1
|
van Niekerk AA, Maluck S, Mag P, Kővágó C, Kerek Á, Jerzsele Á, Steinmetzer T, Pászti-Gere E. Antiviral Drug Candidate Repositioning for Streptococcus suis Infection in Non-Tumorigenic Cell Models. Biomedicines 2024; 12:783. [PMID: 38672139 PMCID: PMC11048155 DOI: 10.3390/biomedicines12040783] [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: 02/28/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
The increasing prevalence of antimicrobial resistance against zoonotic bacteria, including Streptococcus (S.) suis, highlights the need for new therapeutical strategies, including the repositioning of drugs. In this study, susceptibilities of bacterial isolates were tested toward ten different 3-amidinophenyalanine (Phe(3-Am)) derivatives via determination of minimum inhibitory concentration (MIC) values. Some of these protease inhibitors, like compounds MI-432, MI-471, and MI-476, showed excellent antibacterial effects against S. suis. Their drug interaction potential was investigated using human liver microsomal cytochrome P450 (CYP450) measurements. In our work, non-tumorigenic IPEC-J2 cells and primary porcine hepatocytes were infected with S. suis, and the putative beneficial impact of these inhibitors was investigated on cell viability (Neutral red assay), on interleukin (IL)-6 levels (ELISA technique), and on redox balance (Amplex red method). The antibacterial inhibitors prevented S. suis-induced cell death (except MI-432) and decreased proinflammatory IL-6 levels. It was also found that MI-432 and MI-476 had antioxidant effects in an intestinal cell model upon S. suis infection. Concentration-dependent suppression of CYP3A4 function was found via application of all three inhibitors. In conclusion, our study suggests that the potential antiviral Phe(3-Am) derivatives with 2',4' dichloro-biphenyl moieties can be considered as effective drug candidates against S. suis infection due to their antibacterial effects.
Collapse
Affiliation(s)
- Ashley Anzet van Niekerk
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| | - Sara Maluck
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| | - Patrik Mag
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Csaba Kővágó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| | - Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Torsten Steinmetzer
- Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary (Á.J.)
| |
Collapse
|
2
|
Kristensen C, Larsen LE, Trebbien R, Jensen HE. The avian influenza A virus receptor SA-α2,3-Gal is expressed in the porcine nasal mucosa sustaining the pig as a mixing vessel for new influenza viruses. Virus Res 2024; 340:199304. [PMID: 38142890 PMCID: PMC10793167 DOI: 10.1016/j.virusres.2023.199304] [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: 08/12/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Influenza A viruses (IAVs) originate from wild birds but have on several occasions jumped host barriers and are now also circulating in humans and mammals. The IAV host receptors (glycans with galactose linked to a sialic acid (SA) in an α2,3 or α2,6 linkage) are crucial host factors restricting inter-species transmission. In general, avian-origin IAVs show a preference for SA-α2,3 (avian receptor), whereas IAVs isolated from humans and pigs prefer SA-α2,6 (human receptor). N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) are the two major SAs. Neu5Ac is expressed in all species, whereas Neu5Gc is only expressed in a limited number of domestic species such as pigs and horses, but not in humans. Despite that previous studies have shown that the IAV host receptor distribution appears to be similar in pigs and humans, none of these studies have investigated the expression of Neu5Gc-α2,6 in situ in porcine tissues. Thus, the aim of this study was to elucidate the distribution of IAV host receptors expressed in the porcine respiratory tract and relate the expression to the viral tropism of diverse host-adapted IAVs. The IAV receptor (SA-α2,3 and SA-α2,6) distribution and the presence of specifically Neu5Gc-α2,6 in the porcine nasal, tracheal, and lung tissues was investigated by lectin histochemistry. Furthermore, IAV immunohistochemistry was performed on tissues from pigs experimentally infected with IAVs, either adapted to pigs or humans, to investigate the significance of the IAV host receptors and the tropism of the diverse host-adapted IAVs. We document for the first time the expression of the avian receptor on the surface of the porcine nasal mucosa and an equal expression of Neu5Ac-α2,6 and Neu5Gc-α2,6 on the surface of the tracheal epithelium and alveoli. In all IAV-infected pigs, we found a low amount of IAV-positive cells in the trachea despite a high expression of the human receptor. Cumulatively, these findings suggest that optimal IAV replication involves a complex interplay between the viruses and their host receptors and that there might be other less clearly defined host factors that determine the site of replication.
Collapse
Affiliation(s)
- Charlotte Kristensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark.
| | - Lars E Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Ramona Trebbien
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen S, Denmark
| | - Henrik E Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
3
|
Colombo É, Désilets A, Hassanzadeh M, Lemieux G, Marois I, Cliche D, Delbrouck JA, Murza A, Jean F, Marsault E, Richter MV, Leduc R, Boudreault PL. Optimization of Ketobenzothiazole-Based Type II Transmembrane Serine Protease Inhibitors to Block H1N1 Influenza Virus Replication. ChemMedChem 2024; 19:e202300458. [PMID: 37864572 DOI: 10.1002/cmdc.202300458] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Human influenza viruses cause acute respiratory symptoms that can lead to death. Due to the emergence of antiviral drug-resistant strains, there is an urgent requirement for novel antiviral agents and innovative therapeutic strategies. Using the peptidomimetic ketobenzothiazole protease inhibitor RQAR-Kbt (IN-1, aka N-0100) as a starting point, we report how substituting P2 and P4 positions with natural and unnatural amino acids can modulate the inhibition potency toward matriptase, a prototypical type II transmembrane serine protease (TTSP) that acts as a priming protease for influenza viruses. We also introduced modifications of the peptidomimetics N-terminal groups, leading to significant improvements (from μM to nM, 60 times more potent than IN-1) in their ability to inhibit the replication of influenza H1N1 virus in the Calu-3 cell line derived from human lungs. The selectivity towards other proteases has been evaluated and explained using molecular modeling with a crystal structure recently obtained by our group. By targeting host cell TTSPs as a therapeutic approach, it may be possible to overcome the high mutational rate of influenza viruses and consequently prevent potential drug resistance.
Collapse
Affiliation(s)
- Éloïc Colombo
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Antoine Désilets
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Malihe Hassanzadeh
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Gabriel Lemieux
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Isabelle Marois
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
- Current address: Department of Biology, Faculty of Sciences, Université de Sherbrooke, Sherbrooke, J1K 2R1 Québec, Canada
| | - Dominic Cliche
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
| | - Julien A Delbrouck
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
- Current address: Xenon Pharmaceuticals Inc., Burnaby, V5G 4W8, British Columbia, Canada
| | - Alexandre Murza
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - François Jean
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, University of British Columbia, V6T 1Z3, British Columbia, Canada
| | - Eric Marsault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Martin V Richter
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, and Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| |
Collapse
|
4
|
van Eijk N, Schmacke LC, Steinmetzer T, Pilgram O, Poór M, Pászti-Gere E. In vitro testing of host-targeting small molecule antiviral matriptase/TMPRSS2 inhibitors in 2D and 3D cell-based assays. Biomed Pharmacother 2023; 168:115761. [PMID: 37865989 DOI: 10.1016/j.biopha.2023.115761] [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: 07/28/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) pandemic strongly stimulated the development of small molecule antivirals selectively targeting type II transmembrane serine proteases (TTSP), required for the host-cell entry of numerous viruses. A set of 3-amidinophenylalanine derivatives (MI-21, MI-472, MI-477, MI-485, MI-1903 and MI-1904), which inhibit the cleavage of certain viral glycoproteins was characterized in 2D and 3D primary human hepatocyte models on collagen- and Matrigel-coating using a CCK-8 assay to evaluate their cytotoxicity, a resorufin-based method to detect redox imbalances, fluorescence and ultrafiltration experiments to evaluate their interactions with human serum albumin (HSA) and α-acidic glycoprotein (AGP), and luminescence measurement to assess CYP3A4 modulation. For elucidation of selectivity of the applied compounds towards matriptase, transmembrane serine protease 2 (TMPRRS2), thrombin and factor Xa (FXa) Ki values were determined. It was proven that cell viability was only deteriorated by inhibitor MI-1903, and redox status was not influenced by administration of the selected inhibitors at 50 µM for 24 h. MI-472 and MI-477 formed relatively stable complexes with AGP. CYP3A4 inhibition was found to be strong in PHHs exposed to all inhibitors with the exception of MI-21, which seems to be a promising drug candidate also due to its better selectivity towards matriptase and TMPRSS2 over the blood clotting proteases thrombin and FXa. Our in vitro pharmacokinetic screening with these inhibitors helps to select the compounds with the best selectivity and safety profile suitable for a further preclinical characterization without animal sacrifice.
Collapse
Affiliation(s)
- Nicholas van Eijk
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Luna C Schmacke
- Institute of Pharmaceutical Chemistry, Department of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Department of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Oliver Pilgram
- Institute of Pharmaceutical Chemistry, Department of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs H-7624, Hungary; Lab-on-a-Chip Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary
| | - Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary.
| |
Collapse
|
5
|
Bauer L, Benavides FFW, Veldhuis Kroeze EJB, de Wit E, van Riel D. The neuropathogenesis of highly pathogenic avian influenza H5Nx viruses in mammalian species including humans. Trends Neurosci 2023; 46:953-970. [PMID: 37684136 PMCID: PMC10591965 DOI: 10.1016/j.tins.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023]
Abstract
Circulation of highly pathogenic avian influenza (HPAI) H5Nx viruses of the A/Goose/Guangdong/1/96 lineage in birds regularly causes infections of mammals, including humans. In many mammalian species, infections are associated with severe neurological disease, a unique feature of HPAI H5Nx viruses compared with other influenza A viruses. Here, we provide an overview of the neuropathogenesis of HPAI H5Nx virus infection in mammals, centered on three aspects: neuroinvasion, neurotropism, and neurovirulence. We focus on in vitro studies, as well as studies on naturally or experimentally infected mammals. Additionally, we discuss the contribution of viral factors to the neuropathogenesis of HPAI H5Nx virus infections and the efficacy of intervention strategies to prevent neuroinvasion or the development of neurological disease.
Collapse
Affiliation(s)
- Lisa Bauer
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Emmie de Wit
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Debby van Riel
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
6
|
AbuBakar U, Amrani L, Kamarulzaman FA, Karsani SA, Hassandarvish P, Khairat JE. Avian Influenza Virus Tropism in Humans. Viruses 2023; 15:833. [PMID: 37112812 PMCID: PMC10142937 DOI: 10.3390/v15040833] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
An influenza pandemic happens when a novel influenza A virus is able to infect and transmit efficiently to a new, distinct host species. Although the exact timing of pandemics is uncertain, it is known that both viral and host factors play a role in their emergence. Species-specific interactions between the virus and the host cell determine the virus tropism, including binding and entering cells, replicating the viral RNA genome within the host cell nucleus, assembling, maturing and releasing the virus to neighboring cells, tissues or organs before transmitting it between individuals. The influenza A virus has a vast and antigenically varied reservoir. In wild aquatic birds, the infection is typically asymptomatic. Avian influenza virus (AIV) can cross into new species, and occasionally it can acquire the ability to transmit from human to human. A pandemic might occur if a new influenza virus acquires enough adaptive mutations to maintain transmission between people. This review highlights the key determinants AIV must achieve to initiate a human pandemic and describes how AIV mutates to establish tropism and stable human adaptation. Understanding the tropism of AIV may be crucial in preventing virus transmission in humans and may help the design of vaccines, antivirals and therapeutic agents against the virus.
Collapse
Affiliation(s)
- Umarqayum AbuBakar
- Institute of Biological Sciences (ISB), Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Lina Amrani
- Institute of Biological Sciences (ISB), Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Farah Ayuni Kamarulzaman
- Institute of Biological Sciences (ISB), Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Saiful Anuar Karsani
- Institute of Biological Sciences (ISB), Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Pouya Hassandarvish
- Tropical Infectious Diseases Research and Education Center, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Jasmine Elanie Khairat
- Institute of Biological Sciences (ISB), Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
7
|
Abstract
The continuous emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) urges better understanding of the functional motifs in the spike (S) protein and their tolerance to mutations. Here, we focused on the S2′ motif, which, during virus entry, requires cleavage by a host cell protease to release the fusion peptide. Though belonging to an immunogenic region, the SARS-CoV-2 S2′ motif (811-KPSKR-815) has shown hardly any variation, with its three basic (K/R) residues being >99.99% conserved thus far. By creating a series of mutant pseudoviruses bearing the spikes of Wuhan-Hu-1, its G614 mutant or the Delta and Omicron variants, we show that residue K814 (preceding the scissile R815) is dispensable for TMPRSS2 yet favored by the alternative TMPRSS13 protease. Activation by TMPRSS13 was drastically reduced when the SARS-CoV-2 S2′ motif was swapped with that of the low pathogenic 229E coronavirus (685-RVAGR-689), and also, the reverse effect was seen. This swap had no impact on recognition by TMPRSS2. In the Middle East respiratory syndrome coronavirus (MERS-CoV) spike, introducing a dibasic scissile motif was easily accepted by TMPRSS13 but less so by TMPRSS2, confirming that TMPRSS13 favors a sequence rich in K/R residues. Pseudovirus entry experiments in Calu-3 cells confirmed that the S2′ mutations have minor impact on TMPRSS2. Our findings are the first to demonstrate which S2′ residues are important for SARS-CoV-2 spike activation by these two airway proteases, with TMPRSS2 being more tolerant to variation than TMPRSS13. This preemptive insight will help to estimate the impact of S2′ motif changes as they appear in new SARS-CoV-2 variants.
