1
|
Wang S, Pang Z, Fan H, Tong Y. Advances in anti-EV-A71 drug development research. J Adv Res 2024; 56:137-156. [PMID: 37001813 PMCID: PMC10834817 DOI: 10.1016/j.jare.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/05/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Enterovirus A71 (EV-A71) is capable of causing hand, foot and mouth disease (HFMD), which may lead to neurological sequelae and even death. As EV-A71 is resistant to environmental changes and mutates easily, there is still a lack of effective treatments or globally available vaccines. AIM OF REVIEW For more than 50 years since the HFMD epidemic, related drug research has been conducted. Progress in this area can promote the further application of existing potential drugs and develop more efficient and safe antiviral drugs, and provide useful reference for protecting the younger generation and maintaining public health security. KEY SCIENTIFIC CONCEPTS OF REVIEW At present, researchers have identified hundreds of EV-A71 inhibitors based on screening repurposed drugs, targeted structural design, and rational modification of previously effective drugs as the main development strategies. This review systematically introduces the current potential drugs to inhibit EV-A71 infection, including viral inhibitors targeting key sites such as the viral capsid, RNA-dependent RNA polymerase (RdRp), 2C protein, internal ribosome entry site (IRES), 3C proteinase (3Cpro), and 2A proteinase (2Apro), starting from each stage of the viral life cycle. Meanwhile, the progress of host-targeting antiviral drugs and their development are summarized in terms of regulating host immunity, inhibiting autophagy or apoptosis, and regulating the cellular redox environment. In addition, the current clinical methods for the prevention and treatment of HFMD are summarized and discussed with the aim of providing support and recommendations for the treatment of enterovirus infections including EV-A71.
Collapse
Affiliation(s)
- Shuqi Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China; Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
2
|
Wei Y, Liu H, Hu D, He Q, Yao C, Li H, Hu K, Wang J. Recent Advances in Enterovirus A71 Infection and Antiviral Agents. J Transl Med 2024; 104:100298. [PMID: 38008182 DOI: 10.1016/j.labinv.2023.100298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/29/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023] Open
Abstract
Enterovirus A71 (EV-A71) is one of the major causative agents of hand, foot, and mouth disease (HFMD) that majorly affects children. Most of the time, HFMD is a mild disease but can progress to severe complications, such as meningitis, brain stem encephalitis, acute flaccid paralysis, and even death. HFMD caused by EV-A71 has emerged as an acutely infectious disease of highly pathogenic potential in the Asia-Pacific region. In this review, we introduced the properties and life cycle of EV-A71, and the pathogenesis and the pathophysiology of EV-A71 infection, including tissue tropism and host range of virus infection, the diseases caused by the virus, as well as the genes and host cell immune mechanisms of major diseases caused by enterovirus 71 (EV-A71) infection, such as encephalitis and neurologic pulmonary edema. At the same time, clinicopathologic characteristics of EV-A71 infection were introduced. There is currently no specific medication for EV-A71 infection, highlighting the urgency and significance of developing suitable anti-EV-A71 agents. This overview also summarizes the targets of existing anti-EV-A71 agents, including virus entry, translation, polyprotein processing, replication, assembly and release; interferons; interleukins; the mitogen-activated protein kinase, phosphatidylinositol 3-kinase, and protein kinase B signaling pathways; the oxidative stress pathway; the ubiquitin-proteasome system; and so on. Furthermore, it overviews the effects of natural products, monoclonal antibodies, and RNA interference against EV-A71. It also discusses issues limiting the research of antiviral drugs. This review is a systematic and comprehensive summary of the mechanism and pathological characteristics of EV-A71 infection, the latest progress of existing anti-EV-A71 agents. It would provide better understanding and guidance for the research and application of EV-A71 infection and antiviral inhibitors.
Collapse
Affiliation(s)
- Yanhong Wei
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Huihui Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Da Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Qun He
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Chenguang Yao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Hanluo Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China
| | - Kanghong Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, China.
