1
|
Ding J, Zhang Q, Jiang J, Zhou N, Yu Z, Wang Z, Meng X, Daggumati L, Liu T, Wang F, Lu Z, Yang X, Yang Z, Zhang H, Thorek DLJ, Du P, Zhu H. Preclinical Evaluation and Pilot Clinical Study of 18F-Labeled Inhibitor Peptide for Noninvasive Positron Emission Tomography Mapping of Angiotensin Converting Enzyme 2. ACS Pharmacol Transl Sci 2024; 7:1758-1769. [PMID: 38898955 PMCID: PMC11184604 DOI: 10.1021/acsptsci.3c00337] [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: 11/24/2023] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the main molecular target for coronavirus SARS-CoV-2 to enter cells. Molecularly specific tracers that bind to ACE2 with high affinity can be used to determine the tissue distribution of this important receptor, noninvasively. A novel targeting PET imaging probe, [18F]AlF-DX600-BCH, was developed to detect the in vivo expression of ACE2 and monitor response to therapy. Preclinical experiments, including biodistribution, PET imaging, and tissue section analysis, were conducted after tests of in vitro and in vivo stability and pharmacokinetics. The agent was advanced to clinical evaluation in 10 volunteers who received [18F]AlF-DX600-BCH PET/CT at 1 and 2 h after injection (NCT04542863). Preclinical results of both biodistribution and PET demonstrated [18F]AlF-DX600-BCH accumulation in rat kidney (standardized uptake value; SUVkidney/normal > 50), along with specific uptake in testes (SUVtestis/normal > 10) tissues. Kidney, gastrointestinal, and bronchial cell labeling were correlated to ACE2 positive by immunohistochemistry (IHC) staining. In clinical imaging, significant tracer accumulation was predominantly observed in the urinary and reproductive system (SUVrenal cortex = 32.00, SUVtestis = 4.56), and the conjunctiva and nasal mucosa saw elevated uptake in several cases. This work is the first report of a radioisotope probe, [18F]AlF-DX600-BCH, targeting ACE2 with promising preliminary preclinical and translational outlook, thereby demonstrating the potential of noninvasive mapping of ACE2.
Collapse
Affiliation(s)
- Jin Ding
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Qian Zhang
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
- Guizhou
University School of Medicine, Guiyang, 550025 Guizhou, China
| | - Jinquan Jiang
- Department
of Radiology, People’s Hospital of
Deyang City, Deyang, 618000 Sichuan, China
| | - Nina Zhou
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Ziyu Yu
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, 100142 Beijing, China
| | - Zilei Wang
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Xiangxi Meng
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Lasya Daggumati
- Department
of Radiology, Washington University in St.
Louis School of Medicine, St. Louis, Missouri 63110, United States
| | - Teli Liu
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Feng Wang
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Zhihao Lu
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), Department of Gastro-intestinal oncology, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, 100142 Beijing, China
| | - Xing Yang
- Department
of Nuclear Medicine, Peking University First
Hospital, No. 8 Xishiku Street, 100034 Beijing, China
| | - Zhi Yang
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| | - Hanwen Zhang
- Department
of Radiology, Washington University in St.
Louis School of Medicine, St. Louis, Missouri 63110, United States
| | - Daniel L. J. Thorek
- Department
of Radiology, Washington University in St.
Louis School of Medicine, St. Louis, Missouri 63110, United States
| | - Peng Du
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, 100142 Beijing, China
| | - Hua Zhu
- Key
Laboratory of Carcinogenesis and Translational Research (Ministry
of Education/Beijing), NMPA Key Laboratory for Research and Evaluation
of Radiopharmaceuticals (National Medical Products Administration),
Department of Nuclear Medicine, Peking University
Cancer Hospital & Institute, No. 52 Fucheng Road, Beijing 100142, China
| |
Collapse
|
2
|
Trionfetti F, Alonzi T, Bontempi G, Terri M, Battistelli C, Montaldo C, Repele F, Rotili D, Valente S, Zwergel C, Matusali G, Maggi F, Goletti D, Tripodi M, Mai A, Strippoli R. HDAC1-3 inhibition increases SARS-CoV-2 replication and productive infection in lung mesothelial and epithelial cells. Front Cell Infect Microbiol 2023; 13:1257683. [PMID: 38162580 PMCID: PMC10757821 DOI: 10.3389/fcimb.2023.1257683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024] Open
Abstract
Background Despite the significant progress achieved in understanding the pathology and clinical management of SARS-CoV-2 infection, still pathogenic and clinical issues need to be clarified. Treatment with modulators of epigenetic targets, i.e., epidrugs, is a current therapeutic option in several cancers and could represent an approach in the therapy of viral diseases. Results Aim of this study was the analysis of the role of histone deacetylase (HDAC) inhibition in the modulation of SARS-CoV-2 infection of mesothelial cells (MCs).MeT5A cells, a pleura MC line, were pre-treated with different specific class I and IIb HDAC inhibitors. Unexpectedly, treatment with HDAC1-3 inhibitors significantly increased ACE2/TMPRSS2 expression, suggesting a role in favoring SARS-CoV-2 infection. We focused our analysis on the most potent ACE2/TMPRSS2 inducer among the inhibitors analysed, MS-275, a HDAC1-3 inhibitor. ACE2/TMPRSS2 expression was validated by Western Blot (WB) and immunofluorescence. The involvement of HDAC inhibition in receptor induction was confirmed by HDAC1/HDAC2 silencing. In accordance to the ACE2/TMPRSS2 expression data, MS-275 increased SARS-CoV-2 replication and virus propagation in Vero E6 cells.Notably, MS-275 was able to increase ACE2/TMPRSS2 expression and SARS-CoV-2 production, although to a lesser extent, also in the lung adenocarcinoma cell line Calu-3 cells.Mechanistically, treatment with MS-275 increased H3 and H4 histone acetylation at ACE2/TMPRSS2 promoters, increasing their transcription. Conclusion This study highlights a previously unrecognized effect of HDAC1-3 inhibition in increasing SARS-CoV-2 cell entry, replication and productive infection correlating with increased expression of ACE2 and TMPRSS2. These data, while adding basic insight into COVID-19 pathogenesis, warn for the use of HDAC inhibitors in SARS-CoV-2 patients.
