1
|
Alli VJ, Singh SK, Darna M, Suresh V, Sule SA, Jangam A, Kattula B, Pusarapu SL, Thomas J, Sardana Y, Gundla KP, Burra AG, Chandra Sekhar K, Patnaik SS, Reddi B, Muralidharan K, Bokara KK, Addlagatta A, Jadav SS. Development of acylhydrazone linked thiazoles as non-covalent dual inhibitors of SARS-CoV-2 proteases. Eur J Med Chem 2025; 290:117509. [PMID: 40132498 DOI: 10.1016/j.ejmech.2025.117509] [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: 11/20/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
SARS-CoV-2's papain-like protease (PLPro) and main protease (MPro) are essential for viral maturation and replication. Currently, Paxlovid is recommended to treat viral infections, but the emergence of Nirmatrelvir resistance new variants poses serious global risks. Dual targeting agents restrict viral replication, act on other crucial viral pathways, or exert simultaneous protease inhibition, increasing the complexity for the virus to develop resistance, and the design of dual inhibitors is an attractive strategy. Herein, we present research on new thiazole-aryl and thiazole-ester compounds that function as cysteine specific non-covalent competitive dual inhibitors of SARS-CoV-2's papain-like protease (PLPro) and main protease (MPro). Twelve of the 36 compounds demonstrated dual inhibition in the range of nanomolar to low micromolar concentrations while five others exhibit selective PLPro inhibition. Minimal cytotoxicity against two mammalian cell lines and no oral toxicity in rats (LD50 > 2000 mg/kg) were observed. SARS-CoV-2 viral load was successfully reduced by several compounds tested. N-acyl hydrazone (NAH)-thiazole core while forms π-π interactions with the catalytic histidine side chain (H41) in MPro active site, it exhibits a series of hydrophobic and hydrophilic interaction in the interface of BL2 loop and the active site of PLPro. Non-covalent dual inhibition demonstrated by novel NAH-thiazole derivatives in this study provides a path for the development of efficient antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Vidya Jyothi Alli
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Shubham Kumar Singh
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Mounika Darna
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Vavilapalli Suresh
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Swapnil Anil Sule
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Aruna Jangam
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bhavita Kattula
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Sarva Lakshmi Pusarapu
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Jessie Thomas
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Yogesh Sardana
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Krishna Prasad Gundla
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Amarender Goud Burra
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kunta Chandra Sekhar
- Department of Organic Synthesis &Process Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Samata Sai Patnaik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Bharati Reddi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kathirvel Muralidharan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Kiran Kumar Bokara
- CSIR-Centre for Cellular and Molecular Biology, ANNEXE II, Medical Biotechnology Complex, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Anthony Addlagatta
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| | - Surender Singh Jadav
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| |
Collapse
|
2
|
Yadav RP, Jena NR. Paritaprevir as a pan-antiviral against different flaviviruses. Front Mol Biosci 2025; 12:1524951. [PMID: 40248436 PMCID: PMC12003128 DOI: 10.3389/fmolb.2025.1524951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction The flavivirus infections caused by the Zika virus (ZIKV), Dengue virus (DENV), and West Nile virus (WNV) cause mild to serious pathological conditions, such as fever, joint pain, shock, internal bleeding, organ failure, nausea, breathlessness, brain tissue damage, neurodegenerative diseases, and deaths. As currently no efficient vaccine or drug is available to prevent or treat these diseases in humans, it is essential to identify potential drug-like molecules to treat these diseases. For these reasons, several known anti-viral drugs are repurposed against the proteases of ZIKV, WNV, and DENV to inhibit their activities. Methods The GOLD 5.0 molecular docking program was used to dock 20 HIV and HCV drugs against the ZIKV protease. Based on docking scores, 5 drugs were found to bind to the ZIKV protease with high affinities. Subsequently, the AMBER ff14SB force field was employed to simulate these drug-bound complexes of ZIKV protease. The MM/PBSA free energy method was utilized to compute the binding free energies of these complexes. Consequently, the two best ZIKV protease inhibitors were repurposed against the proteases of DENV and WNV. Results and Discussion It is found that out of the 5 drugs, Ritonavir and Paritaprevir bind to the NS2B-NS3 protease of the ZIKV strongly with the Gibbs binding free energies (∆Gbind) of -17.44±3.18 kcal/mol and -14.25±3.11 kcal/mol respectively. Remarkably, Ritonavir binds to the ZIKV Protease about 12 kcal/mol more strongly compared to its binding to the HIV protease. It is further found that Paritaprevir binds to DENV and WNV proteases as strongly as it binds to the ZIKV protease. Hence it is proposed that Paritaprevir may act as a potent pan-antiviral against the Zika, West Nile, and Dengue viral diseases.