| | - Jun Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Le Rouzic A, Fix J, Vinck R, Kappler-Gratias S, Volmer R, Gallardo F, Eléouët JF, Keck M, Cintrat JC, Barbier J, Gillet D, Galloux M. A New Derivative of Retro-2 Displays Antiviral Activity against Respiratory Syncytial Virus. Int J Mol Sci 2023; 25:415. [PMID: 38203585 PMCID: PMC10778932 DOI: 10.3390/ijms25010415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Human respiratory syncytial virus (hRSV) is the most common cause of bronchiolitis and pneumonia in newborns, with all children being infected before the age of two. Reinfections are very common throughout life and can cause severe respiratory infections in the elderly and immunocompromised adults. Although vaccines and preventive antibodies have recently been licensed for use in specific subpopulations of patients, there is still no therapeutic treatment commonly available for these infections. Here, we investigated the potential antiviral activity of Retro-2.2, a derivative of the cellular retrograde transport inhibitor Retro-2, against hRSV. We show that Retro-2.2 inhibits hRSV replication in cell culture and impairs the ability of hRSV to form syncytia. Our results suggest that Retro-2.2 treatment affects virus spread by disrupting the trafficking of the viral de novo synthetized F and G glycoproteins to the plasma membrane, leading to a defect in virion morphogenesis. Taken together, our data show that targeting intracellular transport may be an effective strategy against hRSV infection.
Collapse
Affiliation(s)
- Adrien Le Rouzic
- INRAE Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St Quentin, 78350 Jouy-en-Josas, France; (A.L.R.); (J.F.); (J.-F.E.)
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.V.); (M.K.); (J.B.)
| | - Jenna Fix
- INRAE Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St Quentin, 78350 Jouy-en-Josas, France; (A.L.R.); (J.F.); (J.-F.E.)
| | - Robin Vinck
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.V.); (M.K.); (J.B.)
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris-Saclay, 91191 Gif-sur-Yvette, France;
| | | | - Romain Volmer
- INRAE, IHAP, UMR 1225, ENVT, 31300 Toulouse, France;
| | - Franck Gallardo
- NeoVirTech SAS, 1 Place Pierre Potier, 31000 Toulouse, France; (S.K.-G.); (F.G.)
| | - Jean-François Eléouët
- INRAE Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St Quentin, 78350 Jouy-en-Josas, France; (A.L.R.); (J.F.); (J.-F.E.)
| | - Mathilde Keck
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.V.); (M.K.); (J.B.)
| | - Jean-Christophe Cintrat
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris-Saclay, 91191 Gif-sur-Yvette, France;
| | - Julien Barbier
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.V.); (M.K.); (J.B.)
| | - Daniel Gillet
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (R.V.); (M.K.); (J.B.)
| | - Marie Galloux
- INRAE Unité de Virologie et Immunologie Moléculaires (VIM), Université Paris-Saclay-Versailles St Quentin, 78350 Jouy-en-Josas, France; (A.L.R.); (J.F.); (J.-F.E.)
| |
Collapse
|
4
|
Huang X, Li J, Hong Y, Jiang C, Wu J, Wu M, Sheng R, Liu H, Sun J, Xin Y, Su W. Antiviral effects of the petroleum ether extract of Tournefortia sibirica L. against enterovirus 71 infection in vitro and in vivo. Front Pharmacol 2022; 13:999798. [PMID: 36523495 PMCID: PMC9744809 DOI: 10.3389/fphar.2022.999798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/16/2022] [Indexed: 09/22/2023] Open
Abstract
Enterovirus 71 (EV71) is the major cause of severe hand, foot, and mouth disease (HFMD). Compared to other HFMD pathogens, like coxsackievirus A16 (CVA16), EV71 can invade the central nervous system and cause permanent damage. At present, there are no available antivirals against EV71 for clinical treatment. Herein, multiple Chinese botanical drugs were collected, and 47 types of botanical extracts were extracted using aqueous solutions and organic solvents. Based on the cytopathic effect inhibition assay, petroleum ether extract of Tournefortia sibirica L. (PE-TS) demonstrated 97.25% and 94.75% inhibition rates for EV71 infection (at 250 μg/ml) and CVA16 infection (at 125 μg/ml), respectively, with low cytotoxicity. Preliminary mechanistic studies showed that PE-TS inhibits replication of EV71 genomic RNA and synthesis of the EV71 protein. The released extracellular EV71 progeny virus titer decreased by 3.75 lg under PE-TS treatment. Furthermore, using a newborn mouse model, PE-TS treatment protected 70% and 66.7% of mice from lethal dose EV71 intracranial challenge via administration of intraperitoneal injection at 0.4 mg/g and direct lavage at 0.8 mg/g, respectively. The chemical constituents of the PE-TS were analyzed by Gas Chromatography-Mass Spectrometer (GC-MS), and a total of 60 compounds were identified. Compound-target network analysis and molecular docking implied potential bioactive compounds and their protein targets against EV71 associated pathology. The present study identified antiviral effects of PE-TS against EV71/CVA16 infection in vitro and EV71 infection in vivo, providing a potential antiviral botanical drug extract candidate for HFMD drug development.