Collapse
|
8
|
Improving the selectivity of 3-amidinophenylalanine-derived matriptase inhibitors. Eur J Med Chem 2022; 238:114437. [DOI: 10.1016/j.ejmech.2022.114437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/19/2022]
|
9
|
de Bruin ACM, Funk M, Spronken MI, Gultyaev AP, Fouchier RAM, Richard M. Hemagglutinin Subtype Specificity and Mechanisms of Highly Pathogenic Avian Influenza Virus Genesis. Viruses 2022; 14:1566. [PMID: 35891546 PMCID: PMC9321182 DOI: 10.3390/v14071566] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Highly Pathogenic Avian Influenza Viruses (HPAIVs) arise from low pathogenic precursors following spillover from wild waterfowl into poultry populations. The main virulence determinant of HPAIVs is the presence of a multi-basic cleavage site (MBCS) in the hemagglutinin (HA) glycoprotein. The MBCS allows for HA cleavage and, consequently, activation by ubiquitous proteases, which results in systemic dissemination in terrestrial poultry. Since 1959, 51 independent MBCS acquisition events have been documented, virtually all in HA from the H5 and H7 subtypes. In the present article, data from natural LPAIV to HPAIV conversions and experimental in vitro and in vivo studies were reviewed in order to compile recent advances in understanding HA cleavage efficiency, protease usage, and MBCS acquisition mechanisms. Finally, recent hypotheses that might explain the unique predisposition of the H5 and H7 HA sequences to obtain an MBCS in nature are discussed.
Collapse
Affiliation(s)
- Anja C. M. de Bruin
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; (A.C.M.d.B.); (M.F.); (M.I.S.); (A.P.G.); (R.A.M.F.)
| | - Mathis Funk
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; (A.C.M.d.B.); (M.F.); (M.I.S.); (A.P.G.); (R.A.M.F.)
| | - Monique I. Spronken
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; (A.C.M.d.B.); (M.F.); (M.I.S.); (A.P.G.); (R.A.M.F.)
| | - Alexander P. Gultyaev
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; (A.C.M.d.B.); (M.F.); (M.I.S.); (A.P.G.); (R.A.M.F.)
- Group Imaging and Bioinformatics, Leiden Institute of Advanced Computer Science (LIACS), Leiden University, 2300 RA Leiden, The Netherlands
| | - Ron A. M. Fouchier
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; (A.C.M.d.B.); (M.F.); (M.I.S.); (A.P.G.); (R.A.M.F.)
| | - Mathilde Richard
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; (A.C.M.d.B.); (M.F.); (M.I.S.); (A.P.G.); (R.A.M.F.)
| |
Collapse
|
10
|
Gorący A, Rosik J, Szostak B, Ustianowski Ł, Ustianowska K, Gorący J. Human Cell Organelles in SARS-CoV-2 Infection: An Up-to-Date Overview. Viruses 2022; 14:v14051092. [PMID: 35632833 PMCID: PMC9144443 DOI: 10.3390/v14051092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
Since the end of 2019, the whole world has been struggling with the life-threatening pandemic amongst all age groups and geographic areas caused by Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). The Coronavirus Disease 2019 (COVID-19) pandemic, which has led to more than 468 million cases and over 6 million deaths reported worldwide (as of 20 March 2022), is one of the greatest threats to human health in history. Meanwhile, the lack of specific and irresistible treatment modalities provoked concentrated efforts in scientists around the world. Various mechanisms of cell entry and cellular dysfunction were initially proclaimed. Especially, mitochondria and cell membrane are crucial for the course of infection. The SARS-CoV-2 invasion depends on angiotensin converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2), and cluster of differentiation 147 (CD147), expressed on host cells. Moreover, in this narrative review, we aim to discuss other cell organelles targeted by SARS-CoV-2. Lastly, we briefly summarize the studies on various drugs.
Collapse
Affiliation(s)
- Anna Gorący
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (A.G.); (J.G.)
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Jakub Rosik
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (A.G.); (J.G.)
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Correspondence:
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
| | - Łukasz Ustianowski
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
| | - Klaudia Ustianowska
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
| | - Jarosław Gorący
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (A.G.); (J.G.)
| |
Collapse
|
11
|
Fedor Z, Szentkirályi-Tóth A, Nagy G, Szimrók Z, Varga E, Pászti A, Pászti Z, Jerzsele Á, Pilgram O, Steinmetzer T, Mátis G, Neogrády Z, Pászti-Gere E. Interspecies Comparisons of the Effects of Potential Antiviral 3-Amidinophenylalanine Derivatives on Cytochrome P450 1A2 Isoenzyme. Vet Sci 2022; 9:vetsci9040156. [PMID: 35448654 PMCID: PMC9027957 DOI: 10.3390/vetsci9040156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
In vitro models of animals vulnerable to SARS-CoV-2 infection can support the characterization of effective antiviral drugs, such as synthetic inhibitors of the transmembrane protease serine 2 (TMPRSS2). Changes in cytochrome P450 (CYP) 1A2 activities in the presence of the potential TMPRSS2/matriptase inhibitors (MI) were measured using fluorometric and luminescent assays. Furthermore, the cytotoxicity of these inhibitors was evaluated using the MTS method. In addition, 60 min-long microsomal stability assays were performed using an UPLC-MS/MS procedure to elucidate depletion rates of the inhibitors. CYP1A2 was influenced significantly by MI-463 and MI-1900 in rat microsomes, by MI-432 and MI-482 in beagle microsomes, and by MI-432, MI-463, MI-482, and MI-1900 in cynomolgus monkey microsomes. The IC50 values in monkey microsomes were 1.30 ± 0.14 µM, 2.4 ± 1.4 µM, 0.21 ± 0.09 µM, and 1.1 ± 0.8 µM for inhibitors MI-432, MI-463, MI-482, and MI-1900, respectively. The depletion rates of the parent compounds were lower than 50%, independently of the investigated animal species. The host cell factor TMPRSS2 is of key importance for the cross-species spread of SARS-CoV-2. Studies of the in vitro biotransformation of TMPRSS2 inhibitors provide additional information for the development of new antiviral drugs.
Collapse
Affiliation(s)
- Zsófia Fedor
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Anna Szentkirályi-Tóth
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Gábor Nagy
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Zoltán Szimrók
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Eszter Varga
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Anna Pászti
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Zoltán Pászti
- Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, 1117 Budapest, Hungary;
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
| | - Oliver Pilgram
- Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, 35037 Marburg, Germany; (O.P.); (T.S.)
| | - Torsten Steinmetzer
- Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, 35037 Marburg, Germany; (O.P.); (T.S.)
| | - Gábor Mátis
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, 1078 Budapest, Hungary; (G.M.); (Z.N.)
| | - Zsuzsanna Neogrády
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, 1078 Budapest, Hungary; (G.M.); (Z.N.)
| | - Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary; (Z.F.); (A.S.-T.); (G.N.); (Z.S.); (E.V.); (A.P.); (Á.J.)
- Correspondence:
| |
Collapse
|
12
|
Pászti-Gere E, Szentkirályi A, Fedor Z, Nagy G, Szimrók Z, Pászti Z, Pászti A, Pilgram O, Steinmetzer T, Bodnárová S, Fliszár-Nyúl E, Poór M. In vitro interaction of potential antiviral TMPRSS2 inhibitors with human serum albumin and cytochrome P 450 isoenzymes. Biomed Pharmacother 2022; 146:112513. [PMID: 34915414 PMCID: PMC8668183 DOI: 10.1016/j.biopha.2021.112513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/24/2021] [Accepted: 12/06/2021] [Indexed: 01/25/2023] Open
Abstract
The interactions of four sulfonylated Phe(3-Am)-derived inhibitors (MI-432, MI-463, MI-482 and MI-1900) of type II transmembrane serine proteases (TTSP) such as transmembrane protease serine 2 (TMPRSS2) were examined with serum albumin and cytochrome P450 (CYP) isoenzymes. Complex formation with albumin was investigated using fluorescence spectroscopy. Furthermore, microsomal hepatic CYP1A2, 2C9, 2C19 and 3A4 activities in presence of these inhibitors were determined using fluorometric assays. The inhibitory effects of these compounds on human recombinant CYP3A4 enzyme were also examined. In addition, microsomal stability assays (60-min long) were performed using an UPLC-MS/MS method to determine depletion percentage values of each compound. The inhibitors showed no or only weak interactions with albumin, and did not inhibit CYP1A2, 2C9 and 2C19. However, the compounds tested proved to be potent inhibitors of CYP3A4 in both assays performed. Within one hour, 20%, 12%, 14% and 25% of inhibitors MI-432, MI-463, MI-482 and MI-1900, respectively, were degraded. As essential host cell factor for the replication of the pandemic SARS-CoV-2, the TTSP TMPRSS2 emerged as an important target in drug design. Our study provides further preclinical data on the characterization of this type of inhibitors for numerous trypsin-like serine proteases.
Collapse
Affiliation(s)
- Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary.
| | - Anna Szentkirályi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Zsófia Fedor
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Gábor Nagy
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Zoltán Szimrók
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Zoltán Pászti
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar tudósok körútja 2, Budapest H-1117, Hungary
| | - Anna Pászti
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Oliver Pilgram
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Slávka Bodnárová
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs H-7624, Hungary,Lab-on-a-Chip Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs H-7624, Hungary,Lab-on-a-Chip Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs H-7624, Hungary; Lab-on-a-Chip Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary.
| |
Collapse
|
13
|
Wettstein L, Kirchhoff F, Münch J. The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. Int J Mol Sci 2022; 23:1351. [PMID: 35163273 PMCID: PMC8836196 DOI: 10.3390/ijms23031351] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 01/25/2023] Open
Abstract
TMPRSS2 is a type II transmembrane protease with broad expression in epithelial cells of the respiratory and gastrointestinal tract, the prostate, and other organs. Although the physiological role of TMPRSS2 remains largely elusive, several endogenous substrates have been identified. TMPRSS2 serves as a major cofactor in SARS-CoV-2 entry, and primes glycoproteins of other respiratory viruses as well. Consequently, inhibiting TMPRSS2 activity is a promising strategy to block viral infection. In this review, we provide an overview of the role of TMPRSS2 in the entry processes of different respiratory viruses. We then review the different classes of TMPRSS2 inhibitors and their clinical development, with a focus on COVID-19 treatment.
Collapse
Affiliation(s)
| | | | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (L.W.); (F.K.)
| |
Collapse
|
14
|
de Bruin ACM, Spronken MI, Bestebroer TM, Fouchier RAM, Richard M. Reduced Replication of Highly Pathogenic Avian Influenza Virus in Duck Endothelial Cells Compared to Chicken Endothelial Cells Is Associated with Stronger Antiviral Responses. Viruses 2022; 14:v14010165. [PMID: 35062369 PMCID: PMC8779112 DOI: 10.3390/v14010165] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) cause fatal systemic infections in chickens, which are associated with endotheliotropism. HPAIV infections in wild birds are generally milder and not endotheliotropic. Here, we aimed to elucidate the species-specific endotheliotropism of HPAIVs using primary chicken and duck aortic endothelial cells (chAEC and dAEC respectively). Viral replication kinetics and host responses were assessed in chAEC and dAEC upon inoculation with HPAIV H5N1 and compared to embryonic fibroblasts. Although dAEC were susceptible to HPAIV upon inoculation at high multiplicity of infection, HPAIV replicated to lower levels in dAEC than chAEC during multi-cycle replication. The susceptibility of duck embryonic endothelial cells to HPAIV was confirmed in embryos. Innate immune responses upon HPAIV inoculation differed between chAEC, dAEC, and embryonic fibroblasts. Expression of the pro-inflammatory cytokine IL8 increased in chicken cells but decreased in dAEC. Contrastingly, the induction of antiviral responses was stronger in dAEC than in chAEC, and chicken and duck fibroblasts. Taken together, these data demonstrate that although duck endothelial cells are permissive to HPAIV infection, they display markedly different innate immune responses than chAEC and embryonic fibroblasts. These differences may contribute to the species-dependent differences in endotheliotropism and consequently HPAIV pathogenesis.
