizing effect.
Collapse
Affiliation(s)
- Luigi Margarucci
- Dipartimento di Farmacia, Università degli Studi di Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy.
| | | | | | | | | | | | | |
Collapse
|
19
|
Hosseinzadeh H, Mehri S, Heshmati A, Ramezani M, Sahebkar A, Abnous K. Proteomic screening of molecular targets of crocin. ACTA ACUST UNITED AC 2014; 22:5. [PMID: 24393168 PMCID: PMC3922837 DOI: 10.1186/2008-2231-22-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/04/2013] [Accepted: 09/22/2013] [Indexed: 12/31/2022]
Abstract
Background Traditional drug discovery approaches are mainly relied on the observed phenotypic changes following administration of a plant extract, drug candidate or natural product. Recently, target-based approaches are becoming more popular. The present study aimed to identify the cellular targets of crocin, the bioactive dietary carotenoid present in saffron, using an affinity-based method. Methods Heart, kidney and brain tissues of BALB/c mice were homogenized and extracted for the experiments. Target deconvolution was carried out by first passing cell lysate through an affinity column prepared by covalently attaching crocin to agarose beads. Isolated proteins were separated on a 2D gel, trypsinized in situ and identified by MALDI-TOF/TOF mass spectrometry. MASCOT search engine was used to analyze Mass Data. Results Part of proteome that physically interacts with crocin was found to consist of beta-actin-like protein 2, cytochrome b-c1 complex subunit 1, ATP synthase subunit beta, tubulin beta-3 chain, tubulin beta-6 chain, 14-3-3 protein beta/alpha, V-type proton ATPase catalytic subunitA, 60 kDa heat shock protein, creatine kinase b-type, peroxiredoxin-2, cytochrome b-c1 complex subunit 2, acetyl-coA acetyltransferase, cytochrome c1, proteasome subunit alpha type-6 and proteasome subunit alpha type-4. Conclusion The present findings revealed that crocin physically binds to a wide range of cellular proteins such as structural proteins, membrane transporters, and enzymes involved in ATP and redox homeostasis and signal transduction.
Collapse
Affiliation(s)
| | | | | | | | | | - Khalil Abnous
- Pharmaceutical Research Center, Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, 91775-1365 Mashhad, Iran.
| |
Collapse
|
20
|
Prudent R, Soleilhac E, Barette C, Fauvarque MO, Lafanechère L. Les criblages phénotypiques ou comment faire d’une pierre deux coups. Med Sci (Paris) 2013; 29:897-905. [DOI: 10.1051/medsci/20132910018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Renaud Prudent
- Institut Albert Bonniot, CRI Inserm/Université Joseph Fourier (UJF) U823, équipe 3 Polarité, développement et cancer, rond-point de la Chantourne, 38706 La Tronche Cedex, France
| | | | | | | | | |
Collapse
|
21
|
Margarucci L, Monti MC, Cassiano C, Mozzicafreddo M, Angeletti M, Riccio R, Tosco A, Casapullo A. Chemical proteomics-driven discovery of oleocanthal as an Hsp90 inhibitor. Chem Commun (Camb) 2013; 49:5844-6. [PMID: 23703283 DOI: 10.1039/c3cc41858h] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/27/2022]
Abstract
Hsp90, a key target in cancer therapy, has been identified as the main partner of oleocanthal, an olive oil bioactive compound. A combination of chemical and biological assays disclosed its mechanism of action at the molecular level.