Collapse
Affiliation(s)
- Xinyu Huang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiemin Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yan Hong
- Key Laboratory for Mongolian Medicine R&D Engineering of the Ministry of Education, School of Mongolian Medicine and Pharmacy, Inner Mongolia Minzu University, Tongliao, China
| | - Chenghan Jiang
- College of Agriculture, Yanbian University, Yanji, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Min Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Rui Sheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Hongtao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jie Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Ying Xin
- Key Laboratory for Mongolian Medicine R&D Engineering of the Ministry of Education, School of Mongolian Medicine and Pharmacy, Inner Mongolia Minzu University, Tongliao, China
| | - Weiheng Su
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
5
|
In Vivo Sustained Release of the Retrograde Transport Inhibitor Retro-2.1 Formulated in a Thermosensitive Hydrogel. Int J Mol Sci 2022; 23:ijms232314611. [PMID: 36498939 PMCID: PMC9735573 DOI: 10.3390/ijms232314611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022] Open
Abstract
A recently developed inhibitor of retrograde transport, namely Retro-2.1, proved to be a potent and broad-spectrum lead in vitro against intracellular pathogens, such as toxins, parasites, intracellular bacteria and viruses. To circumvent its low aqueous solubility, a formulation in poly(ethylene glycol)-block-poly(D,L)lactide micelle nanoparticles was developed. This formulation enabled the study of the pharmacokinetic parameters of Retro-2.1 in mice following intravenous and intraperitoneal injections, revealing a short blood circulation time, with an elimination half-life of 5 and 6.7 h, respectively. To explain the poor pharmacokinetic parameters, the metabolic stability of Retro-2.1 was studied in vitro and in vivo, revealing fast cytochrome-P-450-mediated metabolism into a less potent hydroxylated analogue. Subcutaneous injection of Retro-2.1 formulated in a biocompatible and bioresorbable polymer-based thermosensitive hydrogel allowed for sustained release of the drug, with an elimination half-life of 19 h, and better control of its metabolism. This study provides a guideline on how to administer this promising lead in vivo in order to study its efficacy.
Collapse
|
6
|
Cui G, Wang H, Yang C, Zhou X, Wang J, Wang T, Ma T. Berberine prevents lethal EV71 neurological infection in newborn mice. Front Pharmacol 2022; 13:1027566. [PMID: 36386168 PMCID: PMC9640474 DOI: 10.3389/fphar.2022.1027566] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/14/2022] [Indexed: 12/01/2022] Open
Abstract
Enterovirus 71 (EV71) is the major pathogen causing fatal neurological complications of hand, foot, and mouth disease (HFMD) in young children. Currently no effective antiviral therapy is available. In the present study, we found that natural compound Berberine (BBR) displayed potent inhibitory effects on EV71 replication in various neural cells (IC50 of 2.79–4.03 μM). In a newborn mouse model of lethal EV71 infection, Berberine at 2–5 mg/kg markedly reduced mortality and clinical scores. Consistently, the replication of EV71 and pathological changes were attenuated in various infected organs including brain and lung with BBR treatment. Interestingly, EV71 infection in the brain mainly localized in the peripheral zone of brainstem and largely in astrocytes. Primary culture of astrocytes from newborn mouse brain confirmed the efficient EV71 replication that was mostly inhibited by BBR treatment at 5 μM. Further investigations revealed remarkably elevated cellular reactive oxygen species (ROS) levels that coincided with EV71 replication in primary cultured astrocytes and various cell lines. BBR largely abolished the virus-elevated ROS production and greatly diminished EV71 replication by up-regulating NFE2 like bZIP transcription factor 2 (Nrf2) via the kelch like ECH associated protein 1 (Keap)-Nrf2 axis. The nuclear localization of Nrf2 and expression of downstream antioxidant enzymes heme oxygenase 1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1) were increased significantly by BBR treatment. Collectively, our findings revealed that BBR prevents lethal EV71 neurological infection via inhibiting virus replication through regulating Keap-Nrf2 axis and ROS generation in astrocytes of brainstem, thus providing a potential antiviral treatment for severe EV71 infection associated with neurological complications.