Collapse
|
15
|
Wang D, Li C, Chiu MC, Yu Y, Liu X, Zhao X, Huang J, Cheng Z, Yuan S, Poon V, Cai J, Chu H, Chan JF, To KK, Yuen KY, Zhou J. SPINK6 inhibits human airway serine proteases and restricts influenza virus activation. EMBO Mol Med 2022; 14:e14485. [PMID: 34826211 PMCID: PMC9976594 DOI: 10.15252/emmm.202114485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 11/09/2022] Open
Abstract
SPINK6 was identified in human skin as a cellular inhibitor of serine proteases of the KLK family. Airway serine proteases are required to cleave hemagglutinin (HA) of influenza A viruses (IAVs) to initiate an infection in the human airway. We hypothesized that SPINK6 may inhibit common airway serine proteases and restrict IAV activation. We demonstrate that SPINK6 specifically suppresses the proteolytic activity of HAT and KLK5, HAT- and KLK5-mediated HA cleavage, and restricts virus maturation and replication. SPINK6 constrains the activation of progeny virions and impairs viral growth; and vice versa, blocking endogenous SPINK6 enhances HA cleavage and viral growth in physiological-relevant human airway organoids where SPINK6 is intrinsically expressed. In IAV-infected mice, SPINK6 significantly suppresses viral growth and improves mouse survival. Notably, individuals carrying the higher SPINK6 expression allele were protected from human H7N9 infection. Collectively, SPINK6 is a novel host inhibitor of serine proteases in the human airway and restricts IAV activation.
Collapse
Affiliation(s)
- Dong Wang
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Cun Li
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Man Chun Chiu
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Yifei Yu
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Xiaojuan Liu
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Xiaoyu Zhao
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Jingjing Huang
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Zhongshan Cheng
- Applied Bioinformatics CenterSt Jude Children’s Research HospitalMemphisTNUSA
| | - Shuofeng Yuan
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Vincent Poon
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Jian‐Piao Cai
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Hin Chu
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina,State Key Laboratory of Emerging Infectious DiseasesThe University of Hong KongHong KongChina
| | - Jasper Fuk‐Woo Chan
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina,State Key Laboratory of Emerging Infectious DiseasesThe University of Hong KongHong KongChina,Carol Yu Centre for InfectionThe University of Hong KongHong KongChina
| | - Kelvin Kai‐Wang To
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina,State Key Laboratory of Emerging Infectious DiseasesThe University of Hong KongHong KongChina,Carol Yu Centre for InfectionThe University of Hong KongHong KongChina
| | - Kwok Yung Yuen
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina,State Key Laboratory of Emerging Infectious DiseasesThe University of Hong KongHong KongChina,Carol Yu Centre for InfectionThe University of Hong KongHong KongChina
| | - Jie Zhou
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina,State Key Laboratory of Emerging Infectious DiseasesThe University of Hong KongHong KongChina
| |
Collapse
|
16
|
Wu J, Zhang L, Wang X. Host Sex Steroids Interact With Virus Infection: New Insights Into Sex Disparity in Infectious Diseases. Front Microbiol 2021; 12:747347. [PMID: 34803967 PMCID: PMC8600311 DOI: 10.3389/fmicb.2021.747347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/27/2021] [Indexed: 01/08/2023] Open
Abstract
Sex hormones are steroid hormones synthesized from the gonads of animals and tissues such as the placenta and adrenocortical reticular zone. The physiological functions of sex hormones are complex. Sex hormones are not only pathologically correlated with many diseases of the reproductive system, but are etiological factors in some viral infectious diseases, including disease caused by infections of coronaviruses, herpesviruses, hepatitis viruses, and other kinds of human viruses, which either exhibit a male propensity in clinical practice, or crosstalk with androgen receptor (AR)-related pathways in viral pathogenesis. Due to the global pandemic of coronavirus disease 2019 (COVID-19), the role of androgen/AR in viral infectious disease is highlighted again, majorly representing by the recent advances of AR-responsive gene of transmembrane protease/serine subfamily member 2 (TMPRSS2), which proteolytically activates the receptor-mediated virus entry by many coronaviruses and influenza virus, along with the role of androgen-mediated signaling for the transcription of hepatitis B virus (HBV), and the role of sex hormone responsive genes during Zika virus (ZIKV) pathogenesis, et al. Collectively, we propose to provide a comprehensive overview of the role of male sex hormones during multiple phases in the life cycle of different human viruses, which may be partly responsible for the sex-specific prevalence, severity and mortality of some diseases, therefore, may provide clues to develop more efficient prevention and treatment strategies for high-risk populations.
Collapse
Affiliation(s)
- Jinfeng Wu
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Lei Zhang
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xing Wang
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
17
|
Mahoney M, Damalanka VC, Tartell MA, Chung DH, Lourenço AL, Pwee D, Mayer Bridwell AE, Hoffmann M, Voss J, Karmakar P, Azouz NP, Klingler AM, Rothlauf PW, Thompson CE, Lee M, Klampfer L, Stallings CL, Rothenberg ME, Pöhlmann S, Whelan SPJ, O'Donoghue AJ, Craik CS, Janetka JW. A novel class of TMPRSS2 inhibitors potently block SARS-CoV-2 and MERS-CoV viral entry and protect human epithelial lung cells. Proc Natl Acad Sci U S A 2021; 118:e2108728118. [PMID: 34635581 PMCID: PMC8694051 DOI: 10.1073/pnas.2108728118] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2021] [Indexed: 12/14/2022] Open
Abstract
The host cell serine protease TMPRSS2 is an attractive therapeutic target for COVID-19 drug discovery. This protease activates the Spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and of other coronaviruses and is essential for viral spread in the lung. Utilizing rational structure-based drug design (SBDD) coupled to substrate specificity screening of TMPRSS2, we have discovered covalent small-molecule ketobenzothiazole (kbt) TMPRSS2 inhibitors which are structurally distinct from and have significantly improved activity over the existing known inhibitors Camostat and Nafamostat. Lead compound MM3122 (4) has an IC50 (half-maximal inhibitory concentration) of 340 pM against recombinant full-length TMPRSS2 protein, an EC50 (half-maximal effective concentration) of 430 pM in blocking host cell entry into Calu-3 human lung epithelial cells of a newly developed VSV-SARS-CoV-2 chimeric virus, and an EC50 of 74 nM in inhibiting cytopathic effects induced by SARS-CoV-2 virus in Calu-3 cells. Further, MM3122 blocks Middle East respiratory syndrome coronavirus (MERS-CoV) cell entry with an EC50 of 870 pM. MM3122 has excellent metabolic stability, safety, and pharmacokinetics in mice, with a half-life of 8.6 h in plasma and 7.5 h in lung tissue, making it suitable for in vivo efficacy evaluation and a promising drug candidate for COVID-19 treatment.
Collapse
Affiliation(s)
- Matthew Mahoney
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110
- ProteXase Therapeutics, Inc., Saint Louis, MO 63108
| | - Vishnu C Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110
| | - Michael A Tartell
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110
- Program in Virology, Harvard Medical School, Boston, MA 02115
| | - Dong Hee Chung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | - André Luiz Lourenço
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | - Dustin Pwee
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093
| | - Anne E Mayer Bridwell
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen 37077, Germany
- Faculty of Biology and Psychology, Georg-August University Göttingen, Göttingen 37077, Germany
| | - Jorine Voss
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110
| | - Partha Karmakar
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110
| | - Nurit P Azouz
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Andrea M Klingler
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Paul W Rothlauf
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110
- Program in Virology, Harvard Medical School, Boston, MA 02115
| | - Cassandra E Thompson
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110
| | - Melody Lee
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | | | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen 37077, Germany
- Faculty of Biology and Psychology, Georg-August University Göttingen, Göttingen 37077, Germany
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| | - James W Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110;
- ProteXase Therapeutics, Inc., Saint Louis, MO 63108
| |
Collapse
|
18
|
Sato K, Hayashi H, Shimotai Y, Yamaya M, Hongo S, Kawakami K, Matsuzaki Y, Nishimura H. TMPRSS2 Activates Hemagglutinin-Esterase Glycoprotein of Influenza C Virus. J Virol 2021; 95:e0129621. [PMID: 34406864 PMCID: PMC8513465 DOI: 10.1128/jvi.01296-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023] Open
Abstract
Influenza C virus (ICV) has only one kind of spike protein, the hemagglutinin-esterase (HE) glycoprotein. HE functions similarly to hemagglutinin (HA) and neuraminidase of the influenza A and B viruses (IAV and IBV, respectively). It has a monobasic site, which is cleaved by some host enzymes. The cleavage is essential to activating the virus, but the enzyme or enzymes in the respiratory tract have not been identified. This study investigated whether the host serine proteases, transmembrane protease serine S1 member 2 (TMPRSS2) and human airway trypsin-like protease (HAT), which reportedly cleave HA of IAV/IBV, are involved in HE cleavage. We established TMPRSS2- and HAT-expressing MDCK cells (MDCK-TMPRSS2 and MDCK-HAT). ICV showed multicycle replication with HE cleavage without trypsin in MDCK-TMPRSS2 cells as well as IAV did. The HE cleavage and multicycle replication did not appear in MDCK-HAT cells infected with ICV without trypsin, while HA cleavage and multistep growth of IAV appeared in the cells. Amino acid sequences of the HE cleavage site in 352 ICV strains were completely preserved. Camostat and nafamostat suppressed the growth of ICV and IAV in human nasal surface epithelial (HNE) cells. Therefore, this study revealed that, at least, TMPRSS2 is involved in HE cleavage and suggested that nafamostat could be a candidate for therapeutic drugs for ICV infection. IMPORTANCE Influenza C virus (ICV) is a pathogen that causes acute respiratory illness, mostly in children, but there are no anti-ICV drugs. ICV has only one kind of spike protein, the hemagglutinin-esterase (HE) glycoprotein on the virion surface, which possesses receptor-binding, receptor-destroying, and membrane fusion activities. The HE cleavage is essential for the virus to be activated, but the enzyme or enzymes in the respiratory tract have not been identified. This study revealed that transmembrane protease serine S1 member 2 (TMPRSS2), and not human airway trypsin-like protease (HAT), is involved in HE cleavage. This is a novel study on the host enzymes involved in HE cleavage, and the result suggests that the host enzymes, such as TMPRSS2, may be a target for therapeutic drugs of ICV infection.
Collapse
Affiliation(s)
- Ko Sato
- Virus Research Center, Clinical Research Division, Sendai Medical Center, Sendai, Miyagi, Japan
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hideki Hayashi
- Medical University Research Administrator, Nagasaki University School of Medicine, Sakamoto, Nagasaki, Japan
| | - Yoshitaka Shimotai
- Department of Infectious Diseases, Yamagata University Faculty of Medicine, Iida-Nishi, Yamagata, Japan
| | - Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate school of Medicine, Sendai, Miyagi, Japan
| | - Seiji Hongo
- Department of Infectious Diseases, Yamagata University Faculty of Medicine, Iida-Nishi, Yamagata, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yoko Matsuzaki
- Department of Infectious Diseases, Yamagata University Faculty of Medicine, Iida-Nishi, Yamagata, Japan
| | - Hidekazu Nishimura
- Virus Research Center, Clinical Research Division, Sendai Medical Center, Sendai, Miyagi, Japan
| |
Collapse
|
19
|
Kucher AN, Babushkina NP, Sleptcov AA, Nazarenko MS. Genetic Control of Human Infection with SARS-CoV-2. RUSS J GENET+ 2021; 57:627-641. [PMID: 34248311 PMCID: PMC8254434 DOI: 10.1134/s1022795421050057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/26/2020] [Accepted: 08/25/2020] [Indexed: 01/21/2023]
Abstract
In 2019, the SARS-CoV-2 beta-coronavirus, which caused a pandemic of severe acute respiratory viral infection COVID-19 (from COronaVIrus Disease 2019), was first detected. The susceptibility to SARS-CoV-2 and the nature of the course of the COVID-19 clinical picture are determined by many factors, including genetic characteristics of both the pathogen and the human. The SARS-CoV-2 genome has a similarity to the genomes of other coronaviruses, which are pathogenic for humans and cause a severe course of infection: 79% to the SARS-CoV genome and 50% to the MERS-CoV genome. The most significant differences between SARS-CoV-2 and other coronaviruses are recorded in the structure of the gene of the S protein, a key protein responsible for the virus binding to the receptor of the host organism cells. In particular, substitutions in the S protein of SARS-CoV-2, leading to the formation of the furin cleavage site that is absent in other SARS-like coronaviruses, were identified, which may explain the high pathogenicity of SARS-CoV-2. In humans, the genes that are significant for the initial stages of infection include ACE2, ANPEP, DPP4 (encode receptors for coronavirus binding); TMPRSS2, FURIN, TMPRSS11D, CTSL, CTSB (encode proteases involved in the entry of the coronavirus into the cell); DDX1 (the gene of ATP-dependent RNA helicase DDX1, which promotes replication of coronaviruses); and IFITM1, IFITM2, and IFITM3 (encode interferon-induced transmembrane proteins with an antiviral effect). These genes are expressed in many tissues (including those susceptible to the effects of SARS-CoV-2); rare and frequent variants that affect the structure of the encoded protein and its properties and expression level are described in them. A number of common genetic variants with proven functional significance are characterized by the variability in the allele frequency in the world's populations, which can determine interpopulation differences in the prevalence of COVID-19 and in the clinical features of the course of this pathology. The expression level of genes that are important for the formation of the susceptibility to SARS-CoV-2 is affected by epigenetic modifications, comorbidities at the time of infection, taking medications, and bad habits.