Collapse
Affiliation(s)
| | - N. R. Jena
- Discipline of Natural Sciences, Indian Institute of Information Technology, Design and Manufacturing, Jabalpur, India
| |
Collapse
|
3
|
Rezende WS, Neto AM, Corbi JJ, Corbi PP, de Paiva REF, Bergamini FRG. Coordination Compounds as Antivirals against Neglected Tropical Diseases. ChemMedChem 2025; 20:e202400799. [PMID: 39591549 DOI: 10.1002/cmdc.202400799] [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: 10/08/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 11/28/2024]
Abstract
Neglected tropical viral diseases are a burden to social and economic welfare being responsible for higher pathogen-related mortality rates and chronic debilitating patient conditions. Climatic changes have widened up the infectibility ratio of such diseases, with autochthonous transmission in formerly temperate-to-cold environments. The slow-paced development of potential vaccines followed by the inexistence of antiviral drugs for such diseases considerably worsens the situation. Coordination compounds are a class of molecules that have been extensively explored as antiviral drugs for viruses such as poliovirus, HIV and, more recently, SARS-CoV-2, figuring as potential molecules to be explored and capitalized as antivirals against neglected viral strains. In this review the current efforts from the inorganic medicinal chemistry to address viral neglected tropical diseases, with emphasis to coordination compounds, is presented. Since many of neglected viruses are also arthropod-borne viruses, relying on a vector for transmission, coordination entities able to mitigate vectors are also presented as a parallel strategy to prevent and control such diseases.
Collapse
Affiliation(s)
- Wallace S Rezende
- Laboratory of Synthesis of Bioinspired Molecules, Institute of Chemistry, Federal University of Uberlândia-UFU, João Naves de Avila Avenue, 2121, 38408-100, Uberlândia, Minas Gerais, Brazil
| | - Antonio Marçal Neto
- Laboratory of Synthesis of Bioinspired Molecules, Institute of Chemistry, Federal University of Uberlândia-UFU, João Naves de Avila Avenue, 2121, 38408-100, Uberlândia, Minas Gerais, Brazil
| | - Juliano J Corbi
- Department of Hydraulics and Sanitation, University of São Paulo-USP, 13566-590, São Carlos, São Paulo, Brazil
| | - Pedro P Corbi
- Institute of Chemistry, University of Campinas-UNICAMP, PO Box 6154, Campinas, São Paulo, 13083-970, Brazil
| | - Raphael E F de Paiva
- Donostia International Physics Center-DIPC, Paseo Manuel de Lardizabal, 4 Donostia, Euskadi, Gipuzkoa, 20018, Spain
| | - Fernando R G Bergamini
- Laboratory of Synthesis of Bioinspired Molecules, Institute of Chemistry, Federal University of Uberlândia-UFU, João Naves de Avila Avenue, 2121, 38408-100, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
4
|
Martina MG, Rubini D, Radi M, Cagno V. Targeting PI4KB and Src/Abl host kinases as broad-spectrum antiviral strategy: Myth or real opportunity? Antiviral Res 2025; 235:106100. [PMID: 39922541 DOI: 10.1016/j.antiviral.2025.106100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Viruses pose a continuous threat to human health. Limited treatment options exist for current viruses, and the risk of infections with newly emerging or re-emerging viruses is increasing. In a pandemic scenario, having a broad-spectrum antiviral to limit viral spread while developing specific antivirals and vaccines is crucial. Targeting host kinases represents a valuable strategy due to the higher barrier to resistance and the broad-spectrum activity it offers. While cells have redundant kinases for the same biological function, viruses rely on specific kinases for their replication cycle, enabling targeted antiviral action with limited toxicity. This review focuses on two extensively studied kinase targets: the lipid kinase phosphatidylinositol 4-kinase IIIβ (PI4KB) and the tyrosine kinase proteins Src and Abl. Compounds active against these targets are reviewed in terms of the viruses they inhibit, their mechanisms of action and their stage of development. While PI4KB inhibitors have reached clinical trials, those targeting Src and Abl remain largely in the preclinical phase. Nevertheless, opportunities exist to improve potency and further understand the specific roles of these kinases in the life cycle of multiple viruses.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Daniele Rubini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy.