Collapse
|
7
|
Kang N, Gao H, He L, Liu Y, Fan H, Xu Q, Yang S. Ginsenoside Rb1 is an immune-stimulatory agent with antiviral activity against enterovirus 71. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113401. [PMID: 32980486 DOI: 10.1016/j.jep.2020.113401] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to the theory of traditional Chinese medicine, the main pathogenesis of severe hand, foot and mouth disease (HFMD) is that the heat and wet poisons are deeply trapped in the viscera, which causes the deficiency of Qi and Yin in the patient's body. Ginsenoside Rb1 (Rb1) is the most abundant triterpenoid saponin in Panax quinquefolius L., which has the function of Qi-invigorating and Yin-nourishing. Enterovirus 71 (EV71) is one of the causative pathogens of HFMD, especially the form associated with some lethal complications. Therefore, the therapeutic effect of Rb1 on this disease caused by EV71 infection is worth exploring. AIM OF THE STUDY We explored the effective antiviral activities of Rb1 against EV71 in vitro and in vivo and investigated its preliminary antiviral mechanisms. MATERIAL AND METHODS EV71-infected two-day-old suckling mice model was employed to detect the antiviral effects of Rb1 in vivo. To detect the antiviral effects of Rb1 in vitro, cytopathic effect (CPE) reduction assay was performed in EV71-infected Rhabdomyosarcoma (RD) cells. Interferon (IFN)-β interference experiment was employed to detect the antiviral mechanism of Rb1. RESULTS In this paper, we first found that Rb1 exhibited strong antiviral activities in EV71-infected suckling mice when compared to those of ribavirin. Administration of Rb1 reduced the CPE of EV71-infected RD cells in a dose-dependent manner. Moreover, EV71-induced viral protein-1 (VP-1) expression was significantly reduced by Rb1 administration in vitro and in vivo. Furthermore, Rb1 treatment could induce high cellular and humoral immune responses in vivo. Meanwhile, Rb1 contributed to the enhanced Type I IFN responses and IFN-β knockdown reversed the antiviral activity of Rb1 in vitro. CONCLUSION In summary, our findings suggest that Rb1 is an immune-stimulatory agent and provide an insight into therapeutic potentials of Rb1 for the treatment of EV71 infection.
Collapse
Affiliation(s)
- Naixin Kang
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Luan He
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| | - Yanli Liu
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| | - Handong Fan
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, 310036, China.
| | - Qiongming Xu
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China; College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Shilin Yang
- College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
8
|
Identification of an Antiviral Compound from the Pandemic Response Box that Efficiently Inhibits SARS-CoV-2 Infection In Vitro. Microorganisms 2020; 8:microorganisms8121872. [PMID: 33256227 PMCID: PMC7760777 DOI: 10.3390/microorganisms8121872] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
With over 50 million currently confirmed cases worldwide, including more than 1.3 million deaths, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has a major impact on the economy and health care system. Currently, limited prophylactic or therapeutic intervention options are available against SARS-CoV-2. In this study, 400 compounds from the antimicrobial “pandemic response box” library were screened for inhibiting properties against SARS-CoV-2. An initial screen on Vero E6 cells identified five compounds that inhibited SARS-CoV-2 replication. However, validation of the selected hits in a human lung cell line highlighted that only a single compound, namely Retro-2.1, efficiently inhibited SARS-CoV-2 replication. Additional analysis revealed that the antiviral activity of Retro-2.1 occurs at a post-entry stage of the viral replication cycle. Combined, these data demonstrate that stringent in vitro screening of preselected compounds in multiple cell lines refines the rapid identification of new potential antiviral candidate drugs targeting SARS-CoV-2.