Collapse
Affiliation(s)
- A. N. Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - N. P. Babushkina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - A. A. Sleptcov
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - M. S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| |
Collapse
|
20
|
Mahoney M, Damalanka VC, Tartell MA, Chung DH, Lourenco AL, Pwee D, Mayer Bridwell AE, Hoffmann M, Voss J, Karmakar P, Azouz N, Klingler AM, Rothlauf PW, Thompson CE, Lee M, Klampfer L, Stallings C, Rothenberg ME, Pöhlmann S, Whelan SP, O'Donoghue AJ, Craik CS, Janetka JW. A novel class of TMPRSS2 inhibitors potently block SARS-CoV-2 and MERS-CoV viral entry and protect human epithelial lung cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34131661 DOI: 10.1101/2021.05.06.442935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The host cell serine protease TMPRSS2 is an attractive therapeutic target for COVID-19 drug discovery. This protease activates the Spike protein of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and of other coronaviruses and is essential for viral spread in the lung. Utilizing rational structure-based drug design (SBDD) coupled to substrate specificity screening of TMPRSS2, we have discovered a novel class of small molecule ketobenzothiazole TMPRSS2 inhibitors with significantly improved activity over existing irreversible inhibitors Camostat and Nafamostat. Lead compound MM3122 ( 4 ) has an IC 50 of 340 pM against recombinant full-length TMPRSS2 protein, an EC 50 of 430 pM in blocking host cell entry into Calu-3 human lung epithelial cells of a newly developed VSV SARS-CoV-2 chimeric virus, and an EC 50 of 74 nM in inhibiting cytopathic effects induced by SARS-CoV-2 virus in Calu-3 cells. Further, MM3122 blocks Middle East Respiratory Syndrome Coronavirus (MERS-CoV) cell entry with an EC 50 of 870 pM. MM3122 has excellent metabolic stability, safety, and pharmacokinetics in mice with a half-life of 8.6 hours in plasma and 7.5 h in lung tissue, making it suitable for in vivo efficacy evaluation and a promising drug candidate for COVID-19 treatment.
Collapse
|
21
|
Russell CJ. Hemagglutinin Stability and Its Impact on Influenza A Virus Infectivity, Pathogenicity, and Transmissibility in Avians, Mice, Swine, Seals, Ferrets, and Humans. Viruses 2021; 13:746. [PMID: 33923198 PMCID: PMC8145662 DOI: 10.3390/v13050746] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Genetically diverse influenza A viruses (IAVs) circulate in wild aquatic birds. From this reservoir, IAVs sporadically cause outbreaks, epidemics, and pandemics in wild and domestic avians, wild land and sea mammals, horses, canines, felines, swine, humans, and other species. One molecular trait shown to modulate IAV host range is the stability of the hemagglutinin (HA) surface glycoprotein. The HA protein is the major antigen and during virus entry, this trimeric envelope glycoprotein binds sialic acid-containing receptors before being triggered by endosomal low pH to undergo irreversible structural changes that cause membrane fusion. The HA proteins from different IAV isolates can vary in the pH at which HA protein structural changes are triggered, the protein causes membrane fusion, or outside the cell the virion becomes inactivated. HA activation pH values generally range from pH 4.8 to 6.2. Human-adapted HA proteins tend to have relatively stable HA proteins activated at pH 5.5 or below. Here, studies are reviewed that report HA stability values and investigate the biological impact of variations in HA stability on replication, pathogenicity, and transmissibility in experimental animal models. Overall, a stabilized HA protein appears to be necessary for human pandemic potential and should be considered when assessing human pandemic risk.
Collapse
Affiliation(s)
- Charles J Russell
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA
| |
Collapse
|
22
|
Monticelli M, Hay Mele B, Benetti E, Fallerini C, Baldassarri M, Furini S, Frullanti E, Mari F, Andreotti G, Cubellis MV, Renieri A. Protective Role of a TMPRSS2 Variant on Severe COVID-19 Outcome in Young Males and Elderly Women. Genes (Basel) 2021; 12:596. [PMID: 33921689 PMCID: PMC8073081 DOI: 10.3390/genes12040596] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
The protease encoded by the TMPRSS2 gene facilitates viral infections and has been implicated in the pathogenesis of SARS-CoV-2. We analyzed the TMPRSS2 sequence and correlated the protein variants with the clinical features of a cohort of 1177 patients affected by COVID-19 in Italy. Nine relatively common variants (allele frequency > 0.01) and six missense variants which may affect the protease activity according to PolyPhen-2 in HumVar-trained mode were identified. Among them, p.V197M (p.Val197Met) (rs12329760) emerges as a common variant that has a deleterious effect on the protease and a protective effect on the patients. Its role appears particularly relevant in two subgroups of patients-young males and elderly women-and among those affected by co-morbidities, where the variant frequency is higher among individuals who were mildly affected by the disease and did not need hospitalization or oxygen therapy than among those more severely affected, who required oxygen therapy, ventilation or intubation. This study provides useful information for the identification of patients at risk of developing a severe form of COVID-19, and encourages the usage of drugs affecting the expression of TMPRSS2 or inhibiting protein activity.
Collapse
Affiliation(s)
- Maria Monticelli
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
| | - Bruno Hay Mele
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
- Integrative Marine Ecology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Mari
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | | | | | - Maria Vittoria Cubellis
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
- Istituto di Chimica Biomolecolare—CNR, 80078 Pozzuoli, Italy
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
23
|
Gultyaev AP, Spronken MI, Funk M, Fouchier RAM, Richard M. Insertions of codons encoding basic amino acids in H7 hemagglutinins of influenza A viruses occur by recombination with RNA at hotspots near snoRNA binding sites. RNA (NEW YORK, N.Y.) 2021; 27:123-132. [PMID: 33188057 PMCID: PMC7812872 DOI: 10.1261/rna.077495.120] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/06/2020] [Indexed: 06/11/2023]
Abstract
The presence of multiple basic amino acids in the protease cleavage site of the hemagglutinin (HA) protein is the main molecular determinant of virulence of highly pathogenic avian influenza (HPAI) viruses. Recombination of HA RNA with other RNA molecules of host or virus origin is a dominant mechanism of multibasic cleavage site (MBCS) acquisition for H7 subtype HA. Using alignments of HA RNA sequences from documented cases of MBCS insertion due to recombination, we show that such recombination with host RNAs is most likely to occur at particular hotspots in ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), and viral RNAs. The locations of these hotspots in highly abundant RNAs indicate that RNA recombination is facilitated by the binding of small nucleolar RNA (snoRNA) near the recombination points.
Collapse
MESH Headings
- Amino Acids, Basic/genetics
- Amino Acids, Basic/metabolism
- Animals
- Base Pairing
- Base Sequence
- Chickens/virology
- Codon
- Gene Expression Regulation
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Host-Pathogen Interactions/genetics
- Humans
- Influenza A virus/genetics
- Influenza A virus/metabolism
- Influenza A virus/pathogenicity
- Influenza in Birds/virology
- Influenza, Human/virology
- Mutagenesis, Insertional
- RNA, Small Nucleolar/chemistry
- RNA, Small Nucleolar/genetics
- RNA, Small Nucleolar/metabolism
- RNA, Viral/chemistry
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Recombination, Genetic
- Sequence Alignment
- Virulence
Collapse
Affiliation(s)
- Alexander P Gultyaev
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
- Group Imaging and Bioinformatics, Leiden Institute of Advanced Computer Science (LIACS), Leiden University, 2300 RA Leiden, the Netherlands
| | - Monique I Spronken
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Mathis Funk
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Mathilde Richard
- Department of Viroscience, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| |
Collapse
|
24
|
Chepur SV, Pluzhnikov NN, Chubar OV, Bakulina LS, Litvinenko IV, Makarov VA, Gogolevsky AS, Myasnikov VA, Myasnikova IA, Al-Shehadat RI. Respiratory RNA Viruses: How to Be Prepared for an Encounter with New Pandemic Virus Strains. BIOLOGY BULLETIN REVIEWS 2021; 11. [PMCID: PMC8078390 DOI: 10.1134/s207908642102002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The characteristics of the biology of influenza viruses and coronavirus that determine the implementation of the infectious process are presented. With provision for pathogenesis of infection possible effects of serine proteinase inhibitors, heparin, and inhibitors of heparan sulfate receptors in the prevention of cell contamination by viruses are examined. It has been determined that chelators of metals of variable valency and antioxidants should be used for the reduction of replicative activity of viruses and anti-inflammatory therapy. The possibility of a pH-dependent impairment of glycosylation of cellular and viral proteins was traced for chloroquine and its derivatives. The use of low-toxicity drugs as part of adjunct therapy increases the effectiveness of synthetic antiviral drugs and interferons and ensures the safety of baseline therapy.
Collapse
Affiliation(s)
- S. V. Chepur
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - N. N. Pluzhnikov
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - O. V. Chubar
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - L. S. Bakulina
- Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | | | - V. A. Makarov
- Fundamentals of Biotechnology Federal Research Center, 119071 Moscow, Russia
| | - A. S. Gogolevsky
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - V. A. Myasnikov
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - I. A. Myasnikova
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - R. I. Al-Shehadat
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| |
Collapse
|
25
|
Abstract
Many enzymatic activity assays are based on either (1) identifying and quantifying the enzyme with methods such as western blot or enzyme-linked substrate assay (ELISA) or (2) quantifying the enzymatic reaction by monitoring the changing levels of either product or substrate. We have generated an outer membrane protein G (OmpG)-based nanopore approach to distinguish enzyme identity as well as analyze the enzyme's catalytic activity. Here, we engineered an OmpG nanopore with a peptide cut site inserted into one of its loops to detect proteolytic behavior. In addition, we generated an OmpG nanopore with a single-stranded DNA attached to a loop for analyzing nucleolytic cleavage. This OmpG nanopore approach may be highly useful in analyzing specific enzymes in complex biological samples, or in directly determining kinetics of enzyme-substrate complex association and dissociation.
Collapse
|
26
|
Iverson E, Kaler L, Agostino EL, Song D, Duncan GA, Scull MA. Leveraging 3D Model Systems to Understand Viral Interactions with the Respiratory Mucosa. Viruses 2020; 12:E1425. [PMID: 33322395 PMCID: PMC7763686 DOI: 10.3390/v12121425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory viruses remain a significant cause of morbidity and mortality in the human population, underscoring the importance of ongoing basic research into virus-host interactions. However, many critical aspects of infection are difficult, if not impossible, to probe using standard cell lines, 2D culture formats, or even animal models. In vitro systems such as airway epithelial cultures at air-liquid interface, organoids, or 'on-chip' technologies allow interrogation in human cells and recapitulate emergent properties of the airway epithelium-the primary target for respiratory virus infection. While some of these models have been used for over thirty years, ongoing advancements in both culture techniques and analytical tools continue to provide new opportunities to investigate airway epithelial biology and viral infection phenotypes in both normal and diseased host backgrounds. Here we review these models and their application to studying respiratory viruses. Furthermore, given the ability of these systems to recapitulate the extracellular microenvironment, we evaluate their potential to serve as a platform for studies specifically addressing viral interactions at the mucosal surface and detail techniques that can be employed to expand our understanding.
Collapse
Affiliation(s)
- Ethan Iverson
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Logan Kaler
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
| | - Eva L. Agostino
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Gregg A. Duncan
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Margaret A. Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| |
Collapse
|
27
|
Chaudhry F, Lavandero S, Xie X, Sabharwal B, Zheng YY, Correa A, Narula J, Levy P. Manipulation of ACE2 expression in COVID-19. Open Heart 2020; 7:e001424. [PMID: 33443121 PMCID: PMC7757413 DOI: 10.1136/openhrt-2020-001424] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is the virus responsible for the ongoing COVID-19 outbreak. The virus uses ACE2 receptor for viral entry. ACE2 is part of the counter-regulatory renin-angiotensin-aldosterone system and is also expressed in the lower respiratory tract along the alveolar epithelium. There is, however, significant controversy regarding the role of ACE2 expression in COVID-19 pathogenesis. Some have argued that decreasing ACE2 expression would result in decreased susceptibility to the virus by decreasing available binding sites for SARS-CoV-2 and restricting viral entry into the cells. Others have argued that, like the pathogenesis of other viral pneumonias, including those stemming from previous severe acute respiratory syndrome (SARS) viruses, once SARS-CoV-2 binds to ACE2, it downregulates ACE2 expression. Lack of the favourable effects of ACE2 might exaggerate lung injury by a variety of mechanisms. In order to help address this controversy, we conducted a literature search and review of relevant preclinical and clinical publications pertaining to SARS-CoV-2, COVID-19, ACE2, viral pneumonia, SARS, acute respiratory distress syndrome and lung injury. Our review suggests, although controversial, that patients at increased susceptibility to COVID-19 complications may have reduced baseline ACE2, and by modulating ACE2 expression one can possibly improve COVID-19 outcomes. Herein, we elucidate why and how this potential mechanism might work.