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
5
|
Wang F, Han H, Wang C, Wang J, Peng Y, Chen Y, He Y, Deng Z, Li F, Rong Y, Wang D, Liu W, Chen H, Zhang Z. SARS-CoV-2 membrane protein induces neurodegeneration via affecting Golgi-mitochondria interaction. Transl Neurodegener 2024; 13:68. [PMID: 39726060 DOI: 10.1186/s40035-024-00458-1] [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: 05/25/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Neurological complications are a significant concern of Coronavirus Disease 2019 (COVID-19). However, the pathogenic mechanism of neurological symptoms associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is poorly understood. METHODS We used Drosophila as a model to systematically analyze SARS-CoV-2 genes encoding structural and accessory proteins and identified the membrane protein (M) that disrupted mitochondrial functions in vivo. The M protein was stereotaxically injected to further assess its effects in the brains of wild-type (WT) and 5 × FAD mice. Omics technologies, including RNA sequencing and interactome analysis, were performed to explore the mechanisms of the effects of M protein both in vitro and in vivo. RESULTS Systematic analysis of SARS-CoV-2 structural and accessory proteins in Drosophila identified that the M protein induces mitochondrial fragmentation and dysfunction, leading to reduced ATP production, ROS overproduction, and eventually cell death in the indirect flight muscles. In WT mice, M caused hippocampal atrophy, neural apoptosis, glial activation, and mitochondrial damage. These changes were further aggravated in 5 × FAD mice. M was localized to the Golgi apparatus and genetically interacted with four wheel drive (FWD, a Drosophila homolog of mammalian PI4KIIIβ) to regulate Golgi functions in flies. Fwd RNAi, but not PI4KIIIα RNAi, reversed the M-induced Golgi abnormality, mitochondrial fragmentation, and ATP reduction. Inhibition of PI4KIIIβ activity suppressed the M-induced neuronal cell death. Therefore, M induced mitochondrial fragmentation and apoptosis likely through disruption of Golgi-derived PI(4)P-containing vesicles. CONCLUSIONS M disturbs the distribution and function of Golgi, leading to mitochondrial abnormality and eventually neurodegeneration via a PI4KIIIβ-mediated mechanism. This study reveals a potential mechanism for COVID-19 neurological symptoms and opens a new avenue for development of therapeutic strategies targeting SARS-CoV-2 M or mitochondria.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Hailong Han
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Caifang Wang
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Jingfei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Yanni Peng
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Ye Chen
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Yaohui He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361000, China
| | - Zhouyang Deng
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Fang Li
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Yikang Rong
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
| | - Danling Wang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
| | - Wen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361000, China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Zhuohua Zhang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China.
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
6
|
Han H, Liu H, Steiner R, Zhao Z, Jin ZG. Inhibition of protein kinase D and its substrate phosphatidylinositol-4 kinase III beta blocks common human coronavirus replication. Microbiol Spectr 2024; 12:e0150124. [PMID: 39540754 PMCID: PMC11619529 DOI: 10.1128/spectrum.01501-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the infection of a coronavirus, named as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coronaviruses can be replicated in the infected host cells. Coronavirus replication involves various steps, including membrane fusion, peri-nuclear particle formation, and matrix vesicle transport to the cell membrane via the endoplasmic reticulum-Golgi-lysosome route. Recent studies have suggested that protein kinase D (PKD) plays a crucial role in regulation of vesicle formation and trafficking in the trans-Golgi network (TGN). Thus, we hypothesize that inhibiting PKD and its associated pathway could be an effective strategy to limit viral replication. Here, we report that molecular and pharmacological inhibition of PKD and its substrate phosphatidylinositol-4 kinase III beta (PI4KIIIβ) significantly diminishes the replication of common human coronaviruses. Specifically, we found that the PKD-silencing siRNA and the PKD inhibitor CRT0066101 have broad-spectrum antiviral activity against HCoV-OC43, HCoV-NL63, and HCoV-229E in cultured cells. Mechanistic studies revealed that the deactivation of PKD reduced the activation of PI4KIIIβ, thereby blocking the transport of viral particles in the host cells. Furthermore, the PI4KIIIβ inhibitor, BQR695, also exhibited antiviral activity against those coronaviruses. In conclusion, PKD and its substrate, PI4KIIIβ, may serve as novel antiviral targets for human coronaviruses and warrant further investigation. IMPORTANCE Human coronaviruses can lead to a range of clinical symptoms, from asymptomatic infection to severe illness and death, with a limited array of antiviral drugs available. Protein kinase D (PKD) is involved in various cellular processes, such as cell proliferation, apoptosis, and membrane fission of the Golgi apparatus. However, the specific role of PKD in the human coronavirus life cycle remains unclear. In this study, we found that PKD inhibitors effectively attenuated human coronavirus replication at the trans-Golgi network (TGN) stage in the viral life cycle. Furthermore, inhibiting PKD reduced PI4KIIIβ activation, thereby blocking viral replication in the host cells. Importantly, PI4KIIIβ inhibitors also blocked human coronavirus replication. Thus, PKD may represent a promising therapeutic target against both current circulating and future emerging coronaviruses.