Collapse
Affiliation(s)
- Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Michela Terri
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | | | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Federica Repele
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
3
|
Li X, Yin W, Li A, Li D, Gao X, Wang R, Cui B, Qiu S, Li R, Jia L, Zuo C, Zhang L, Li M. ACE2 PET to reveal the dynamic patterns of ACE2 recovery in an infection model with pseudocorona virus. J Med Virol 2023; 95:e28470. [PMID: 36606602 DOI: 10.1002/jmv.28470] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/27/2022] [Accepted: 12/25/2022] [Indexed: 01/07/2023]
Abstract
Due to the COVID-19 pandemic, a series of sequelae, such as fatigue, tachypnea, and ageusia, appeared in long COVID patients, but the pathological basis was still uncertain. The targeted radiopharmaceuticals were of potential to systemically and dynamically trace the pathological changes. For the key ACE2 protein in the virus-host interaction, 68 Ga-cyc-DX600 was developed on the basis of DX600 as a PET tracer of ACE2 fluctuation and maintained the ability in differentiating ACE and ACE2. In the temporary infection model inhaled with the radio-traceable pseudovirus in the upper respiratory tract of male humanized ACE2 (hACE2) mice, organ-specific ACE2 dysfunction in acute period and the following ACE2 recovery in a relatively long period was visualized and quantified by ACE2 PET, revealing a complex pattern of virus concentration-dependent degree and time period-dependent tendency of ACE2 recovery, mainly a sudden decrease of apparent ACE2 in the heart, liver, kidneys, lungs, and so on, but the liver was of a quick functional compensation on ACE2 expression after a temporary decrease. ACE2 expression of most organs has recovered to a normal level at 15 days post inhalation, with brain and genitals still of a decreased SUVACE2 ; meanwhile, kidneys were of an increased SUVACE2 . These findings on ACE2 PET were further verified by western blot. When compared with high-resolution computed tomography on structural changes and FDG PET on glycometabolism, ACE2 PET was superior in an earlier diagnostic window during infection and more comprehensive understanding of functional dysfunction post-infection. In the respective ACE2 PET/CT and ACE2 PET/MR scans of a volunteer, the repeatability of SUVACE2 and the ACE2 specificity were further confirmed. In conclusion, 68 Ga-cyc-DX600 was developed as an ACE2-specific tracer, and the corresponding ACE2 PET revealed the dynamic patterns of functional ACE2 recovery and provided a reference and approach to explore the ACE2-related pathological basis of sequelae in long COVID.