Collapse
Affiliation(s)
- Luigi Margarucci
- Dipartimento di Farmacia, Università degli Studi di Salerno, Via Ponte don Melillo, 84084 Fisciano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Arai T, Uehata M, Akatsuka H, Kamiyama T. A quantitative analysis to unveil specific binding proteins for bioactive compounds. Protein Eng Des Sel 2013; 26:249-54. [DOI: 10.1093/protein/gzs103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
|
23
|
Pieretti S, Haanstra JR, Mazet M, Perozzo R, Bergamini C, Prati F, Fato R, Lenaz G, Capranico G, Brun R, Bakker BM, Michels PAM, Scapozza L, Bolognesi ML, Cavalli A. Naphthoquinone derivatives exert their antitrypanosomal activity via a multi-target mechanism. PLoS Negl Trop Dis 2013; 7:e2012. [PMID: 23350008 PMCID: PMC3547856 DOI: 10.1371/journal.pntd.0002012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/19/2012] [Accepted: 12/03/2012] [Indexed: 11/21/2022] Open
Abstract
Background and Methodology Recently, we reported on a new class of naphthoquinone derivatives showing a promising anti-trypanosomatid profile in cell-based experiments. The lead of this series (B6, 2-phenoxy-1,4-naphthoquinone) showed an ED50 of 80 nM against Trypanosoma brucei rhodesiense, and a selectivity index of 74 with respect to mammalian cells. A multitarget profile for this compound is easily conceivable, because quinones, as natural products, serve plants as potent defense chemicals with an intrinsic multifunctional mechanism of action. To disclose such a multitarget profile of B6, we exploited a chemical proteomics approach. Principal Findings A functionalized congener of B6 was immobilized on a solid matrix and used to isolate target proteins from Trypanosoma brucei lysates. Mass analysis delivered two enzymes, i.e. glycosomal glycerol kinase and glycosomal glyceraldehyde-3-phosphate dehydrogenase, as potential molecular targets for B6. Both enzymes were recombinantly expressed and purified, and used for chemical validation. Indeed, B6 was able to inhibit both enzymes with IC50 values in the micromolar range. The multifunctional profile was further characterized in experiments using permeabilized Trypanosoma brucei cells and mitochondrial cell fractions. It turned out that B6 was also able to generate oxygen radicals, a mechanism that may additionally contribute to its observed potent trypanocidal activity. Conclusions and Significance Overall, B6 showed a multitarget mechanism of action, which provides a molecular explanation of its promising anti-trypanosomatid activity. Furthermore, the forward chemical genetics approach here applied may be viable in the molecular characterization of novel multitarget ligands. The multitarget approach can represent a promising strategy for the discovery of innovative drug candidates against neglected tropical diseases. However, multitarget drug discovery can be very demanding, because of the highly time-consuming step related to the fine balancing of the biological activities against selected targets. An innovative workflow for discovering multitarget drugs can be envisioned: i) design and synthesis of natural-like compounds; ii) test them using phenotypic cell-based assays; iii) fishing potential targets by means of chemical proteomics. This workflow might rapidly provide new hit candidates that can be further progressed to the hit-to-lead and lead optimization steps of the drug discovery process. The two latter steps can benefit from information on the molecular target(s), which may be identified by chemical proteomics. Herein, we report on the elucidation of the mode of action of a new series of anti-trypanosomal naphthoquinone compounds, previously tested using cell-based assays, by means of chemical proteomics, classical biochemistry, molecular and system biology.
Collapse
Affiliation(s)
- Simone Pieretti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
- Department of Biochemistry, University of Bologna, Bologna, Italy
| | - Jurgen R. Haanstra
- Department of Pediatrics, Centre for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Molecular Cell Physiology, Faculty of Earth and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Muriel Mazet
- Research Unit for Tropical Diseases, de Duve Institute and Laboratory of Biochemistry, Université Catholique de Louvain, Brussels, Belgium
| | - Remo Perozzo
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | | | - Federica Prati
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
| | - Romana Fato
- Department of Biochemistry, University of Bologna, Bologna, Italy
| | - Giorgio Lenaz
- Department of Biochemistry, University of Bologna, Bologna, Italy
| | | | - Reto Brun
- Swiss Tropical Institute, Basel, Switzerland
| | - Barbara M. Bakker
- Department of Pediatrics, Centre for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Molecular Cell Physiology, Faculty of Earth and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul A. M. Michels
- Research Unit for Tropical Diseases, de Duve Institute and Laboratory of Biochemistry, Université Catholique de Louvain, Brussels, Belgium
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- * E-mail: (MLB); (AC)
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
- * E-mail: (MLB); (AC)
| |
Collapse
|
24
|
Wang K, Yang T, Wu Q, Zhao X, Nice EC, Huang C. Chemistry-based functional proteomics for drug target deconvolution. Expert Rev Proteomics 2013; 9:293-310. [PMID: 22809208 DOI: 10.1586/epr.12.19] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/05/2023]
Abstract
Drug target deconvolution, a process that identifies targets to small molecules in complex biological samples, which underlie the biological responses that are observed when a drug is administered, plays an important role in current drug discovery. Despite the fact that genomics and proteomics have provided a flood of information that contributes to the progress of drug target identification and validation, the current approach to drug target deconvolution still poses dilemmas. Chemistry-based functional proteomics, a multidisciplinary strategy, has become the preferred method of choice to deconvolute drug target pools, based on direct interactions between small molecules and their protein targets. This approach has already identified a broad panel of previously undefined enzymes with potential as drug targets and defined targets that can rationalize side effects and toxicity for new drug candidates and existing therapeutics. Herein, the authors discuss both activity-based protein profiling and compound-centric chemical proteomics approaches used in chemistry-based functional proteomics and their applications for the identification and characterization of small molecular targets.