Collapse
|
9
|
The macrophage microtubule network acts as a key cellular controller of the intracellular fate of Leishmania infantum. PLoS Negl Trop Dis 2020; 14:e0008396. [PMID: 32722702 PMCID: PMC7386624 DOI: 10.1371/journal.pntd.0008396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/16/2020] [Indexed: 11/19/2022] Open
Abstract
The parasitophorous vacuoles (PVs) that insulate Leishmania spp. in host macrophages are vacuolar compartments wherein promastigote forms differentiate into amastigote that are the replicative form of the parasite and are also more resistant to host responses. We revisited the biogenesis of tight-fitting PVs that insulate L. infantum in promastigote-infected macrophage-like RAW 264.7 cells by time-dependent confocal laser multidimensional imaging analysis. Pharmacological disassembly of the cellular microtubule network and silencing of the dynein gene led to an impaired interaction of L. infantum-containing phagosomes with late endosomes and lysosomes, resulting in the tight-fitting parasite-containing phagosomes never transforming into mature PVs. Analysis of the shape of the L. infantum parasite within PVs, showed that factors that impair promastigote-amastigote differentiation can also result in PVs whose maturation is arrested. These findings highlight the importance of the MT-dependent interaction of L. infantum-containing phagosomes with the host macrophage endolysosomal pathway to secure the intracellular fate of the parasite. Kinetoplastid parasites of the genus Leishmania are responsible for a diverse spectrum of mammalian infectious diseases, the leishmaniases, including cutaneous, mucocutaneous, and mucosal pathologies. Infectious metacyclic promastigotes of infected female Phlebotomus sandflies are injected into the host at the site of the bite during the sandfly blood meal, after which they are internalized by host professional phagocytic neutrophils and macrophages. Leishmania infantum is an etiological agent of potentially fatal visceral pathology. This study molecularly dissects the maturation of L. infantum-containing phagosomes/parasitophorous vacuoles (PVs) in host macrophages. We reveal the requirement of vacuolar movement along macrophage microtubule tracks for the phagosome trafficking toward the endolysosomal pathway necessary for the development of the mature tight-fitting PV crucial for L. infantum survival and proliferation.
Collapse
|
10
|
Abdelkafi H, Michau A, Pons V, Ngadjeua F, Clerget A, Ait Ouarab L, Buisson DA, Montoir D, Caramelle L, Gillet D, Barbier J, Cintrat JC. Structure-Activity Relationship Studies of Retro-1 Analogues against Shiga Toxin. J Med Chem 2020; 63:8114-8133. [PMID: 32648758 DOI: 10.1021/acs.jmedchem.0c00298] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High-throughput screening has shown that Retro-1 inhibits ricin and Shiga toxins by diminishing their intracellular trafficking via the retrograde route, from early endosomes to the Golgi apparatus. To improve the activity of Retro-1, a structure-activity relationship (SAR) study was undertaken and yielded an analogue with a roughly 70-fold better half-maximal effective concentration (EC50) against Shiga toxin cytotoxicity measured in a cell protein synthesis assay.
Collapse
Affiliation(s)
- Hajer Abdelkafi
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France
| | - Aurélien Michau
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - Valérie Pons
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France
| | - Flora Ngadjeua
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - Alexandra Clerget
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - Lilia Ait Ouarab
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France
| | - David-Alexandre Buisson
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France
| | - David Montoir
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France
| | - Lucie Caramelle
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - Daniel Gillet
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - Julien Barbier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, France
| | - Jean-Christophe Cintrat
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, France
| |
Collapse
|
11
|
Functional dissection of the retrograde Shiga toxin trafficking inhibitor Retro-2. Nat Chem Biol 2020; 16:327-336. [PMID: 32080624 PMCID: PMC7039708 DOI: 10.1038/s41589-020-0474-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/10/2020] [Indexed: 11/29/2022]
Abstract
The retrograde transport inhibitor Retro-2 has a protective effect on cells and in mice against Shiga-like toxins and ricin. Retro-2 causes toxin accumulation in early endosomes, and relocalization of the Golgi SNARE protein syntaxin-5 to the endoplasmic reticulum. The molecular mechanisms by which this is achieved remain unknown. Here, we show that Retro-2 targets the endoplasmic reticulum exit site component Sec16A, affecting anterograde transport of syntaxin-5 from the endoplasmic reticulum to the Golgi. The formation of canonical SNARE complexes involving syntaxin-5 is not affected in Retro-2-treated cells. In contrast, the interaction of syntaxin-5 with a newly discovered binding partner, the retrograde trafficking chaperone GPP130, is abolished, and we show that GPP130 must indeed bind to syntaxin-5 to drive Shiga toxin transport from endosomes to the Golgi. We thereby identify Sec16A as a druggable target, and provide evidence for a non-SNARE function for syntaxin-5 in interaction with the GPP130.