Collapse
Affiliation(s)
- Farhan Chaudhry
- Department of Emergency Medicine and Integrative Biosciences Center, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Basera Sabharwal
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Ashish Correa
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jagat Narula
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Phillip Levy
- Department of Emergency Medicine and Integrative Biosciences Center, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
28
|
Makarov V, Riabova O, Ekins S, Pluzhnikov N, Chepur S. The past, present and future of RNA respiratory viruses: influenza and coronaviruses. Pathog Dis 2020; 78:ftaa046. [PMID: 32860686 PMCID: PMC7499567 DOI: 10.1093/femspd/ftaa046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Influenza virus and coronaviruses continue to cause pandemics across the globe. We now have a greater understanding of their functions. Unfortunately, the number of drugs in our armory to defend us against them is inadequate. This may require us to think about what mechanisms to address. Here, we review the biological properties of these viruses, their genetic evolution and antiviral therapies that can be used or have been attempted. We will describe several classes of drugs such as serine protease inhibitors, heparin, heparan sulfate receptor inhibitors, chelating agents, immunomodulators and many others. We also briefly describe some of the drug repurposing efforts that have taken place in an effort to rapidly identify molecules to treat patients with COVID-19. While we put a heavy emphasis on the past and present efforts, we also provide some thoughts about what we need to do to prepare for respiratory viral threats in the future.
Collapse
Affiliation(s)
- Vadim Makarov
- Federal Research Center Fundamentals of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow 119071, Russia
| | - Olga Riabova
- Federal Research Center Fundamentals of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow 119071, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Nikolay Pluzhnikov
- State Research Institute of Military Medicine of the Ministry of Defence of the Russian Federation, St Petersburg 195043, Russia
| | - Sergei Chepur
- State Research Institute of Military Medicine of the Ministry of Defence of the Russian Federation, St Petersburg 195043, Russia
| |
Collapse
|
29
|
Abstract
The world is currently embroiled in a pandemic of coronavirus disease 2019 (COVID-19), a respiratory illness caused by the novel betacoronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The severity of COVID-19 disease ranges from asymptomatic to fatal acute respiratory distress syndrome. In few patients, the disease undergoes phenotypic differentiation between 7 and 14 days of acute illness, either resulting in full recovery or symptom escalation. However, the mechanism of such variation is not clear, but the facts suggest that patient's immune status, comorbidities, and the systemic effects of the viral infection (potentially depending on the SARS-CoV-2 strain involved) play a key role. Subsequently, patients with the most severe symptoms tend to have poor outcomes, manifest severe hypoxia, and possess elevated levels of pro-inflammatory cytokines (including IL-1β, IL-6, IFN-γ, and TNF-α) along with elevated levels of the anti-inflammatory cytokine IL-10, marked lymphopenia, and elevated neutrophil-to-lymphocyte ratios. Based on the available evidence, we propose a mechanism wherein SARS-CoV-2 infection induces direct organ damage while also fueling an IL-6-mediated cytokine release syndrome (CRS) and hypoxia, resulting in escalating systemic inflammation, multi-organ damage, and end-organ failure. Elevated IL-6 and hypoxia together predisposes patients to pulmonary hypertension, and the presence of asymptomatic hypoxia in COVID-19 further compounds this problem. Due to the similar downstream mediators, we discuss the potential synergistic effects and systemic ramifications of SARS-CoV-2 and influenza virus during co-infection, a phenomenon we have termed "COVI-Flu." Additionally, the differences between CRS and cytokine storm are highlighted. Finally, novel management approaches, clinical trials, and therapeutic strategies toward both SARS-CoV-2 and COVI-Flu infection are discussed, highlighting host response optimization and systemic inflammation reduction.
Collapse
|
30
|
Inhibition of TMPRSS2 by HAI-2 reduces prostate cancer cell invasion and metastasis. Oncogene 2020; 39:5950-5963. [PMID: 32778768 PMCID: PMC7416816 DOI: 10.1038/s41388-020-01413-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/13/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022]
Abstract
TMPRSS2 is an important membrane-anchored serine protease involved in human prostate cancer progression and metastasis. A serine protease physiologically often comes together with a cognate inhibitor for execution of proteolytically biologic function; however, TMPRSS2's cognate inhibitor is still elusive. To identify the cognate inhibitor of TMPRSS2, in this study, we applied co-immunoprecipitation and LC/MS/MS analysis and isolated hepatocyte growth factor activator inhibitors (HAIs) to be potential inhibitor candidates for TMPRSS2. Moreover, the recombinant HAI-2 proteins exhibited a better inhibitory effect on TMPRSS2 proteolytic activity than HAI-1, and recombinant HAI-2 proteins had a high affinity to form a complex with TMPRSS2. The immunofluorescence images further showed that TMPRSS2 was co-localized to HAI-2. Both KD1 and KD2 domain of HAI-2 showed comparable inhibitory effects on TMPRSS2 proteolytic activity. In addition, HAI-2 overexpression could suppress the induction effect of TMPRSS2 on pro-HGF activation, extracellular matrix degradation and prostate cancer cell invasion. We further determined that the expression levels of TMPRSS2 were inversely correlated with HAI-2 levels during prostate cancer progression. In orthotopic xenograft animal model, TMPRSS2 overexpression promoted prostate cancer metastasis, and HAI-2 overexpression efficiently blocked TMPRSS2-induced metastasis. In summary, the results together indicate that HAI-2 can function as a cognate inhibitor for TMPRSS2 in human prostate cancer cells and may serve as a potential factor to suppress TMPRSS2-mediated malignancy.
Collapse
|
31
|
Bestle D, Heindl MR, Limburg H, Van Lam van T, Pilgram O, Moulton H, Stein DA, Hardes K, Eickmann M, Dolnik O, Rohde C, Klenk HD, Garten W, Steinmetzer T, Böttcher-Friebertshäuser E. TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci Alliance 2020; 3:3/9/e202000786. [PMID: 32703818 PMCID: PMC7383062 DOI: 10.26508/lsa.202000786] [Citation(s) in RCA: 550] [Impact Index Per Article: 137.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 11/24/2022] Open
Abstract
The novel emerged SARS-CoV-2 has rapidly spread around the world causing acute infection of the respiratory tract (COVID-19) that can result in severe disease and lethality. For SARS-CoV-2 to enter cells, its surface glycoprotein spike (S) must be cleaved at two different sites by host cell proteases, which therefore represent potential drug targets. In the present study, we show that S can be cleaved by the proprotein convertase furin at the S1/S2 site and the transmembrane serine protease 2 (TMPRSS2) at the S2' site. We demonstrate that TMPRSS2 is essential for activation of SARS-CoV-2 S in Calu-3 human airway epithelial cells through antisense-mediated knockdown of TMPRSS2 expression. Furthermore, SARS-CoV-2 replication was also strongly inhibited by the synthetic furin inhibitor MI-1851 in human airway cells. In contrast, inhibition of endosomal cathepsins by E64d did not affect virus replication. Combining various TMPRSS2 inhibitors with furin inhibitor MI-1851 produced more potent antiviral activity against SARS-CoV-2 than an equimolar amount of any single serine protease inhibitor. Therefore, this approach has considerable therapeutic potential for treatment of COVID-19.
Collapse
Affiliation(s)
- Dorothea Bestle
- Institute of Virology, Philipps-University, Marburg, Germany
| | | | - Hannah Limburg
- Institute of Virology, Philipps-University, Marburg, Germany
| | - Thuy Van Lam van
- Institute of Pharmaceutical Chemistry, Philipps-University, Marburg, Germany
| | - Oliver Pilgram
- Institute of Pharmaceutical Chemistry, Philipps-University, Marburg, Germany
| | - Hong Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - David A Stein
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Kornelia Hardes
- Institute of Pharmaceutical Chemistry, Philipps-University, Marburg, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology, Gießen, Germany
| | - Markus Eickmann
- Institute of Virology, Philipps-University, Marburg, Germany.,German Center for Infection Research (DZIF), Marburg-Gießen-Langen Site, Emerging Infections Unit, Philipps-University, Marburg, Germany
| | - Olga Dolnik
- Institute of Virology, Philipps-University, Marburg, Germany.,German Center for Infection Research (DZIF), Marburg-Gießen-Langen Site, Emerging Infections Unit, Philipps-University, Marburg, Germany
| | - Cornelius Rohde
- Institute of Virology, Philipps-University, Marburg, Germany.,German Center for Infection Research (DZIF), Marburg-Gießen-Langen Site, Emerging Infections Unit, Philipps-University, Marburg, Germany
| | | | - Wolfgang Garten
- Institute of Virology, Philipps-University, Marburg, Germany
| | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Philipps-University, Marburg, Germany
| | | |
Collapse
|
32
|
Sallenave JM, Guillot L. Innate Immune Signaling and Proteolytic Pathways in the Resolution or Exacerbation of SARS-CoV-2 in Covid-19: Key Therapeutic Targets? Front Immunol 2020; 11:1229. [PMID: 32574272 PMCID: PMC7270404 DOI: 10.3389/fimmu.2020.01229] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
COVID-19 is caused by the Severe Acute Respiratory Syndrome (SARS) coronavirus (Cov)-2, an enveloped virus with a positive-polarity, single-stranded RNA genome. The initial outbreak of the pandemic began in December 2019, and it is affecting the human health of the global community. In common with previous pandemics (Influenza H1N1 and SARS-CoV) and the epidemics of Middle east respiratory syndrome (MERS)-CoV, CoVs target bronchial and alveolar epithelial cells. Virus protein ligands (e.g., haemagglutinin or trimeric spike glycoprotein for Influenza and CoV, respectively) interact with cellular receptors, such as (depending on the virus) either sialic acids, Dipeptidyl peptidase 4 (DPP4), or angiotensin-converting enzyme 2 (ACE2). Host proteases, e.g., cathepsins, furin, or members of the type II transmembrane serine proteases (TTSP) family, such as Transmembrane protease serine 2 (TMPRSS2), are involved in virus entry by proteolytically activating virus ligands. Also involved are Toll Like Receptor (TLR) family members, which upregulate anti-viral and pro-inflammatory mediators [interleukin (IL)-6 and IL-8 and type I and type III Interferons among others], through the activation of Nuclear Factor (NF)-kB. When these events (virus cellular entry and innate immune responses) are uncontrolled, a deleterious systemic response is sometimes encountered in infected patients, leading to the well-described "cytokine storm" and an ensuing multiple organ failure promoted by a downregulation of dendritic cell, macrophage, and T-cell function. We aim to describe how the lung and systemic host innate immune responses affect survival either positively, through downregulating initial viral load, or negatively, by triggering uncontrolled inflammation. An emphasis will be put on host cellular signaling pathways and proteases involved with a view on tackling these therapeutically.
Collapse
Affiliation(s)
- Jean-Michel Sallenave
- INSERM UMR1152, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Hôpital Bichat, Université de Paris, Paris, France
| | - Loïc Guillot
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
33
|
Lambertz RLO, Gerhauser I, Nehlmeier I, Gärtner S, Winkler M, Leist SR, Kollmus H, Pöhlmann S, Schughart K. H2 influenza A virus is not pathogenic in Tmprss2 knock-out mice. Virol J 2020; 17:56. [PMID: 32321537 PMCID: PMC7178614 DOI: 10.1186/s12985-020-01323-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/08/2020] [Indexed: 12/29/2022] Open
Abstract
The host cell protease TMPRSS2 cleaves the influenza A virus (IAV) hemagglutinin (HA). Several reports have described resistance of Tmprss2−/− knock-out (KO) mice to IAV infection but IAV of the H2 subtype have not been examined yet. Here, we demonstrate that TMPRSS2 is able to cleave H2-HA in cell culture and that Tmprss2−/− mice are resistant to infection with a re-assorted PR8_HA(H2) virus. Infection of KO mice did not cause major body weight loss or death. Furthermore, no significant increase in lung weights and no virus replication were observed in Tmprss2−/− mice. Finally, only minor tissue damage and infiltration of immune cells were detected and no virus-positive cells were found in histological sections of Tmprss2−/− mice. In summary, our studies indicate that TMPRSS2 is required for H2 IAV spread and pathogenesis in mice. These findings extend previous results pointing towards a central role of TMPRSS2 in IAV infection and validate host proteases as a potential target for antiviral therapy.