Collapse
Affiliation(s)
- Kui Wang
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, PR China
| | | | | | | | | | | |
Collapse
|
25
|
Cassiano C, Monti MC, Festa C, Zampella A, Riccio R, Casapullo A. Chemical Proteomics Reveals Heat Shock Protein 60 To Be the Main Cellular Target of the Marine Bioactive Sesterterpene Suvanine. Chembiochem 2012; 13:1953-8. [DOI: 10.1002/cbic.201200291] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/04/2012] [Indexed: 01/23/2023]
|
26
|
Margarucci L, Monti MC, Mencarelli A, Cassiano C, Fiorucci S, Riccio R, Zampella A, Casapullo A. Heat shock proteins as key biological targets of the marine natural cyclopeptide perthamide C. MOLECULAR BIOSYSTEMS 2012; 8:1412-7. [DOI: 10.1039/c2mb05507d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022]
|
27
|
Sakamoto S, Hatakeyama M, Ito T, Handa H. Tools and methodologies capable of isolating and identifying a target molecule for a bioactive compound. Bioorg Med Chem 2011; 20:1990-2001. [PMID: 22264760 DOI: 10.1016/j.bmc.2011.12.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/10/2011] [Revised: 12/09/2011] [Accepted: 12/13/2011] [Indexed: 01/29/2023]
Abstract
Elucidating the mechanism of action of bioactive compounds, such as commonly used pharmaceutical drugs and biologically active natural products, in the cells and the living body is important in drug discovery research. To this end, isolation and identification of target protein(s) for the bioactive compound are essential in understanding its function fully. And, development of reliable tools and methodologies capable of addressing efficiently identification and characterization of the target proteins based on the bioactive compounds accelerates drug discovery research. Affinity-based isolation and identification of target molecules for the bioactive compounds is a classic, but still powerful approach. This paper introduces recent progress on affinity chromatography system, focusing on development of practical affinity matrices and useful affinity-based methodologies on target identification. Beneficial affinity chromatography systems with using practical tools and useful methodologies facilitate chemical biology and drug discovery research.
Collapse
Affiliation(s)
- Satoshi Sakamoto
- Department of Biological Information, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | |
Collapse
|
28
|
Wang D, Zetterström CE, Gabrielsen M, Beckham KSH, Tree JJ, Macdonald SE, Byron O, Mitchell TJ, Gally DL, Herzyk P, Mahajan A, Uvell H, Burchmore R, Smith BO, Elofsson M, Roe AJ. Identification of bacterial target proteins for the salicylidene acylhydrazide class of virulence-blocking compounds. J Biol Chem 2011; 286:29922-31. [PMID: 21724850 PMCID: PMC3191033 DOI: 10.1074/jbc.m111.233858] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022] Open
Abstract
A class of anti-virulence compounds, the salicylidene acylhydrazides, has been widely reported to block the function of the type three secretion system of several Gram-negative pathogens by a previously unknown mechanism. In this work we provide the first identification of bacterial proteins that are targeted by this group of compounds. We provide evidence that their mode of action is likely to result from a synergistic effect arising from a perturbation of the function of several conserved proteins. We also examine the contribution of selected target proteins to the pathogenicity of Yersinia pseudotuberculosis and to expression of virulence genes in Escherichia coli O157.
Collapse
Affiliation(s)
- Dai Wang
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Margarucci L, Monti MC, Fontanella B, Riccio R, Casapullo A. Chemical proteomics reveals bolinaquinone as a clathrin-mediated endocytosis inhibitor. ACTA ACUST UNITED AC 2011; 7:480-5. [DOI: 10.1039/c0mb00126k] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
|
30
|
Yu WL, Guizzunti G, Foley TL, Burkart MD, La Clair JJ. An optimized immunoaffinity fluorescent method for natural product target elucidation. JOURNAL OF NATURAL PRODUCTS 2010; 73:1659-1666. [PMID: 20836515 DOI: 10.1021/np100371k] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/29/2023]
Abstract
Understanding the mode of action of small molecules is an integral facet of drug discovery. We report an optimized immunoaffinity fluorescent method that allows one to conduct parallel studies at both the cellular and molecular level using a single probe construct. Viability of the method has been evaluated analytically and applied using glycyrrhetic acid as a model.