Collapse
Affiliation(s)
- Huijuan Han
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Zhijun Zhao
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zheng-Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
7
|
Martina MG, Carlen V, Van der Reysen S, Bianchi E, Cabella N, Crespan E, Radi M, Cagno V. Bithiazole inhibitors of PI4KB show broad-spectrum antiviral activity against different viral families. Antiviral Res 2024; 231:106003. [PMID: 39265657 DOI: 10.1016/j.antiviral.2024.106003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/29/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
Broad-spectrum antivirals can be extremely important for pandemic preparedness. Targeting host factors dispensable for the host but indispensable for the virus can result in high barrier to resistance and a large range of viruses targeted. PI4KB is a lipid kinase involved in the replication of several RNA viruses, but common inhibitors of this target are mainly active against members of the Picornaviridae family. Herein we describe the optimization of bithiazole PI4KB inhibitors as broad-spectrum antivirals (BSAs) active against different members of the Picornaviridae, Coronaviridae, Flaviviridae and Poxviridae families. Since some of these viruses are transmitted via respiratory route, the efficacy of one of the most promising compounds was evaluated in an airway model. The molecule showed complete viral inhibition and absence of toxicity. These results pave the road for the development of new BSAs.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | - Vincent Carlen
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland
| | - Sarah Van der Reysen
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | - Elena Bianchi
- Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Noemi Cabella
- Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Emmanuele Crespan
- Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Marco Radi
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy.
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
8
|
Barbieri F, Carlen V, Martina MG, Sannio F, Cancade S, Perini C, Restori M, Crespan E, Maga G, Docquier JD, Cagno V, Radi M. 4-Trifluoromethyl bithiazoles as broad-spectrum antimicrobial agents for virus-related bacterial infections or co-infections. RSC Med Chem 2024; 15:1589-1600. [PMID: 38784463 PMCID: PMC11110737 DOI: 10.1039/d3md00686g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/07/2024] [Indexed: 05/25/2024] Open
Abstract
Respiratory tract infections involving a variety of microorganisms such as viruses, bacteria, and fungi are a prominent cause of morbidity and mortality globally, exacerbating various pre-existing respiratory and non-respiratory conditions. Moreover, the ability of bacteria and viruses to coexist might impact the development and severity of lung infections, promoting bacterial colonization and subsequent disease exacerbation. Secondary bacterial infections following viral infections represent a complex challenge to be overcome from a therapeutic point of view. We report herein our efforts in the development of new bithiazole derivatives showing broad-spectrum antimicrobial activity against both viruses and bacteria. A series of 4-trifluoromethyl bithiazole analogues was synthesized and screened against selected viruses (hRVA16, EVD68, and ZIKV) and a panel of Gram-positive and Gram-negative bacteria. Among them, two promising broad-spectrum antimicrobial compounds (8a and 8j) have been identified: both compounds showed low micromolar activity against all tested viruses, 8a showed synergistic activity against E. coli and A. baumannii in the presence of a subinhibitory concentration of colistin, while 8j showed a broader spectrum of activity against Gram-positive and Gram-negative bacteria. Activity against antibiotic-resistant clinical isolates is also reported. Given the ever-increasing need to adequately address viral and bacterial infections or co-infections, this study paves the way for the development of new agents with broad antimicrobial properties and synergistic activity with common antivirals and antibacterials.
Collapse
Affiliation(s)
- Francesca Barbieri
- Dipartimento di Scienze degli Alimenti e del Farmaco (DipALIFAR), Università degli Studi di Parma Viale delle Scienze, 27/A 43124 Parma Italy
| | - Vincent Carlen
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne 1011 Lausanne Switzerland
| | - Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco (DipALIFAR), Università degli Studi di Parma Viale delle Scienze, 27/A 43124 Parma Italy
| | - Filomena Sannio
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena Viale Bracci 16 53100 Siena Italy
| | - Sacha Cancade
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena Viale Bracci 16 53100 Siena Italy
| | - Cecilia Perini
- Institute of Molecular Genetics IGM-CNR "Luigi Luca Cavalli-Sforza" Via Abbiategrasso 207 I-27100 Pavia Italy
| | - Margherita Restori
- Dipartimento di Scienze degli Alimenti e del Farmaco (DipALIFAR), Università degli Studi di Parma Viale delle Scienze, 27/A 43124 Parma Italy
| | - Emmanuele Crespan
- Institute of Molecular Genetics IGM-CNR "Luigi Luca Cavalli-Sforza" Via Abbiategrasso 207 I-27100 Pavia Italy
| | - Giovanni Maga
- Institute of Molecular Genetics IGM-CNR "Luigi Luca Cavalli-Sforza" Via Abbiategrasso 207 I-27100 Pavia Italy
| | - Jean-Denis Docquier
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena Viale Bracci 16 53100 Siena Italy
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne 1011 Lausanne Switzerland
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco (DipALIFAR), Università degli Studi di Parma Viale delle Scienze, 27/A 43124 Parma Italy
| |
Collapse
|
9
|
G Vishakantegowda A, Hwang D, Chakrasali P, Jung E, Lee JY, Shin JS, Jung YS. Highly potent and selective phosphatidylinositol 4-kinase IIIβ inhibitors as broad-spectrum anti-rhinoviral agents. RSC Med Chem 2024; 15:704-719. [PMID: 38389877 PMCID: PMC10880896 DOI: 10.1039/d3md00630a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 12/08/2023] [Indexed: 02/24/2024] Open