Collapse
|
12
|
Morgens DW, Chan C, Kane AJ, Weir NR, Li A, Dubreuil MM, Tsui CK, Hess GT, Lavertu A, Han K, Polyakov N, Zhou J, Handy EL, Alabi P, Dombroski A, Yao D, Altman RB, Sello JK, Denic V, Bassik MC. Retro-2 protects cells from ricin toxicity by inhibiting ASNA1-mediated ER targeting and insertion of tail-anchored proteins. eLife 2019; 8:48434. [PMID: 31674906 PMCID: PMC6858068 DOI: 10.7554/elife.48434] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
The small molecule Retro-2 prevents ricin toxicity through a poorly-defined mechanism of action (MOA), which involves halting retrograde vesicle transport to the endoplasmic reticulum (ER). CRISPRi genetic interaction analysis revealed Retro-2 activity resembles disruption of the transmembrane domain recognition complex (TRC) pathway, which mediates post-translational ER-targeting and insertion of tail-anchored (TA) proteins, including SNAREs required for retrograde transport. Cell-based and in vitro assays show that Retro-2 blocks delivery of newly-synthesized TA-proteins to the ER-targeting factor ASNA1 (TRC40). An ASNA1 point mutant identified using CRISPR-mediated mutagenesis abolishes both the cytoprotective effect of Retro-2 against ricin and its inhibitory effect on ASNA1-mediated ER-targeting. Together, our work explains how Retro-2 prevents retrograde trafficking of toxins by inhibiting TA-protein targeting, describes a general CRISPR strategy for predicting the MOA of small molecules, and paves the way for drugging the TRC pathway to treat broad classes of viruses known to be inhibited by Retro-2.
Collapse
Affiliation(s)
- David W Morgens
- Department of Genetics, Stanford University, Stanford, United States
| | - Charlene Chan
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Andrew J Kane
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Nicholas R Weir
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Amy Li
- Department of Genetics, Stanford University, Stanford, United States
| | | | - C Kimberly Tsui
- Department of Genetics, Stanford University, Stanford, United States
| | - Gaelen T Hess
- Department of Genetics, Stanford University, Stanford, United States
| | - Adam Lavertu
- Biomedical Informatics Training Program, Stanford University, Stanford, United States
| | - Kyuho Han
- Department of Genetics, Stanford University, Stanford, United States
| | - Nicole Polyakov
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Jing Zhou
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Emma L Handy
- Department of Chemistry, Brown University, Providence, United States
| | - Philip Alabi
- Department of Chemistry, Brown University, Providence, United States
| | - Amanda Dombroski
- Department of Chemistry, Brown University, Providence, United States
| | - David Yao
- Department of Genetics, Stanford University, Stanford, United States
| | - Russ B Altman
- Department of Genetics, Stanford University, Stanford, United States.,Bioengineering, Stanford University, Stanford, United States
| | - Jason K Sello
- Department of Chemistry, Brown University, Providence, United States
| | - Vladimir Denic
- Department of Molecular and Cellular Biology, Northwest Labs, Harvard University, Cambridge, United States
| | - Michael C Bassik
- Department of Genetics, Stanford University, Stanford, United States.,Program in Cancer Biology, Stanford University, Stanford, United States.,Stanford University Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford, United States
| |
Collapse
|
13
|
Lin JY, Kung YA, Shih SR. Antivirals and vaccines for Enterovirus A71. J Biomed Sci 2019; 26:65. [PMID: 31481071 PMCID: PMC6720414 DOI: 10.1186/s12929-019-0560-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/23/2019] [Indexed: 01/23/2023] Open
Abstract
Enterovirus A71 (EV-A71) is an important emerging virus posing a threat to children under five years old. EV-A71 infection in infants or young children can cause hand-foot-and-mouth disease, herpangina, or severe neurological complications. However, there are still no effective antivirals for treatment of these infections. In this review, we summarize the antiviral compounds developed to date based on various targets of the EV-A71 life cycle. Moreover, development of a vaccine would be the most effective approach to prevent EV-A71 infection. Therefore, we also summarize the development and clinical progress of various candidate EV-A71 vaccines, including inactivated whole virus, recombinant VP1 protein, synthetic peptides, viral-like particles, and live attenuated vaccines.