Collapse
Affiliation(s)
- Wei-Luen Yu
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, USA
| | | | | | | | | |
Collapse
|
31
|
Gug F, Oumata N, Tribouillard-Tanvier D, Voisset C, Desban N, Bach S, Blondel M, Galons H. Synthesis of conjugates of 6-aminophenanthridine and guanabenz, two structurally unrelated prion inhibitors, for the determination of their cellular targets by affinity chromatography. Bioconjug Chem 2010; 21:279-88. [PMID: 20092293 DOI: 10.1021/bc900314n] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
The synthesis of affinity matrices for 6-aminophenanthridine (6AP) and 2,6-dichlorobenzylidenaminoguanidine (Guanabenz, GA), two unrelated prion inhibitors, is described. In both cases, the same simple spacer, epsilon-aminocaproylaminopentanol, was introduced by a Mitsunobu reaction and the choice of the anchoring position of the linker was determined by the study of the residual antiprion activity of the corresponding 6AP or GA conjugates. Very recently, these two affinity matrices were used for chromatography assays leading to the identification of ribosome (via the rRNA) as a common target of these two antiprion drugs. Here, we show, using competition experiments with Quinacrine (QC) and Chlorpromazine (CPZ), two other antiprion drugs, that QC, but not CPZ, may also directly target the rRNA.
Collapse
Affiliation(s)
- Fabienne Gug
- Laboratoire de Chimie Organique 2, INSERM U648, Universite Paris-Descartes, 4 avenue de l'Observatoire, 75270 Paris cedex 06, France
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Coley W, Kehn-Hall K, Van Duyne R, Kashanchi F. Novel HIV-1 therapeutics through targeting altered host cell pathways. Expert Opin Biol Ther 2009; 9:1369-82. [PMID: 19732026 DOI: 10.1517/14712590903257781] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/27/2023]
Abstract
The emergence of drug-resistant HIV-1 strains presents a challenge for the design of new drugs. Anti-HIV compounds currently in use are the subject of advanced clinical trials using either HIV-1 reverse transcriptase, viral protease or integrase inhibitors. Recent studies show an increase in the number of HIV-1 variants resistant to anti-retroviral agents in newly infected individuals. Targeting host cell factors involved in the regulation of HIV-1 replication might be one way to combat HIV-1 resistance to the currently available anti-viral agents. A specific inhibition of HIV-1 gene expression could be expected from the development of compounds targeting host cell factors that participate in the activation of the HIV-1 LTR promoter. Here we discuss how targeting the host can be accomplished either by using small molecules to alter the function of the host's proteins such as p53 or cdk9, or by utilizing new advances in siRNA therapies to knock down essential host factors such as CCR5 and CXCR4. Finally, we will discuss how the viral protein interactomes should be used to better design therapeutics against HIV-1.