Collapse
Affiliation(s)
- Ruth Lydia Olga Lambertz
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sabine Gärtner
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Michael Winkler
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sarah Rebecca Leist
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Current Address: Department of Epidemiology, University of North Carolina, Chapel Hill, USA
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany.,Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany. .,University of Veterinary Medicine Hannover, Hannover, Germany. .,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
34
|
Beerens N, Heutink R, Harders F, Bossers A, Koch G, Peeters B. Emergence and Selection of a Highly Pathogenic Avian Influenza H7N3 Virus. J Virol 2020; 94:e01818-19. [PMID: 31969434 PMCID: PMC7108855 DOI: 10.1128/jvi.01818-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/09/2020] [Indexed: 01/21/2023] Open
Abstract
Low-pathogenicity avian influenza (LPAI) viruses of subtypes H5 and H7 have the ability to spontaneously mutate to highly pathogenic (HPAI) virus variants, causing high mortality in poultry. The highly pathogenic phenotype is caused by mutation of the hemagglutinin (HA) cleavage site, but additional mutations may play a role. Evidence from the field for the switch to high pathogenicity remains scarce. This study provides direct evidence for LPAI-to-HPAI virus mutation during H7N3 infection of a turkey farm in the Netherlands. No severe clinical symptoms were reported at the farm, but deep sequencing of isolates from the infected turkeys revealed a minority of HPAI virus sequences (0.06%) in the virus population. The HPAI virus contained a 12-nucleotide insertion in the HA cleavage site that was likely introduced by a single event as no intermediates with shorter inserts were identified. This suggests nonhomologous recombination as the mechanism of insertion. Analysis of different organs of the infected turkeys showed the largest amount of HPAI virus in the lung (4.4%). The HPAI virus was rapidly selected in experimentally infected chickens after both intravenous and intranasal/intratracheal inoculation with a mixed virus preparation. Full-genome sequencing revealed that both pathotypes contained a deletion in the stalk region of the neuraminidase protein. We identified additional mutations in HA and polymerase basic protein 1 (PB1) in the HPAI virus, which were already present as minority variants in the LPAI virus population. Our findings provide more insight into the molecular changes and mechanisms involved in the emergence and selection of HPAI viruses.IMPORTANCE Low-pathogenicity avian influenza (LPAI) viruses circulate in wild birds and can be transmitted to poultry. LPAI viruses can mutate to become highly pathogenic avian influenza (HPAI) viruses causing severe disease and death in poultry. Little is known about this switch to high pathogenicity. We isolated an LPAI H7N3 virus from an infected turkey farm and showed that this contains small amounts of HPAI virus. The HPAI virus rapidly outcompeted the LPAI virus in chickens that were experimentally infected with this mixture of viruses. We analyzed the genome sequences of the LPAI and HPAI viruses and identified several changes that may be important for a virus to become highly pathogenic. This knowledge may be used for timely identification of LPAI viruses that pose a risk of becoming highly pathogenic in the field.
Collapse
Affiliation(s)
- Nancy Beerens
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Rene Heutink
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Frank Harders
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Alex Bossers
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Guus Koch
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Ben Peeters
- Wageningen Bioveterinary Research, Lelystad, The Netherlands
| |
Collapse
|
35
|
Parvin R, Schinkoethe J, Grund C, Ulrich R, Bönte F, Behr KP, Voss M, Samad MA, Hassan KE, Luttermann C, Beer M, Harder T. Comparison of pathogenicity of subtype H9 avian influenza wild-type viruses from a wide geographic origin expressing mono-, di-, or tri-basic hemagglutinin cleavage sites. Vet Res 2020; 51:48. [PMID: 32234073 PMCID: PMC7106749 DOI: 10.1186/s13567-020-00771-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/11/2020] [Indexed: 01/18/2023] Open
Abstract
An intravenous pathogenicity index (IVPI) of > 1.2 in chickens or, in case of subtypes H5 and H7, expression of a polybasic hemagglutinin cleavage site (HACS), signals high pathogenicity (HP). Viruses of the H9N2-G1 lineage, which spread across Asia and Africa, are classified to be of low pathogenicity although, in the field, they became associated with severe clinical signs and epizootics in chickens. Here we report on a pre-eminent trait of recent H9N2-G1 isolates from Bangladesh and India, which express a tribasic HACS (motif PAKSKR-GLF; reminiscent of an HPAIV-like polybasic HACS) and compare their features to H9Nx viruses with di- and monobasic HACS from other phylogenetic and geographic origins. In an in vitro assay, the tribasic HACS of H9N2 was processed by furin-like proteases similar to bona fide H5 HPAIV while some dibasic sites showed increased cleavability but monobasic HACS none. Yet, all viruses remained trypsin-dependent in cell culture. In ovo, only tribasic H9N2 viruses were found to replicate in a grossly extended spectrum of embryonic organs. In contrast to all subtype H5/H7 HPAI viruses, tribasic H9N2 viruses did not replicate in endothelial cells either in the chorio-allantoic membrane or in other embryonic tissues. By IVPI, all H9Nx isolates proved to be of low pathogenicity. Pathogenicity assessment of tribasic H9N2-G1 viruses remains problematic. It cannot be excluded that the formation of a third basic amino acid in the HACS forms an intermediate step towards a gain in pathogenicity. Continued observation of the evolution of these viruses in the field is recommended.
Collapse
Affiliation(s)
- Rokshana Parvin
- Institute of Diagnostic Virology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute (FLI), Suedufer 10, 17493, Greifswald-Insel Riems, Germany.,Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Jan Schinkoethe
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 33, 04103, Leipzig, Germany
| | - Christian Grund
- Institute of Diagnostic Virology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute (FLI), Suedufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Reiner Ulrich
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 33, 04103, Leipzig, Germany
| | - Franziska Bönte
- University of Applied Sciences Wedel, Feldstraße 143, 22880, Wedel, Germany
| | - Klaus P Behr
- AniCon Labor GmbH, Mühlenstraße, 49685, Höltinghausen, Germany
| | - Matthias Voss
- Lohmann Tierzucht GmbH, Veterinär-Labor, Abschnede 64, 27472, Cuxhaven, Germany
| | - Mohammed A Samad
- NRL-AI, Bangladesh Livestock Research Institute (BLRI), Savar, Dhaka, Bangladesh
| | - Kareem E Hassan
- Institute of Diagnostic Virology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute (FLI), Suedufer 10, 17493, Greifswald-Insel Riems, Germany.,Poultry Diseases Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Christine Luttermann
- Institute of Immunology, Friedrich-Loeffler-Institute, Greifswald-Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute (FLI), Suedufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute (FLI), Suedufer 10, 17493, Greifswald-Insel Riems, Germany.
| |
Collapse
|
36
|
Laporte M, Stevaert A, Raeymaekers V, Boogaerts T, Nehlmeier I, Chiu W, Benkheil M, Vanaudenaerde B, Pöhlmann S, Naesens L. Hemagglutinin Cleavability, Acid Stability, and Temperature Dependence Optimize Influenza B Virus for Replication in Human Airways. J Virol 2019; 94:e01430-19. [PMID: 31597759 PMCID: PMC6912116 DOI: 10.1128/jvi.01430-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/28/2019] [Indexed: 12/15/2022] Open
Abstract
Influenza A virus (IAV) and influenza B virus (IBV) cause yearly epidemics with significant morbidity and mortality. When zoonotic IAVs enter the human population, the viral hemagglutinin (HA) requires adaptation to achieve sustained virus transmission. In contrast, IBV has been circulating in humans, its only host, for a long period of time. Whether this entailed adaptation of IBV HA to the human airways is unknown. To address this question, we compared two seasonal IAVs (A/H1N1 and A/H3N2) and two IBVs (B/Victoria and B/Yamagata lineages) with regard to host-dependent activity of HA as the mediator of membrane fusion during viral entry. We first investigated proteolytic activation of HA by covering all type II transmembrane serine protease (TTSP) and kallikrein enzymes, many of which proved to be present in human respiratory epithelium. The IBV HA0 precursor is cleaved by a broader panel of TTSPs and activated with much higher efficiency than IAV HA0. Accordingly, knockdown of a single protease, TMPRSS2, abrogated spread of IAV but not IBV in human respiratory epithelial cells. Second, the HA fusion pH values proved similar for IBV and human-adapted IAVs (with one exception being the HA of 1918 IAV). Third, IBV HA exhibited higher expression at 33°C, a temperature required for membrane fusion by B/Victoria HA. This indicates pronounced adaptation of IBV HA to the mildly acidic pH and cooler temperature of human upper airways. These distinct and intrinsic features of IBV HA are compatible with extensive host adaptation during prolonged circulation of this respiratory virus in the human population.IMPORTANCE Influenza epidemics are caused by influenza A and influenza B viruses (IAV and IBV, respectively). IBV causes substantial disease; however, it is far less studied than IAV. While IAV originates from animal reservoirs, IBV circulates in humans only. Virus spread requires that the viral hemagglutinin (HA) is active and sufficiently stable in human airways. We resolve here how these mechanisms differ between IBV and IAV. Whereas human IAVs rely on one particular protease for HA activation, this is not the case for IBV. Superior activation of IBV by several proteases should enhance shedding of infectious particles. IBV HA exhibits acid stability and a preference for 33°C, indicating pronounced adaptation to the human upper airways, where the pH is mildly acidic and a cooler temperature exists. These adaptive features are rationalized by the long existence of IBV in humans and may have broader relevance for understanding the biology and evolution of respiratory viruses.
Collapse
MESH Headings
- Cell Line
- Epithelial Cells/pathology
- Epithelial Cells/virology
- Gene Expression Regulation
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Host-Pathogen Interactions/genetics
- Humans
- Hydrogen-Ion Concentration
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/metabolism
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/metabolism
- Influenza A Virus, H3N2 Subtype/pathogenicity
- Influenza B virus/genetics
- Influenza B virus/metabolism
- Influenza B virus/pathogenicity
- Influenza, Human/pathology
- Influenza, Human/virology
- Kallikreins/classification
- Kallikreins/genetics
- Kallikreins/metabolism
- Lung/pathology
- Lung/virology
- Membrane Fusion
- Membrane Proteins/classification
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Proteolysis
- Respiratory Mucosa/pathology
- Respiratory Mucosa/virology
- Serine Endopeptidases/deficiency
- Serine Endopeptidases/genetics
- Serine Proteases/classification
- Serine Proteases/genetics
- Serine Proteases/metabolism
- Species Specificity
- Temperature
- Virus Internalization
- Virus Replication/genetics
Collapse
Affiliation(s)
- Manon Laporte
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Annelies Stevaert
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Valerie Raeymaekers
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Talitha Boogaerts
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Winston Chiu
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Mohammed Benkheil
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Bart Vanaudenaerde
- Katholieke Universiteit Leuven, Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Pneumology, University Hospital Leuven, Leuven, Belgium
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Lieve Naesens
- Katholieke Universiteit Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| |
Collapse
|
37
|
Whittaker GR, Straus MR. Human matriptase/ST 14 proteolytically cleaves H7N9 hemagglutinin and facilitates the activation of influenza A/Shanghai/2/2013 virus in cell culture. Influenza Other Respir Viruses 2019; 14:189-195. [PMID: 31820577 PMCID: PMC7040964 DOI: 10.1111/irv.12707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/07/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Influenza is a zoonotic disease that infects millions of people each year resulting in hundreds of thousands of deaths, and in turn devastating pandemics. Influenza is caused by influenza viruses, including influenza A virus (IAV). There are many subtypes of IAV but only a few seem to be able to adapt to humans and to cause disease. In 2013, an H7N9 IAV subtype emerged in China that does not cause clinical symptoms in its chicken host but leads to severe infections when transmitted into humans. Since 2013, there have been six epidemic waves of H7N9 with 1567 laboratory-confirmed human infections and 615 deaths. Pathogenicity of IAV is complex, but a crucial feature contributing to virulence is the activation of the hemagglutinin (HA) fusion protein by host proteases that triggers membrane fusion and leads to subsequent virus propagation. METHODS 293T, VERO, and MDCK cells were used to conduct Western blot analysis, immunofluorescence assays, and pseudoparticle and live virus infections, and to evaluate H7N9 HA cleavage-activation. RESULTS/CONCLUSIONS We show that human matriptase/ST 14 is able to cleave H7N9 HA. Cleavage of H7N9 HA expressed in cell culture results in fusogenic HA and syncytia formation. In infection studies with viral pseudoparticles carrying matriptase/ST 14-activated H7N9 HA, we observed a high infectivity of cells. Finally, human matriptase/ST 14 also activated H7N9 live virus which resulted in high infectivity. Our data demonstrate that human matriptase/ST 14 is a likely candidate protease to promote H7N9 infections in humans.