Abstract
Human rhinoviruses (hRVs) cause upper and lower respiratory tract infections and exacerbate asthma and chronic obstructive pulmonary disease. hRVs comprise more than 160 strains with considerable genetic variation. Their high diversity and strain-specific interactions with antisera hinder the development of anti-hRV therapeutic agents. Phosphatidylinositol-4-kinase IIIβ (PI4KIIIβ) is a key enzyme in the phosphoinositide signalling pathway that is crucial for the replication and survival of various viruses. We identified novel PI4KIIIβ inhibitors, N-(4-methyl-5-arylthiazol)-2-amide derivatives, by generating a hit compound, 1a, from the high-throughput screening of a chemical library, followed by the optimization study of 1a. Inhibitor 7e exhibited the highest activity (EC50 = 0.008, 0.0068, and 0.0076 μM for hRV-B14, hRV-A16, and hRV-A21, respectively) and high toxicity (CC50 = 6.1 μM). Inhibitor 7f showed good activity and low toxicity and provided the highest selectivity index (SI ≥ 4638, >3116, and >2793 for hRV-B14, hRV-A16, and hRV-A21, respectively). Furthermore, 7f showed broad-spectrum activities against various hRVs, coxsackieviruses, and other enteroviruses, such as EV-A71 and EV-D68. The binding mode of the inhibitors was investigated using 7f, and the experimental results of plaque reduction, replicon and cytotoxicity, and time-of-drug-addition assays suggested that 7f acts as a PI4KIIIβ inhibitor. The kinase inhibition activity of this series of compounds against PI4KIIIα and PI4KIIIβ was assessed, and 7f demonstrated kinase inhibition activity with an IC50 value of 0.016 μM for PI4KIIIβ, but not for PI4KIIIα (>10 μM). Therefore, 7f represents a highly potent and selective PI4KIIIβ inhibitor for the further development of antiviral therapy against hRVs or other enteroviruses.
Collapse
Affiliation(s)
- Avinash G Vishakantegowda
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
| | - Dasom Hwang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Laboratory of Veterinary Virology, College of Veterinary Medicine, Chungbuk National University Cheongju 28644 Republic of Korea
| | - Prashant Chakrasali
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Joo-Youn Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Young-Sik Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
| |
Collapse
|
10
|
Pacetti M, Pismataro MC, Felicetti T, Giammarino F, Bonomini A, Tiecco M, Bertagnin C, Barreca ML, Germani R, Cecchetti V, Vicenti I, Tabarrini O, Zazzi M, Loregian A, Massari S. Switching the three-component Biginelli-like reaction conditions for the regioselective synthesis of new 2-amino[1,2,4]triazolo[1,5- a]pyrimidines. Org Biomol Chem 2024; 22:767-783. [PMID: 38167738 DOI: 10.1039/d3ob01861j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Among the eight different triazolopyrimidine isomers existing in nature, 1,2,4-triazolo[1,5-a]pyrimidine (TZP) is one of the most studied and used isomers in medicinal chemistry. For some years, our group has been involved in developing regioselective one-pot procedures for the synthesis of 2-amino-7-aryl-5-methyl- and 2-amino-5-aryl-7-methyl-TZPs of interest in the preparation of antiviral agents. In this work, taking advantage of a Biginelli-like multicomponent reaction (MCR), we report the identification of finely tunable conditions to regioselectively synthesize C-6 ester-substituted amino-TZP analogues, both in dihydro and oxidized forms. Indeed, the use of mild acidic conditions is strongly directed toward the regioselective synthesis of 5-aryl-7-methyl C-6-substituted TZP analogues, while the use of neutral ionic liquids shifted the regioselectivity towards 7-aryl-5-methyl derivatives. In addition, the novel synthesized scaffolds were functionalized at the C-2 position and evaluated for their antiviral activity against RNA viruses (influenza virus, flaviviruses, and SARS-CoV-2). Compounds 25 and 26 emerged as promising anti-flavivirus agents, showing activity in the low micromolar range.
Collapse
Affiliation(s)
- Martina Pacetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | | | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | - Federica Giammarino
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Anna Bonomini
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Matteo Tiecco
- Chemistry Interdisciplinary Project (ChIP), School of Pharmacy, University of Camerino, 62032 Camerino, MC, Italy
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | | | - Raimondo Germani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | - Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| |
Collapse
|
11
|
Raymonda MH, Rodríguez-Sánchez I, Schafer XL, Smorodintsev-Schiller L, Harris IS, Munger J. Cytomegalovirus-induced inactivation of TSC2 disrupts the coupling of fatty acid biosynthesis to glucose availability resulting in a vulnerability to glucose starvation. mBio 2024; 15:e0303123. [PMID: 38117060 PMCID: PMC10790783 DOI: 10.1128/mbio.03031-23] [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: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
IMPORTANCE Viruses modulate host cell metabolism to support the mass production of viral progeny. For human cytomegalovirus, we find that the viral UL38 protein is critical for driving these pro-viral metabolic changes. However, our results indicate that these changes come at a cost, as UL38 induces an anabolic rigidity that leads to a metabolic vulnerability. We find that UL38 decouples the link between glucose availability and fatty acid biosynthetic activity. Normal cells respond to glucose limitation by down-regulating fatty acid biosynthesis. Expression of UL38 results in the inability to modulate fatty acid biosynthesis in response to glucose limitation, which results in cell death. We find this vulnerability in the context of viral infection, but this linkage between fatty acid biosynthesis, glucose availability, and cell death could have broader implications in other contexts or pathologies that rely on glycolytic remodeling, for example, oncogenesis.