Collapse
Affiliation(s)
- William Coley
- George Washington University, School of Medicine, Department of Microbiology, Immunology and Tropical Medicine, Washington, DC 20037, USA
| | | | | | | |
Collapse
|
33
|
Budin G, Dimala MM, Lamour V, Oudet P, Mioskowski C, Meunier S, Brino L, Wagner A. A Chemical Labeling Strategy for Proteomics under Nondenaturing Conditions. Chembiochem 2009; 11:79-82. [DOI: 10.1002/cbic.200900641] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/07/2023]
|
34
|
Saxena C, Bonacci TM, Huss KL, Bloem LJ, Higgs RE, Hale JE. Capture of Drug Targets from Live Cells Using a Multipurpose Immuno-Chemo-Proteomics Tool. J Proteome Res 2009; 8:3951-7. [DOI: 10.1021/pr900277x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Affiliation(s)
- Chaitanya Saxena
- Integrative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Tabetha M. Bonacci
- Integrative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Karen L. Huss
- Integrative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Laura J. Bloem
- Integrative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Richard E. Higgs
- Integrative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - John E. Hale
- Integrative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| |
Collapse
|
35
|
Corbel C, Haddoub R, Guiffant D, Lozach O, Gueyrard D, Lemoine J, Ratin M, Meijer L, Bach S, Goekjian P. Identification of potential cellular targets of aloisine A by affinity chromatography. Bioorg Med Chem 2009; 17:5572-82. [PMID: 19596197 DOI: 10.1016/j.bmc.2009.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2009] [Revised: 06/05/2009] [Accepted: 06/14/2009] [Indexed: 11/26/2022]
Abstract
Affinity chromatography was used to identify potential cellular targets of aloisine A (7-n-butyl-6-(4'-hydroxyphenyl)-5H-pyrrolo[2,3b]pyrazine), a potent inhibitor of cyclin-dependent kinases. This technique is based on the immobilization of the drug on a solid matrix, followed by identification of specifically bound proteins. To this end, both aloisine A and the protein-kinase inactive control N-methyl aloisine, bearing extended linker chains have been synthesized. We present the preparation of such analogues having the triethylene glycol chain at different positions of the molecule, as well as their immobilization on an agarose-based matrix. Affinity chromatography of various biological extracts on the aloisine matrices allowed the identification of both protein kinases and non-kinase proteins as potential cellular targets of aloisine.
Collapse
Affiliation(s)
- Caroline Corbel
- CNRS USR-3151, Protein Phosphorylation and Human Disease, Station Biologique, B.P.74, F-29682 Roscoff Cedex, Bretagne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Saxena C, Zhen E, Higgs RE, Hale JE. An immuno-chemo-proteomics method for drug target deconvolution. J Proteome Res 2008; 7:3490-7. [PMID: 18590316 DOI: 10.1021/pr800222q] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
Abstract
Chemical proteomics is an emerging technique for drug target deconvolution and profiling the toxicity of known drugs. With the use of this technique, the specificity of a small molecule inhibitor toward its potential targets can be characterized and information thus obtained can be used in optimizing lead compounds. Most commonly, small molecules are immobilized on solid supports and used as affinity chromatography resins to bind targets. However, it is difficult to evaluate the effect of immobilization on the affinity of the compounds to their targets. Here, we describe the development and application of a soluble probe where a small molecule was coupled with a peptide epitope which was used to affinity isolate binding proteins from cell lysate. The soluble probe allowed direct verification that the compound after coupling with peptide epitope retained its binding characteristics. The PKC-alpha inhibitor Bisindolylmaleimide-III was coupled with a peptide containing the FLAG epitope. Following incubation with cellular lysates, the compound and associated proteins were affinity isolated using anti-FLAG antibody beads. Using this approach, we identified the known Bisindolylmaleimide-III targets, PKC-alpha, GSK3-beta, CaMKII, adenosine kinase, CDK2, and quinine reductase type 2, as well as previously unidentified targets PKAC-alpha, prohibitin, VDAC and heme binding proteins. This method was directly compared to the solid-phase method (small molecule was immobilized to a solid support) providing an orthogonal strategy to aid in target deconvolution and help to eliminate false positives originating from nonspecific binding of the proteins to the matrix.
Collapse
Affiliation(s)
- Chaitanya Saxena
- Integrative Biology, Greenfield Laboratories, Eli Lilly and Company, Greenfield, IN 46140, USA.
| | | | | | | |
Collapse
|
37
|
Hübel K, Lessmann T, Waldmann H. Chemical biology--identification of small molecule modulators of cellular activity by natural product inspired synthesis. Chem Soc Rev 2008; 37:1361-74. [PMID: 18568162 DOI: 10.1039/b704729k] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023]
Abstract
The aim of this tutorial review is to introduce the reader to the concept, synthesis and application of natural product-inspired compound collections as an important field in chemical biology. This review will discuss how potentially interesting scaffolds can be identified (structural classification of natural products), synthesized in an appropriate manner (including stereoselective transformations for solid phase-bound compounds) and tested in biological assays (cell-based screening as well as biochemical in vitro assays). These approaches will provide the opportunity to identify new and interesting compounds as well as new targets for chemical biology and medicinal chemistry research.