Collapse
Affiliation(s)
- Gary R Whittaker
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Marco R Straus
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
38
|
Callies LK, Tadeo D, Simper J, Bugge TH, Szabo R. Iterative, multiplexed CRISPR-mediated gene editing for functional analysis of complex protease gene clusters. J Biol Chem 2019; 294:15987-15996. [PMID: 31501243 DOI: 10.1074/jbc.ra119.009773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
Elucidation of gene function by reverse genetics in animal models frequently is complicated by the functional redundancy of homologous genes. This obstacle often is compounded by the tight clustering of homologous genes, which precludes the generation of multigene-deficient animals through standard interbreeding of single-deficient animals. Here, we describe an iterative, multiplexed CRISPR-based approach for simultaneous gene editing in the complex seven-member human airway trypsin-like protease/differentially expressed in a squamous cell carcinoma (HAT/DESC) cluster of membrane-anchored serine proteases. Through four cycles of targeting, we generated a library of 18 unique congenic mouse strains lacking combinations of HAT/DESC proteases, including a mouse strain deficient in all seven proteases. Using this library, we demonstrate that HAT/DESC proteases are dispensable for term development, postnatal health, and fertility and that the recently described function of the HAT-like 4 protease in epidermal barrier formation is unique among all HAT/DESC proteases. The study demonstrates the potential of iterative, multiplexed CRISPR-mediated gene editing for functional analysis of multigene clusters, and it provides a large array of new congenic mouse strains for the study of HAT/DESC proteases in physiological and in pathophysiological processes.
Collapse
Affiliation(s)
- LuLu K Callies
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel Tadeo
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Jan Simper
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Roman Szabo
- Proteases and Tissue Remodeling Section, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
39
|
Poux C, Dondalska A, Bergenstråhle J, Pålsson S, Contreras V, Arasa C, Järver P, Albert J, Busse DC, LeGrand R, Lundeberg J, Tregoning JS, Spetz AL. A Single-Stranded Oligonucleotide Inhibits Toll-Like Receptor 3 Activation and Reduces Influenza A (H1N1) Infection. Front Immunol 2019; 10:2161. [PMID: 31572376 PMCID: PMC6751283 DOI: 10.3389/fimmu.2019.02161] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/28/2019] [Indexed: 12/18/2022] Open
Abstract
The initiation of an immune response is dependent on the activation and maturation of dendritic cells after sensing pathogen associated molecular patterns by pattern recognition receptors. However, the response needs to be balanced as excessive pro-inflammatory cytokine production in response to viral or stress-induced pattern recognition receptor signaling has been associated with severe influenza A virus (IAV) infection. Here, we use an inhibitor of Toll-like receptor (TLR)3, a single-stranded oligonucleotide (ssON) with the capacity to inhibit certain endocytic routes, or a TLR3 agonist (synthetic double-stranded RNA PolyI:C), to evaluate modulation of innate responses during H1N1 IAV infection. Since IAV utilizes cellular endocytic machinery for viral entry, we also assessed ssON's capacity to affect IAV infection. We first show that IAV infected human monocyte-derived dendritic cells (MoDC) were unable to up-regulate the co-stimulatory molecules CD80 and CD86 required for T cell activation. Exogenous TLR3 stimulation did not overcome the IAV-mediated inhibition of co-stimulatory molecule expression in MoDC. However, TLR3 stimulation using PolyI:C led to an augmented pro-inflammatory cytokine response. We reveal that ssON effectively inhibited PolyI:C-mediated pro-inflammatory cytokine production in MoDC, notably, ssON treatment maintained an interferon response induced by IAV infection. Accordingly, RNAseq analyses revealed robust up-regulation of interferon-stimulated genes in IAV cultures treated with ssON. We next measured reduced IAV production in MoDC treated with ssON and found a length requirement for its anti-viral activity, which overlapped with its capacity to inhibit uptake of PolyI:C. Hence, in cases wherein an overreacting TLR3 activation contributes to IAV pathogenesis, ssON can reduce this signaling pathway. Furthermore, concomitant treatment with ssON and IAV infection in mice resulted in maintained weight and reduced viral load in the lungs. Therefore, extracellular ssON provides a mechanism for immune regulation of TLR3-mediated responses and suppression of IAV infection in vitro and in vivo in mice.
Collapse
Affiliation(s)
- Candice Poux
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Aleksandra Dondalska
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Joseph Bergenstråhle
- Science for Life Laboratory, Department of Gene Technology, Royal Institute of Technology, Stockholm, Sweden
| | - Sandra Pålsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Vanessa Contreras
- CEA, UMR1184, IDMIT Department, Institut de Biologie François Jacob, DRF, Fontenay-aux-Roses, France
| | - Claudia Arasa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Peter Järver
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - David C Busse
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Roger LeGrand
- CEA, UMR1184, IDMIT Department, Institut de Biologie François Jacob, DRF, Fontenay-aux-Roses, France
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, Royal Institute of Technology, Stockholm, Sweden
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
40
|
Izaguirre G. The Proteolytic Regulation of Virus Cell Entry by Furin and Other Proprotein Convertases. Viruses 2019; 11:v11090837. [PMID: 31505793 PMCID: PMC6784293 DOI: 10.3390/v11090837] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
A wide variety of viruses exploit furin and other proprotein convertases (PCs) of the constitutive protein secretion pathway in order to regulate their cell entry mechanism and infectivity. Surface proteins of enveloped, as well as non-enveloped, viruses become processed by these proteases intracellularly during morphogenesis or extracellularly after egress and during entry in order to produce mature virions activated for infection. Although viruses also take advantage of other proteases, it is when some viruses become reactive with PCs that they may develop high pathogenicity. Besides reacting with furin, some viruses may also react with the PCs of the other specificity group constituted by PC4/PC5/PACE4/PC7. The targeting of PCs for inhibition may result in a useful strategy to treat infections with some highly pathogenic viruses. A wide variety of PC inhibitors have been developed and tested for their antiviral activity in cell-based assays.
Collapse
Affiliation(s)
- Gonzalo Izaguirre
- College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
41
|
Braun E, Sauter D. Furin-mediated protein processing in infectious diseases and cancer. Clin Transl Immunology 2019; 8:e1073. [PMID: 31406574 PMCID: PMC6682551 DOI: 10.1002/cti2.1073] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/10/2019] [Accepted: 07/13/2019] [Indexed: 12/17/2022] Open
Abstract
Proteolytic cleavage regulates numerous processes in health and disease. One key player is the ubiquitously expressed serine protease furin, which cleaves a plethora of proteins at polybasic recognition motifs. Mammalian substrates of furin include cytokines, hormones, growth factors and receptors. Thus, it is not surprising that aberrant furin activity is associated with a variety of disorders including cancer. Furthermore, the enzymatic activity of furin is exploited by numerous viral and bacterial pathogens, thereby enhancing their virulence and spread. In this review, we describe the physiological and pathophysiological substrates of furin and discuss how dysregulation of a simple proteolytic cleavage event may promote infectious diseases and cancer. One major focus is the role of furin in viral glycoprotein maturation and pathogenicity. We also outline cellular mechanisms regulating the expression and activation of furin and summarise current approaches that target this protease for therapeutic intervention.
Collapse
Affiliation(s)
- Elisabeth Braun
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| | - Daniel Sauter
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| |
Collapse
|
42
|
Löw K, Hardes K, Fedeli C, Seidah NG, Constam DB, Pasquato A, Steinmetzer T, Roulin A, Kunz S. A novel cell-based sensor detecting the activity of individual basic proprotein convertases. FEBS J 2019; 286:4597-4620. [PMID: 31276291 DOI: 10.1111/febs.14979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/13/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023]
Abstract
The basic proprotein convertases (PCs) furin, PC1/3, PC2, PC5/6, PACE4, PC4, and PC7 are promising drug targets for human diseases. However, developing selective inhibitors remains challenging due to overlapping substrate recognition motifs and limited structural information. Classical drug screening approaches for basic PC inhibitors involve homogeneous biochemical assays using soluble recombinant enzymes combined with fluorogenic substrate peptides that may not accurately recapitulate the complex cellular context of the basic PC-substrate interaction. Herein we report basic PC sensor (BPCS), a novel cell-based molecular sensor that allows rapid screening of candidate inhibitors and their selectivity toward individual basic PCs within mammalian cells. BPCS consists of Gaussia luciferase linked to a sortilin-1 membrane anchor via a cleavage motif that allows efficient release of luciferase specifically if individual basic PCs are provided in the same membrane. Screening of selected candidate peptidomimetic inhibitors revealed that BPCS can readily distinguish between general and selective PC inhibitors in a high-throughput screening format. The robust and cost-effective assay format of BPCS makes it suitable to identify novel specific small-molecule inhibitors against basic PCs for therapeutic application. Its cell-based nature will allow screening for drug targets in addition to the catalytically active mature enzyme, including maturation, transport, and cellular factors that modulate the enzyme's activity. This broadened 'target range' will enhance the likelihood to identify novel small-molecule compounds that inhibit basic PCs in a direct or indirect manner and represents a conceptual advantage.
Collapse
Affiliation(s)
- Karin Löw
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Switzerland
| | - Kornelia Hardes
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Germany
| | - Chiara Fedeli
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, (Affiliated to the University of Montreal), Canada
| | - Daniel B Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL) SV ISREC, Switzerland
| | - Antonella Pasquato
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| | - Torsten Steinmetzer
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Germany
| | - Alexandre Roulin
- Department of Ecology and Evolution, University of Lausanne, Switzerland
| | - Stefan Kunz
- Institute of Microbiology, University Hospital Center, University of Lausanne, Switzerland
| |
Collapse
|
43
|
Lambertz RLO, Gerhauser I, Nehlmeier I, Leist SR, Kollmus H, Pöhlmann S, Schughart K. Tmprss2 knock-out mice are resistant to H10 influenza A virus pathogenesis. J Gen Virol 2019; 100:1073-1078. [PMID: 31099738 DOI: 10.1099/jgv.0.001274] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The surface protein haemagglutinin (HA) of influenza A viruses (IAV) needs to be cleaved by a host protease to become functional. Here, we investigated if IAV of the H10 subtype also requires TMPRSS2 for replication and pathogenesis in mice. We first showed in cell culture that TMPRSS2 is able to cleave H10-HA. When Tmprss2-/- deficient mice were infected with a re-assorted virus H10-HA, they did not lose body weight and no viral replication was observed in contrast to wild-type mice. Histopathological analysis showed that inflammatory lesions in the lung of Tmprss2-/- mice were reduced compared to wild-type mice. In addition, no viral antigen was detected in the lungs of Tmprss2-/- mice and no evidence for HA cleavage was observed. We conclude from these studies that TMPRSS2 activity is also essential for in vivo replication and pathogenesis of H10 IAV.
Collapse
Affiliation(s)
- Ruth L O Lambertz
- 1 Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ingo Gerhauser
- 2 Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Inga Nehlmeier
- 3 Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Sarah R Leist
- 1 Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Heike Kollmus
- 1 Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Pöhlmann
- 3 Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany.,4 Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Klaus Schughart
- 1 Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,5 University of Veterinary Medicine Hannover, Hannover, Germany.,6 Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
44
|
Viral Factors Important for Efficient Replication of Influenza A Viruses in Cells of the Central Nervous System. J Virol 2019; 93:JVI.02273-18. [PMID: 30867311 PMCID: PMC6532103 DOI: 10.1128/jvi.02273-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/05/2019] [Indexed: 11/20/2022] Open
Abstract
Central nervous system (CNS) disease is one of the most common extrarespiratory tract complications of influenza A virus infections, and the frequency and severity differ between seasonal, pandemic, and zoonotic influenza viruses. However, little is known about the interaction of these viruses with cells of the CNS. Differences among seasonal, pandemic, and zoonotic influenza viruses in replication efficacy in CNS cells, in vitro, suggest that the presence of an alternative HA cleavage mechanism and ability to attach are important viral factors. Identifying these viral factors and detailed knowledge of the interaction between influenza virus and CNS cells are important to prevent and treat this potentially lethal CNS disease. Central nervous system (CNS) disease is one of the most common extrarespiratory tract complications of influenza A virus infections. Remarkably, zoonotic H5N1 virus infections are more frequently associated with CNS disease than seasonal or pandemic influenza viruses. Little is known about the interaction between influenza A viruses and cells of the CNS; therefore, it is currently unknown which viral factors are important for efficient replication. Here, we determined the replication kinetics of a seasonal, pandemic, zoonotic, and lab-adapted influenza A virus in human neuron-like (SK-N-SH) and astrocyte-like (U87-MG) cells and primary mouse cortex neurons. In general, highly pathogenic avian influenza (HPAI) H5N1 virus replicated most efficiently in all cells, which was associated with efficient attachment and infection. Seasonal H3N2 and to a lesser extent pandemic H1N1 virus replicated in a trypsin-dependent manner in SK-N-SH but not in U87-MG cells. In the absence of trypsin, only HPAI H5N1 and WSN viruses replicated. Removal of the multibasic cleavage site (MBCS) from HPAI H5N1 virus attenuated, but did not abrogate, replication. Taken together, our results showed that the MBCS and, to a lesser extent, the ability to attach are important determinants for efficient replication of HPAI H5N1 virus in cells of the CNS. This suggests that both an alternative hemagglutinin (HA) cleavage mechanism and preference for α-2,3-linked sialic acids allowing efficient attachment contribute to the ability of influenza A viruses to replicate efficiently in cells of the CNS. This study further improves our knowledge on potential viral factors important for the neurotropic potential of influenza A viruses. IMPORTANCE Central nervous system (CNS) disease is one of the most common extrarespiratory tract complications of influenza A virus infections, and the frequency and severity differ between seasonal, pandemic, and zoonotic influenza viruses. However, little is known about the interaction of these viruses with cells of the CNS. Differences among seasonal, pandemic, and zoonotic influenza viruses in replication efficacy in CNS cells, in vitro, suggest that the presence of an alternative HA cleavage mechanism and ability to attach are important viral factors. Identifying these viral factors and detailed knowledge of the interaction between influenza virus and CNS cells are important to prevent and treat this potentially lethal CNS disease.