Collapse
Affiliation(s)
- Matthew H. Raymonda
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Irene Rodríguez-Sánchez
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Xenia L. Schafer
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Leonid Smorodintsev-Schiller
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, USA
| | - Isaac S. Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, USA
| | - Joshua Munger
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
12
|
Aggarwal R, Jain N, Dubey GP, Singh S, Chandra R. Visible Light-Prompted Regioselective Synthesis of Novel 5-Aroyl/hetaroyl-2',4-dimethyl-2,4'-bithiazoles as DNA- and BSA-Targeting Agents. Biomacromolecules 2023; 24:4798-4818. [PMID: 37729507 DOI: 10.1021/acs.biomac.3c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Organic transformations mediated by visible light have gained popularity in recent years as they are green, renewable, inexpensive, and clean and yield excellent products. The present study describes cyclo-condensation of 2-methylthiazole-4-carbothioamide with differently substituted α-bromo-1,3-diketones achieved by utilizing a white light-emitting diode (LED) (9W) to accomplish the regioselective synthesis of novel 5-aroyl/hetaroyl-2',4-dimethyl-2,4'-bithiazole derivatives as DNA/bovine serum albumin (BSA)-targeting agents. The structure characterization of the exact regioisomer was achieved unequivocally by heteronuclear two-dimensional nuclear magnetic resonance (2D-NMR) spectroscopy [1H-13C] HMBC; [1H-13C] HMQC; and [1H-15N] HMBC. In silico toxicity studies indicated that the synthesized compounds exhibit low toxicity risks and adhere to the rules of oral bioavailability without any exception. Computational molecular modeling of the bithiazole derivatives with the dodecamer sequence of the DNA duplex and BSA identified 5-(4-chlorobenzoyl)-2',4-dimethyl-2,4'-bithiazole 7g as the most suitable derivative that can interact effectively with these biomolecules. Furthermore, theoretical results concurred with the ex vivo binding mode of the 7g with calf thymus DNA (ct-DNA) and BSA through a variety of spectroscopic techniques, viz., ultraviolet-visible (UV-visible), circular dichroism (CD), steady-state fluorescence, and competitive displacement assay, along with viscosity measurements.
Collapse
Affiliation(s)
- Ranjana Aggarwal
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India
- CSIR-National Institute of Science Communication and Policy Research, New Delhi 110012, India
| | - Naman Jain
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India
| | - Gyan Prakash Dubey
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, New Delhi 110007, India
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi, New Delhi 110007, India
| |
Collapse
|
13
|
Grazia Martina M, Giammarino F, Vicenti I, Groaz E, Rozenski J, Incerti M, Sannio F, Docquier JD, Zazzi M, Radi M. Nucleoside Derivatives of 2,6-Diaminopurine Antivirals: Base-Modified Nucleosides with Broad-Spectrum Antimicrobial Properties. ChemMedChem 2023; 18:e202300200. [PMID: 37221137 DOI: 10.1002/cmdc.202300200] [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: 04/12/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 05/25/2023]
Abstract
The plethora of viral outbreaks experienced in the last decade, together with the widespread distribution of many re-emerging and newly emerging viruses, emphasize the urgent need for novel broad-spectrum antivirals as tools for early intervention in case of future epidemics. Non-natural nucleosides have been at the forefront for the treatment of infectious diseases for many years and still represent one of the most successful classes of antiviral molecules on the market. In the attempt to explore the biologically relevant chemical space of this class of antimicrobials, we describe herein the development of novel base-modified nucleosides by converting previously identified 2,6-diaminopurine antivirals into the corresponding D/L ribonucleosides, acyclic nucleosides and prodrug derivatives. A phenotypic screening against viruses belonging to different families (Flaviviridae, Coronaviridae, Retroviridae) and against a panel of Gram-positive and Gram-negative bacteria, allowed to identify a few interesting molecules with broad-spectrum antimicrobial activities.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze, 27/A, 43124, Parma, Italy
| | - Federica Giammarino
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Ilaria Vicenti
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Elisabetta Groaz
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Jef Rozenski
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium
| | - Matteo Incerti
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze, 27/A, 43124, Parma, Italy
| | - Filomena Sannio
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Jean Denis Docquier
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, Viale Bracci 16, 53100, Siena, Italy
- Laboratoire de Bactériologie Moléculaire, Centre d'Ingénierie des Protéines, University of Liège, Allée du 6 Août, 4000, Liège, Belgium
| | - Maurizio Zazzi
- Dipartimento di Biotecnologie Mediche, Università degli Studi di Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze, 27/A, 43124, Parma, Italy