Collapse
Affiliation(s)
- Jing-Yi Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Yu-An Kung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
14
|
Antiviral Efficacy of Flavonoids against Enterovirus 71 Infection in Vitro and in Newborn Mice. Viruses 2019; 11:v11070625. [PMID: 31284698 PMCID: PMC6669683 DOI: 10.3390/v11070625] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 12/26/2022] Open
Abstract
Enterovirus 71 (EV71) infection is known to cause hand, foot, and mouth disease (HFMD), which is associated with neurological complications; however, there is currently no effective treatment for this infection. Flavonoids are a large group of naturally occurring compounds with multiple bioactivities, and the inhibitory effects of several flavonoids against EV71 have been studied in cell cultures; however, to date, there are no reported data on their effects in animal models. In this study, we confirmed the in vitro activities of eight flavonoids against EV71 infection, based on the inhibition of cytopathic effects. Moreover, these flavonoids were found to reduce viral genomic RNA replication and protein synthesis. We further demonstrated the protective efficacy of these flavonoids in newborn mice challenged with a lethal dose of EV71. Apigenin, luteolin, kaempferol, formononetin, and penduletin conferred survival protection of 88.89%, 91.67%, 88.89%, 75%, and 66.67%, respectively, from the lethal EV71 challenge. In addition, isorhamnetin provided the highest mice survival protection of 100% at a dose of 10 mg/kg. This study, to the best of our knowledge, is the first to evaluate the in vivo anti-EV7l activities of multiple flavonoids, and we accordingly identified flavonoids as potential leading compounds for anti-EV71 drug development.
Collapse
|
15
|
Desai D, Lauver M, Ostman A, Cruz L, Ferguson K, Jin G, Roper B, Brosius D, Lukacher A, Amin S, Buchkovich N. Inhibition of diverse opportunistic viruses by structurally optimized retrograde trafficking inhibitors. Bioorg Med Chem 2019; 27:1795-1803. [PMID: 30890396 DOI: 10.1016/j.bmc.2019.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/04/2019] [Accepted: 03/12/2019] [Indexed: 10/27/2022]
Abstract
Opportunistic viruses are a major problem for immunosuppressed individuals, particularly following organ or stem cell transplantation. Current treatments are non-existent or suffer from problems such as high toxicity or development of resistant strains. We previously published that a trafficking inhibitor that targets a host protein greatly reduces the replication of human cytomegalovirus. This inhibitor was also shown to be moderately effective against polyomaviruses, another family of opportunistic viruses. We have developed a panel of analogues for this inhibitor and have shown that these analogues maintain their high efficacy against HCMV, while substantially lowering the concentration required to inhibit polyomavirus replication. By targeting a host protein these compounds are able to inhibit the replication of two very different viruses. These observations open up the possibility of pan-viral inhibitors for immunosuppressed individuals that are effective against multiple, diverse opportunistic viruses.
Collapse
Affiliation(s)
- Dhimant Desai
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Matthew Lauver
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Alexandria Ostman
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Linda Cruz
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Kevin Ferguson
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Ge Jin
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Brianne Roper
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Daniel Brosius
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Aron Lukacher
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Shantu Amin
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Nick Buchkovich
- Department of Microbiology & Immunology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, United States.
| |
Collapse
|
16
|
Immunocompetent and Immunodeficient Mouse Models for Enterovirus 71 Pathogenesis and Therapy. Viruses 2018; 10:v10120674. [PMID: 30487421 PMCID: PMC6316343 DOI: 10.3390/v10120674] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
Enterovirus 71 (EV71) is a global health threat. Children infected with EV71 could develop hand-foot-and-mouth disease (HFMD), encephalitis, paralysis, pulmonary edema, and death. At present, no effective treatment for EV71 is available. We reviewed here various mouse models for EV71 pathogenesis and therapy. Earlier studies relied on the use of mouse-adapted EV71 strains. To avoid artificial mutations arising de novo during the serial passages, recent studies used EV71 clinical isolates without adaptation. Several human receptors for EV71 were shown to facilitate viral entry in cell culture. However, in vivo infection with human SCARB2 receptor transgenic mice appeared to be more limited to certain strains and genotypes of EV71. Efficacy of oral infection in these transgenic models is extremely low. Intriguingly, despite the lack of human receptors, immunodeficient neonatal mouse models can still be infected with EV71 clinical isolates via oral or intraperitoneal routes. Crossbreeding between SCARB2 transgenic and stat1 knockout mice generated a more sensitive and user-friendly hybrid mouse model. Infected hybrid mice developed a higher incidence and earlier onset of CNS disease and death. Different pathogenesis profiles were observed in models deficient in various arms of innate or humoral immunity. These models are being actively used for antiviral research.