Collapse
Affiliation(s)
- Katja Hübel
- Max-Planck-Institute of Molecular Physiology, Otto Hahn-Str. 11, 44227 Dortmund, Germany
| | | | | |
Collapse
|
38
|
Leroy D, Doerig C. Drugging the Plasmodium kinome: the benefits of academia-industry synergy. Trends Pharmacol Sci 2008; 29:241-9. [PMID: 18394721 DOI: 10.1016/j.tips.2008.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2007] [Revised: 02/25/2008] [Accepted: 02/25/2008] [Indexed: 01/21/2023]
Abstract
Malaria remains a major killer in many parts of the world. Recently, the development of nonprofit organisations aimed at fighting this deadly scourge incited academic and industrial scientists to merge their expertise in drug-target validation and lead discovery. Expectations are clear: identification and characterisation of new molecules showing high efficacy, low toxicity and little propensity to induce resistance in the parasite. In this context, protein kinase inhibitors represent an attractive possibility. Here, we compare traditional target-based drug-discovery approaches with innovative exploratory paths (parallel screening, cell-based assays, integrated systems biology and allosteric inhibition) and discuss the benefits of acadaemia-industry cooperation. Early characterisation of distribution, metabolism, pharmacokinetic (DMPK) and toxicology parameters are considered as well.
Collapse
Affiliation(s)
- Didier Leroy
- Merck-Serono International S.A., Geneva Research Center, 9, Chemin des Mines, Case postale 54, CH-1211 Genève 20, Switzerland.
| | | |
Collapse
|
39
|
Abstract
Drug discovery to lessen the burden of chronic renal failure and end-stage renal disease remains a principle goal of translational research in nephrology. In this review, we provide an overview of the current development of small molecule cyclin-dependent kinase (CDK)/glycogen synthase kinase-3 (GSK-3) inhibitors as therapeutic agents for parenchymal renal diseases. The emergence of this drug family has resulted from the recognition that CDKs and GSK-3s play critical roles in the progression and regression of many kidney diseases. CDK/GSK-3 inhibitors suppress pathogenic proliferation, apoptosis, and inflammation, and promote regeneration of injured tissue. Preclinical efficacy has now been demonstrated in mesangial proliferative glomerulonephritis, crescentic glomerulonephritis, collapsing glomerulopathy, proliferative lupus nephritis, polycystic kidney diseases, diabetic nephropathy, and several forms of acute kidney injury. Novel biomarkers of therapy are aiding the process of drug development. This review will highlight these advancements in renal therapeutics.
Collapse
|
40
|
Abstract
Recognition of some of the limitations of target-based drug discovery has recently led to the renaissance of a more holistic approach in which complex biological systems are investigated for phenotypic changes upon exposure to small molecules. The subsequent identification of the molecular targets that underlie an observed phenotypic response--termed target deconvolution--is an important aspect of current drug discovery, as knowledge of the molecular targets will greatly aid drug development. Here, the broad panel of experimental strategies that can be applied to target deconvolution is critically reviewed.
Collapse
|
41
|
Rix U, Hantschel O, Dürnberger G, Remsing Rix LL, Planyavsky M, Fernbach NV, Kaupe I, Bennett KL, Valent P, Colinge J, Köcher T, Superti-Furga G. Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 2007; 110:4055-63. [PMID: 17720881 DOI: 10.1182/blood-2007-07-102061] [Citation(s) in RCA: 517] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
The BCR-ABL tyrosine kinase inhibitor imatinib represents the current frontline therapy in chronic myeloid leukemia. Because many patients develop imatinib resistance, 2 second-generation drugs, nilotinib and dasatinib, displaying increased potency against BCR-ABL were developed. To predict potential side effects and novel medical uses, we generated comprehensive drug-protein interaction profiles by chemical proteomics for all 3 drugs. Our studies yielded 4 major findings: (1) The interaction profiles of the 3 drugs displayed strong differences and only a small overlap covering the ABL kinases. (2) Dasatinib bound in excess of 30 Tyr and Ser/Thr kinases, including major regulators of the immune system, suggesting that dasatinib might have a particular impact on immune function. (3) Despite the high specificity of nilotinib, the receptor tyrosine kinase DDR1 was identified and validated as an additional major target. (4) The oxidoreductase NQO2 was bound and inhibited by imatinib and nilotinib at physiologically relevant drug concentrations, representing the first nonkinase target of these drugs.