| |
Collapse
|
14
|
Raymonda MH, Rodríguez-Sánchez I, Schafer XL, Smorodintsev-Schiller L, Harris IS, Munger J. Cytomegalovirus-induced inactivation of TSC2 disrupts the coupling of fatty acid biosynthesis to glucose availability resulting in a vulnerability to glucose limitation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541212. [PMID: 37292722 PMCID: PMC10245705 DOI: 10.1101/2023.05.17.541212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Human cytomegalovirus (HCMV) modulates cellular metabolism to support productive infection, and the HCMV UL38 protein drives many aspects of this HCMV-induced metabolic program. However, it remains to be determined whether virally-induced metabolic alterations might induce novel therapeutic vulnerabilities in virally infected cells. Here, we explore how HCMV infection and the UL38 protein modulate cellular metabolism and how these changes alter the response to nutrient limitation. We find that expression of UL38, either in the context of HCMV infection or in isolation, sensitizes cells to glucose limitation resulting in cell death. This sensitivity is mediated through UL38's inactivation of the TSC complex subunit 2 (TSC2) protein, a central metabolic regulator that possesses tumor-suppressive properties. Further, expression of UL38 or the inactivation of TSC2 results in anabolic rigidity in that the resulting increased levels of fatty acid biosynthesis are insensitive to glucose limitation. This failure to regulate fatty acid biosynthesis in response to glucose availability sensitizes cells to glucose limitation, resulting in cell death unless fatty acid biosynthesis is inhibited. These experiments identify a regulatory circuit between glycolysis and fatty acid biosynthesis that is critical for cell survival upon glucose limitation and highlight a metabolic vulnerability associated with viral infection and the inactivation of normal metabolic regulatory controls.
Collapse
Affiliation(s)
- Matthew H. Raymonda
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Irene Rodríguez-Sánchez
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Xenia L. Schafer
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Leonid Smorodintsev-Schiller
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, USA
| | - Isaac S. Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, USA
| | - Joshua Munger
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
15
|
Privileged Scaffold Decoration for the Identification of the First Trisubstituted Triazine with Anti-SARS-CoV-2 Activity. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248829. [PMID: 36557962 PMCID: PMC9782877 DOI: 10.3390/molecules27248829] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022]
Abstract
Current therapy against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) are based on the use of Remdesivir 1, Molnupiravir 2, and the recently identified Nirmatrelvir 3. Unfortunately, these three drugs showed some limitations regarding potency and possible drug-drug interactions. A series of derivatives coming from a decoration approach of the privileged scaffold s-triazines were synthesized and evaluated against SAR-CoV-2. One derivative emerged as the hit of the series for its micromolar antiviral activity and low cytotoxicity. Mode of action and pharmacokinetic in vitro preliminary studies further confirm the role as candidates for a future optimization campaign of the most active derivative identified with this work.
Collapse
|
16
|
El-Lateef HMA, El-Dabea T, Khalaf MM, Abu-Dief AM. Development of Metal Complexes for Treatment of Coronaviruses. Int J Mol Sci 2022; 23:6418. [PMID: 35742870 PMCID: PMC9223400 DOI: 10.3390/ijms23126418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/29/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Coronavirus disease (SARS-CoV-2) is a global epidemic. This pandemic, which has been linked to high rates of death, has forced some countries throughout the world to implement complete lockdowns in order to contain the spread of infection. Because of the advent of new coronavirus variants, it is critical to find effective treatments and vaccines to prevent the virus's rapid spread over the world. In this regard, metal complexes have attained immense interest as antibody modifiers and antiviral therapies, and they have a lot of promise towards SARS-CoV-2 and their suggested mechanisms of action are discussed, i.e., a new series of metal complexes' medicinal vital role in treatment of specific proteins or SARS-CoV-2 are described. The structures of the obtained metal complexes were fully elucidated by different analytical and spectroscopic techniques also. Molecular docking and pharmacophore studies presented that most of complexes studied influenced good binding affinity to the main protease SARS-CoV-2, which also was attained as from the RCSB pdb (Protein Data Bank) data PDB ID: 6 W41, to expect the action of metal complexes in contradiction of COVID-19. Experimental research is required to determine the pharmacokinetics of most of the complexes analyzed for the treatment of SARS-CoV-2-related disease. Finally, the toxicity of a metal-containing inorganic complex will thus be discussed by its capability to transfer metals which may bind with targeted site.