Collapse
|
17
|
Inhibition of Retrograde Transport Limits Polyomavirus Infection In Vivo. mSphere 2017; 2:mSphere00494-17. [PMID: 29152583 PMCID: PMC5687923 DOI: 10.1128/mspheredirect.00494-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023] Open
Abstract
PyVs can cause significant morbidity and mortality in immunocompromised individuals. No clinically efficacious anti-PyV therapeutic agents are available. A recently identified inhibitor of retrograde transport, Retro-2cycl, blocks movement of PyV virion-containing vesicles from early endosomes to the endoplasmic reticulum, an early step in the PyV life cycle. Retro-2cycl and its derivatives have been shown to inhibit infection by human PyVs in tissue culture. Here, we demonstrate that a derivative of Retro-2cycl, Retro-2.1, reduces infection by MuPyV in the kidneys of acutely infected mice. Mimicking the common clinical scenario of PyV resurgence, we further show that MuPyV levels increase in the kidneys of immunocompromised, persistently infected mice and that this increase is inhibited by Retro-2.1. These data provide the first evidence for control of a natural PyV infection in vivo by administration of an inhibitor of retrograde transport. Polyomaviruses (PyVs) silently infect most humans, but they can cause life-threatening diseases in immunocompromised individuals. The JC polyomavirus (JCPyV) induces progressive multifocal leukoencephalopathy, a severe demyelinating disease in multiple sclerosis patients receiving immunomodulatory therapy, and BK polyomavirus (BKPyV)-associated nephropathy is a major cause of kidney allograft failure. No effective anti-PyV agents are available. Several compounds have been reported to possess anti-PyV activity in vitro, but none have shown efficacy in clinical trials. Productive PyV infection involves usurping the cellular retrograde vesicular transport pathway to enable endocytosed virions to navigate to the endoplasmic reticulum where virion uncoating begins. Compounds inhibiting this pathway have been shown to reduce infection by simian virus 40 (SV40), JCPyV, and BKPyV in tissue culture. In this study, we investigated the potential of Retro-2.1, a retrograde transport inhibitor, to limit infection by mouse polyomavirus (MuPyV) in vivo. We found that Retro-2.1 significantly reduced MuPyV levels in the kidney during acute infection without affecting renal function or the MuPyV-specific CD8 T cell response. To approximate the clinical setting of PyV resurgence in immunocompromised hosts, we showed that antibody-mediated depletion of T cells in persistently infected mice elevated MuPyV levels in the kidney and that Retro-2.1 blunted this increase in virus levels. In summary, these data indicate that inhibition of retrograde vesicular transport in vivo controls infection in a natural PyV mouse model and supports development of these compounds as potential therapeutic agents for individuals at risk for human PyV-associated diseases. IMPORTANCE PyVs can cause significant morbidity and mortality in immunocompromised individuals. No clinically efficacious anti-PyV therapeutic agents are available. A recently identified inhibitor of retrograde transport, Retro-2cycl, blocks movement of PyV virion-containing vesicles from early endosomes to the endoplasmic reticulum, an early step in the PyV life cycle. Retro-2cycl and its derivatives have been shown to inhibit infection by human PyVs in tissue culture. Here, we demonstrate that a derivative of Retro-2cycl, Retro-2.1, reduces infection by MuPyV in the kidneys of acutely infected mice. Mimicking the common clinical scenario of PyV resurgence, we further show that MuPyV levels increase in the kidneys of immunocompromised, persistently infected mice and that this increase is inhibited by Retro-2.1. These data provide the first evidence for control of a natural PyV infection in vivo by administration of an inhibitor of retrograde transport.
Collapse
|
18
|
ABMA, a small molecule that inhibits intracellular toxins and pathogens by interfering with late endosomal compartments. Sci Rep 2017; 7:15567. [PMID: 29138439 PMCID: PMC5686106 DOI: 10.1038/s41598-017-15466-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/24/2017] [Indexed: 12/30/2022] Open
Abstract
Intracellular pathogenic microorganisms and toxins exploit host cell mechanisms to enter, exert their deleterious effects as well as hijack host nutrition for their development. A potential approach to treat multiple pathogen infections and that should not induce drug resistance is the use of small molecules that target host components. We identified the compound 1-adamantyl (5-bromo-2-methoxybenzyl) amine (ABMA) from a cell-based high throughput screening for its capacity to protect human cells and mice against ricin toxin without toxicity. This compound efficiently protects cells against various toxins and pathogens including viruses, intracellular bacteria and parasite. ABMA provokes Rab7-positive late endosomal compartment accumulation in mammalian cells without affecting other organelles (early endosomes, lysosomes, the Golgi apparatus, the endoplasmic reticulum or the nucleus). As the mechanism of action of ABMA is restricted to host-endosomal compartments, it reduces cell infection by pathogens that depend on this pathway to invade cells. ABMA may represent a novel class of broad-spectrum compounds with therapeutic potential against diverse severe infectious diseases.
Collapse
|