Collapse
|
45
|
Magnen M, Elsässer BM, Zbodakova O, Kasparek P, Gueugnon F, Petit-Courty A, Sedlacek R, Goettig P, Courty Y. Kallikrein-related peptidase 5 and seasonal influenza viruses, limitations of the experimental models for activating proteases. Biol Chem 2019; 399:1053-1064. [PMID: 29883316 DOI: 10.1515/hsz-2017-0340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/15/2018] [Indexed: 11/15/2022]
Abstract
Every year, influenza A virus (IAV) affects and kills many people worldwide. The viral hemagglutinin (HA) is a critical actor in influenza virus infectivity which needs to be cleaved by host serine proteases to exert its activity. KLK5 has been identified as an activating protease in humans with a preference for the H3N2 IAV subtype. We investigated the origin of this preference using influenza A/Puerto Rico/8/34 (PR8, H1N1) and A/Scotland/20/74 (Scotland, H3N2) viruses. Pretreatment of noninfectious virions with human KLK5 increased infectivity of Scotland IAV in MDCK cells and triggered influenza pneumonia in mice. These effects were not observed with the PR8 IAV. Molecular modeling and in vitro enzymatic studies of peptide substrates and recombinant HAs revealed that the sequences around the cleavage site do not represent the sole determinant of the KLK5 preference for the H3N2 subtype. Using mouse Klk5 and Klk5-deficient mice, we demonstrated in vitro and in vivo that the mouse ortholog protease is not an IAV activating enzyme. This may be explained by unfavorable interactions between H3 HA and mKlk5. Our data highlight the limitations of some approaches used to identify IAV-activating proteases.
Collapse
Affiliation(s)
- Mélia Magnen
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| | | | - Olga Zbodakova
- Laboratory of Transgenic Models of Diseases, Division BIOCEV, Institute of Molecular Genetics, CZ-25250 Vestec, Czech Republic
| | - Petr Kasparek
- Laboratory of Transgenic Models of Diseases, Division BIOCEV, Institute of Molecular Genetics, CZ-25250 Vestec, Czech Republic
| | - Fabien Gueugnon
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| | - Agnès Petit-Courty
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Division BIOCEV, Institute of Molecular Genetics, CZ-25250 Vestec, Czech Republic
| | - Peter Goettig
- Department of Biosciences, University of Salzburg, A-5020 Salzburg, Austria
| | - Yves Courty
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| |
Collapse
|
46
|
Magnen M, Gueugnon F, Petit-Courty A, Baranek T, Sizaret D, Brewah YA, Humbles AA, Si-Tahar M, Courty Y. Tissue kallikrein regulates alveolar macrophage apoptosis early in influenza virus infection. Am J Physiol Lung Cell Mol Physiol 2019; 316:L1127-L1140. [PMID: 30908937 DOI: 10.1152/ajplung.00379.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Host cell proteases are involved in influenza pathogenesis. We examined the role of tissue kallikrein 1 (KLK1) by comparing wild-type (WT) and KLK1-deficient mice infected with influenza H3N2 virus. The levels of KLK1 in lung tissue and in bronchoalveolar lavage (BAL) fluid increased substantially during infection. KLK1 did not promote virus infectivity despite its trypsin-like activity, but it did decrease the initial virus load. We examined two cell types involved in the early control of pathogen infections, alveolar macrophages (AMs) and natural killer (NK) cells to learn more about the antiviral action of KLK1. Inactivating the Klk1 gene or treating WT mice with an anti-KLK1 monoclonal antibody to remove KLK1 activity accelerated the initial virus-induced apoptotic depletion of AMs. Intranasal instillation of deficient mice with recombinant KLK1 (rKLK1) reversed the phenotype. The levels of granulocyte-macrophage colony-stimulating factor in infected BAL fluid were significantly lower in KLK1-deficient mice than in WT mice. Treating lung epithelial cells with rKLK1 increased secretion of this factor known to enhance AM resistance to pathogen-induced apoptosis. The recruitment of NK cells to the air spaces peaked 3 days after infection in WT mice but not in KLK1-deficient mice, as did increases in several NK-attracting chemokines (CCL2, CCL3, CCL5, and CXCL10) in BAL. Chronic obstructive pulmonary disease (COPD) patients are highly susceptible to viral infection, and we observed that the KLK1 mRNA levels decreased with increasing COPD severity. Our findings indicate that KLK1 intervenes early in the antiviral defense modulating the severity of influenza infection. Decreased KLK1 expression in COPD patients could contribute to the worsening of influenza.
Collapse
Affiliation(s)
- Melia Magnen
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| | - Fabien Gueugnon
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| | - Agnès Petit-Courty
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| | - Thomas Baranek
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| | - Damien Sizaret
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| | | | | | - Mustapha Si-Tahar
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| | - Yves Courty
- INSERM, U1100-Centre d'Etude des Pathologies Respiratoires , Tours , France.,Université de Tours , Tours , France
| |
Collapse
|
47
|
Lambertz RLO, Pippel J, Gerhauser I, Kollmus H, Anhlan D, Hrincius ER, Krausze J, Kühn N, Schughart K. Exchange of amino acids in the H1-haemagglutinin to H3 residues is required for efficient influenza A virus replication and pathology in Tmprss2 knock-out mice. J Gen Virol 2018; 99:1187-1198. [PMID: 30084768 PMCID: PMC6230768 DOI: 10.1099/jgv.0.001128] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The haemagglutinin (HA) of H1N1 and H3N2 influenza A virus (IAV) subtypes has to be activated by host proteases. Previous studies showed that H1N1 virus cannot replicate efficiently in Tmprss2-/- knock-out mice whereas H3N2 viruses are able to replicate to the same levels in Tmprss2-/- as in wild type (WT) mice. Here, we investigated the sequence requirements for the HA molecule that allow IAV to replicate efficiently in the absence of TMPRSS2. We showed that replacement of the H3 for the H1-loop sequence (amino acids 320 to 329, at the C-terminus of HA1) was not sufficient for equal levels of virus replication or severe pathology in Tmprss2-/- knock-out mice compared to WT mice. However, exchange of a distant amino acid from H1 to H3 sequence (E31D) in addition to the HA-loop substitution resulted in virus replication in Tmprss2-/- knock-out mice that was comparable to WT mice. The higher virus replication and lung damage was associated with increased epithelial damage and higher mortality. Our results provide further evidence and insights into host proteases as a promising target for therapeutic intervention of IAV infections.
Collapse
Affiliation(s)
- Ruth L O Lambertz
- 1Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jan Pippel
- 2Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ingo Gerhauser
- 3Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Heike Kollmus
- 1Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Darisuren Anhlan
- 4Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University (WWU) Muenster, Muenster, Germany
| | - Eike R Hrincius
- 4Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University (WWU) Muenster, Muenster, Germany
| | - Joern Krausze
- 2Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Nora Kühn
- 1Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Schughart
- 1Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,6Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.,5University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
48
|
Zmora P, Hoffmann M, Kollmus H, Moldenhauer AS, Danov O, Braun A, Winkler M, Schughart K, Pöhlmann S. TMPRSS11A activates the influenza A virus hemagglutinin and the MERS coronavirus spike protein and is insensitive against blockade by HAI-1. J Biol Chem 2018; 293:13863-13873. [PMID: 29976755 PMCID: PMC6130959 DOI: 10.1074/jbc.ra118.001273] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/15/2018] [Indexed: 12/25/2022] Open
Abstract
The influenza virus hemagglutinin (HA) facilitates viral entry into target cells. Cleavage of HA by host cell proteases is essential for viral infectivity, and the responsible enzymes are potential targets for antiviral intervention. The type II transmembrane serine protease (TTSP) TMPRSS2 has been identified as an HA activator in cell culture and in the infected host. However, it is less clear whether TMPRSS2-related enzymes can also activate HA for spread in target cells. Moreover, the activity of cellular serine protease inhibitors against HA-activating TTSPs is poorly understood. Here, we show that TMPRSS11A, another member of the TTSP family, cleaves and activates the influenza A virus (FLUAV) HA and the Middle East respiratory syndrome coronavirus spike protein (MERS-S). Moreover, we demonstrate that TMPRSS11A is expressed in murine tracheal epithelium, which is a target of FLUAV infection, and in human trachea, suggesting that the protease could support FLUAV spread in patients. Finally, we show that HA activation by the TMPRSS11A-related enzymes human airway tryptase and DESC1, but not TMPRSS11A itself, is blocked by the cellular serine protease inhibitor hepatocyte growth factor activator inhibitor type-1 (HAI-1). Our results suggest that TMPRSS11A could promote FLUAV spread in target cells and that HA-activating TTSPs exhibit differential sensitivity to blockade by cellular serine protease inhibitors.
Collapse
Affiliation(s)
- Pawel Zmora
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany,
| | - Markus Hoffmann
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Heike Kollmus
- the Department of Infection Genetics, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Anna-Sophie Moldenhauer
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Olga Danov
- the Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of REBIRTH Cluster of Excellence, 30625 Hannover, Germany, and
| | - Armin Braun
- the Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of REBIRTH Cluster of Excellence, 30625 Hannover, Germany, and
| | - Michael Winkler
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Klaus Schughart
- the Department of Infection Genetics, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany.,the University of Veterinary Medicine Hannover, 30599 Hannover, Germany.,the Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Stefan Pöhlmann
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany, .,the Faculty of Biology and Psychology, University of Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
49
|
Böttcher-Friebertshäuser E, Garten W, Klenk HD. The Antiviral Potential of Host Protease Inhibitors. ACTIVATION OF VIRUSES BY HOST PROTEASES 2018. [PMCID: PMC7122247 DOI: 10.1007/978-3-319-75474-1_11] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The replication of numerous pathogenic viruses depends on host proteases, which therefore emerged as potential antiviral drug targets. In some cases, e.g., for influenza viruses, their function during the viral propagation cycle is relatively well understood, where they cleave and activate viral surface glycoproteins. For other viruses, e.g., Ebola virus, the function of host proteases during replication is still not clear. Host proteases may also contribute to the pathogenicity of virus infection by activating proinflammatory cytokines. For some coronaviruses, human proteases can also serve in a nonproteolytical fashion simply as receptors for virus entry. However, blocking of such protein-protein contacts is challenging, because receptor surfaces are often flat and difficult to address with small molecules. In contrast, many proteases possess well-defined binding pockets. Therefore, they can be considered as well-druggable targets, especially, if they are extracellularly active. The number of their experimental crystal structures is steadily increasing, which is an important prerequisite for a rational structure-based inhibitor design using computational chemistry tools in combination with classical medicinal chemistry approaches. Moreover, host proteases can be considered as stable targets, and their inhibition should prevent rapid resistance developments, which is often observed when addressing viral proteins. Otherwise, the inhibition of host proteases can also affect normal physiological processes leading to a higher probability of side effects and a narrow therapeutic window. Therefore, they should be preferably used in combination therapies with additional antiviral drugs. This strategy should provide a stronger antiviral efficacy, allow to use lower drug doses, and minimize side effects. Despite numerous experimental findings on their antiviral activity, no small-molecule inhibitors of host proteases have been approved for the treatment of virus infections, so far.
Collapse
Affiliation(s)
| | - Wolfgang Garten
- Institut für Virologie, Philipps Universität, Marburg, Germany
| | | |
Collapse
|
50
|
Böttcher-Friebertshäuser E, Garten W, Klenk HD. Characterization of Proprotein Convertases and Their Involvement in Virus Propagation. ACTIVATION OF VIRUSES BY HOST PROTEASES 2018. [PMCID: PMC7122180 DOI: 10.1007/978-3-319-75474-1_9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Wolfgang Garten
- Institut für Virologie, Philipps Universität, Marburg, Germany
| | | |
Collapse
|