Collapse
Affiliation(s)
- Hany M. Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Tarek El-Dabea
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Mai M. Khalaf
- Department of Chemistry, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
| | - Ahmed M. Abu-Dief
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82534, Egypt;
- Chemistry Department, College of Science, Taibah University, Madinah 344, Saudi Arabia
| |
Collapse
|
17
|
Martina MG, Sannio F, Crespan E, Pavone M, Simoncini A, Barbieri F, Perini C, Pesce E, Maga G, Pedemonte N, Docquier JD, Radi M. Towards Innovative Antibacterial-Correctors for Cystic Fibrosis Targeting the Lung Microbiome with a Multifunctional Effect. ChemMedChem 2022; 17:e202200277. [PMID: 35638249 DOI: 10.1002/cmdc.202200277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/30/2022] [Indexed: 11/10/2022]
Abstract
Cystic Fibrosis (CF) is a genetic disease caused by loss-of-function mutations in the CFTR gene, which codes for a defective ion channel. This causes an electrolyte imbalance and results in a spiral of negative effects on multiple organs, most notably the accumulation of thick mucus in the lungs, chronic respiratory tract infections and inflammation leading to pulmonary exacerbation and premature death. Progressive decline of lung function is mainly linked to persistent or recurring infections, mostly caused by bacteria, which require treatments with antibiotics and represent one of the major life-limiting factors in subjects with CF. Treatment of such a complex disease require multiple drugs with a consequent therapeutic burden and complications caused by drug-drug interactions and rapid emergence of bacterial drug resistance. We report herein our recent efforts in developing innovative multifunctional antibiotics specifically tailored to CF by a direct action on bacterial topoisomerases and a potential indirect effect on the pulmonary mucociliary clearance mediated by ΔF508-CFTR correction. The obtained results may pave the way for the development of a simplified therapeutic approach with a single agent acting as multifunctional antibacterial-corrector.
Collapse
Affiliation(s)
- Maria Grazia Martina
- University of Parma: Universita degli Studi di Parma, Department of Food and Drug, ITALY
| | - Filomena Sannio
- University of Siena: Universita degli Studi di Siena, Dipartimento di Biotecnologie Mediche, ITALY
| | - Emmanuele Crespan
- CNR: Consiglio Nazionale delle Ricerche, Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", ITALY
| | - Marialaura Pavone
- University of Parma: Universita degli Studi di Parma, Department of Food and Drug, ITALY
| | - Alice Simoncini
- University of Parma: Universita degli Studi di Parma, Department of Food and Drug, ITALY
| | - Francesca Barbieri
- University of Parma: Universita degli Studi di Parma, Department of Food and Drug, ITALY
| | - Cecilia Perini
- CNR: Consiglio Nazionale delle Ricerche, Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", ITALY
| | - Emanuela Pesce
- Istituto Giannina Gaslini Istituto Pediatrico di Ricovero e Cura a Carattere Scientifico: Istituto Giannina Gaslini, U.O.C. Genetica Medica, ITALY
| | - Giovanni Maga
- CNR: Consiglio Nazionale delle Ricerche, Istituto di Genetica Molecolare, IGM-CNR "Luigi Luca Cavalli-Sforza", ITALY
| | - Nicoletta Pedemonte
- Istituto Giannina Gaslini Istituto Pediatrico di Ricovero e Cura a Carattere Scientifico: Istituto Giannina Gaslini, U.O.C. Genetica Medica, ITALY
| | - Jean-Denis Docquier
- University of Siena: Universita degli Studi di Siena, Dipartimento di Biotecnologie Mediche, ITALY
| | - Marco Radi
- University of Parma, Department of Food and Drug, Viale delle Scienze, 27/A, 43124, Parma, ITALY
| |
Collapse
|
18
|
In Vitro Anti-SARS-CoV-2 Activity of Selected Metal Compounds and Potential Molecular Basis for Their Actions Based on Computational Study. Biomolecules 2021; 11:biom11121858. [PMID: 34944502 PMCID: PMC8699537 DOI: 10.3390/biom11121858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Metal-based drugs represent a rich source of chemical substances of potential interest for the treatment of COVID-19. To this end, we have developed a small but representative panel of nine metal compounds, including both synthesized and commercially available complexes, suitable for medical application and tested them in vitro against the SARS-CoV-2 virus. The screening revealed that three compounds from the panel, i.e., the organogold(III) compound Aubipyc, the ruthenium(III) complex KP1019, and antimony trichloride (SbCl3), are endowed with notable antiviral properties and an acceptable cytotoxicity profile. These initial findings prompted us to perform a computational study to unveil the likely molecular basis of their antiviral actions. Calculations evidenced that the metalation of nucleophile sites in SARS-CoV-2 proteins or nucleobase strands, induced by Aubipyc, SbCl3, and KP1019, is likely to occur. Remarkably, we found that only the deprotonated forms of Cys and Sec residues can react favorably with these metallodrugs. The mechanistic implications of these findings are discussed.
Collapse
|