Collapse
MESH Headings
- Benzamides
- Dasatinib
- Discoidin Domain Receptor 1
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Drug Screening Assays, Antitumor
- Fusion Proteins, bcr-abl
- Humans
- Imatinib Mesylate
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Piperazines/chemistry
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/metabolism
- Proteomics
- Pyrimidines/chemistry
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Quinone Reductases/antagonists & inhibitors
- Quinone Reductases/metabolism
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor Protein-Tyrosine Kinases/metabolism
- Thiazoles/chemistry
- Thiazoles/pharmacology
- Thiazoles/therapeutic use
Collapse
Affiliation(s)
- Uwe Rix
- Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hantschel O, Rix U, Schmidt U, Bürckstümmer T, Kneidinger M, Schütze G, Colinge J, Bennett KL, Ellmeier W, Valent P, Superti-Furga G. The Btk tyrosine kinase is a major target of the Bcr-Abl inhibitor dasatinib. Proc Natl Acad Sci U S A 2007; 104:13283-8. [PMID: 17684099 PMCID: PMC1940229 DOI: 10.1073/pnas.0702654104] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/13/2023] Open
Abstract
Dasatinib is a small-molecule kinase inhibitor used for the treatment of imatinib-resistant chronic myelogenous leukemia (CML). We have analyzed the kinases targeted by dasatinib by using an unbiased chemical proteomics approach to detect binding proteins directly from lysates of CML cells. Besides Abl and Src kinases, we have identified the Tec kinases Btk and Tec, but not Itk, as major binders of dasatinib. The kinase activity of Btk and Tec, but not of Itk, was inhibited by nanomolar concentrations of dasatinib in vitro and in cultured cells. We identified the gatekeeper residue as the critical determinant of dasatinib susceptibility. Mutation of Thr-474 in Btk to Ile and Thr-442 in Tec to Ile conferred resistance to dasatinib, whereas mutation of the corresponding residue in Itk (Phe-435) to Thr sensitized the otherwise insensitive Itk to dasatinib. The configuration of this residue may be a predictor for dasatinib sensitivity across the kinome. Analysis of mast cells derived from Btk-deficient mice suggested that inhibition of Btk by dasatinib may be responsible for the observed reduction in histamine release upon dasatinib treatment. Furthermore, dasatinib inhibited histamine release in primary human basophils and secretion of proinflammatory cytokines in immune cells. The observed inhibition of Tec kinases by dasatinib predicts immunosuppressive (side) effects of this drug and may offer therapeutic opportunities for inflammatory and immunological disorders.
Collapse
Affiliation(s)
- Oliver Hantschel
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
| | - Uwe Rix
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
| | - Uwe Schmidt
- Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria; and
| | - Tilmann Bürckstümmer
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
| | - Michael Kneidinger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Gregor Schütze
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
| | - Jacques Colinge
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
| | - Keiryn L. Bennett
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
| | - Wilfried Ellmeier
- Institute of Immunology, Medical University of Vienna, Lazarettgasse 19, 1090 Vienna, Austria; and
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Giulio Superti-Furga
- *Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 19, 1090 Vienna, Austria
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
43
|
Tribouillard D, Gug F, Galons H, Bach S, Saupe SJ, Blondel M. Antiprion drugs as chemical tools to uncover mechanisms of prion propagation. Prion 2007; 1:48-52. [PMID: 19164923 DOI: 10.4161/pri.1.1.4053] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022] Open
Abstract
A number of drugs active against prions either in vitro, in cellular systems or in vivo in animal models have been isolated in various screening assays. In this minireview, we would like to suggest, that in addition to their direct interest as potential therapeutic agents, these molecules could be used as original research tools to understand prion propagation. The use of antiprion compounds as tool to understand fundamentals of prion propagation relies on reverse screening approaches. These global genetic and/or biochemical approaches aim to identify the intracellular target(s) and mechanism of action of the drugs. Once those are known, the biological activity of the compounds can be optimized on a rational basis, their potential side effects understood and minimized. In vitro enzyme-based screening assays can then be designed to allow discovery of new, more potent and selective molecules. Here we describe the main comprehensive biochemical and genetical approaches to realize reverse screening approaches based on antiprion drugs. We will finish by discussing the interest of using drug inactivation of specific targets as a substitute to genetic inactivation.
Collapse
Affiliation(s)
- Déborah Tribouillard
- INSERM U613, CHU de Brest, Faculté de Médecine de l'Université de Bretagne Occidentale, 46 rue Félix Le Dantec, Brest, France
| | | | | | | | | | | |
Collapse
|