Collapse
Affiliation(s)
- Xiao Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Wei Yin
- Department of Radiology, Shanghai Changhai Hospital, Shanghai, China
| | - Ao Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Danni Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Xiaolong Gao
- Department of Radiology, Luodian Hospital, Baoshan District, Shanghai, China
| | - Ruizhi Wang
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Bin Cui
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Shuang Qiu
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Rou Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Lina Jia
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Changjing Zuo
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Lan Zhang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Ming Li
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Pozdnyakova N, Krisanova N, Pastukhov A, Tarasenko A, Dudarenko M, Chernykh A, Pashenko A, Ryabukhin S, Tolstanova G, Volochnyuk D, Borisova T. Neuromodulation by selective angiotensin-converting enzyme 2 inhibitors. Neuroscience 2022; 498:155-173. [PMID: 35817218 DOI: 10.1016/j.neuroscience.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/23/2022] [Accepted: 07/02/2022] [Indexed: 11/25/2022]
Abstract
Here, neuromodulatory effects of selective angiotensin-converting enzyme 2 (ACE2) inhibitors were investigated. Two different types of small molecule ligands for ACE2 inhibition were selected using chemical genetic approach, they were synthesized using developed chemical method and tested using presynaptic rat brain nerve terminals (synaptosomes). EBC-36032 (1 µM) increased in a dose-dependent manner spontaneous and stimulated ROS generation in nerve terminals that was of non-mitochondrial origin. Another inhibitor EBC-36033 (MLN-4760) was inert regarding modulation of ROS generation. EBC-36032 and EBC-36033 (100 µM) did not modulate the exocytotic release of L-[14C]glutamate, whereas both inhibitors decreased the initial rate of uptake, but not accumulation (10 min) of L-[14C]glutamate by nerve terminals. EBC-36032 (100 µM) decreased the exocytotic release as well as the initial rate and accumulation of [3H]GABA by nerve terminals. EBC-36032 and EBC-36033 did not change the extracellular levels and transporter-mediated release of [3H]GABA and L-[14C]glutamate, and tonic leakage of [3H]GABA from nerve terminals. Therefore, synthesized selective ACE2 inhibitors decreased uptake of glutamate and GABA as well as exocytosis of GABA at the presynaptic level. The initial rate of glutamate uptake was the only parameter that was mitigated by both ACE2 inhibitors despite stereochemistry issues. In terms of ACE2-targeted antiviral/anti-SARS-CoV-2 and other therapies, novel ACE2 inhibitors should be checked on the subject of possible renin-angiotensin system (RAS)-independent neurological side effects.
Collapse
Affiliation(s)
- Natalia Pozdnyakova
- The Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kyiv 01054, Ukraine
| | - Natalia Krisanova
- The Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kyiv 01054, Ukraine
| | - Artem Pastukhov
- The Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kyiv 01054, Ukraine
| | - Alla Tarasenko
- The Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kyiv 01054, Ukraine
| | - Marina Dudarenko
- The Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kyiv 01054, Ukraine
| | - Anton Chernykh
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, Kyiv 01033, Ukraine; Enamine Ltd, 78 Chervonotkatska Street, Kyiv 02094, Ukraine
| | - Alexander Pashenko
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, Kyiv 01033, Ukraine; Enamine Ltd, 78 Chervonotkatska Street, Kyiv 02094, Ukraine
| | - Sergey Ryabukhin
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, Kyiv 01033, Ukraine; Enamine Ltd, 78 Chervonotkatska Street, Kyiv 02094, Ukraine
| | - Ganna Tolstanova
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, Kyiv 01033, Ukraine
| | - Dmitriy Volochnyuk
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, Kyiv 01033, Ukraine; Enamine Ltd, 78 Chervonotkatska Street, Kyiv 02094, Ukraine; Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Murmanska Street, Kyiv 02094, Ukraine
| | - Tatiana Borisova
- The Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kyiv 01054, Ukraine.
| |
Collapse
|
5
|
Chen Y, Zhang YN, Yan R, Wang G, Zhang Y, Zhang ZR, Li Y, Ou J, Chu W, Liang Z, Wang Y, Chen YL, Chen G, Wang Q, Zhou Q, Zhang B, Wang C. ACE2-targeting monoclonal antibody as potent and broad-spectrum coronavirus blocker. Signal Transduct Target Ther 2021; 6:315. [PMID: 34433803 PMCID: PMC8385704 DOI: 10.1038/s41392-021-00740-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
The evolution of coronaviruses, such as SARS-CoV-2, makes broad-spectrum coronavirus preventional or therapeutical strategies highly sought after. Here we report a human angiotensin-converting enzyme 2 (ACE2)-targeting monoclonal antibody, 3E8, blocked the S1-subunits and pseudo-typed virus constructs from multiple coronaviruses including SARS-CoV-2, SARS-CoV-2 mutant variants (SARS-CoV-2-D614G, B.1.1.7, B.1.351, B.1.617.1, and P.1), SARS-CoV and HCoV-NL63, without markedly affecting the physiological activities of ACE2 or causing severe toxicity in ACE2 “knock-in” mice. 3E8 also blocked live SARS-CoV-2 infection in vitro and in a prophylactic mouse model of COVID-19. Cryo-EM and “alanine walk” studies revealed the key binding residues on ACE2 interacting with the CDR3 domain of 3E8 heavy chain. Although full evaluation of safety in non-human primates is necessary before clinical development of 3E8, we provided a potentially potent and “broad-spectrum” management strategy against all coronaviruses that utilize ACE2 as entry receptors and disclosed an anti-coronavirus epitope on human ACE2.
Collapse
Affiliation(s)
- Yuning Chen
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Nan Zhang
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-ScienceChinese Academy of Sciences, Wuhan, Hubei, China
| | - Renhong Yan
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, and Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Guifeng Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyuan Zhang
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, and Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Zhe-Rui Zhang
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-ScienceChinese Academy of Sciences, Wuhan, Hubei, China
| | - Yaning Li
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jianxia Ou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wendi Chu
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhijuan Liang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yongmei Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yi-Li Chen
- Dartsbio Pharmaceuticals, Zhongshan, Guangdong, China.,Fudan University, School of Pharmacy, Shanghai, China
| | - Ganjun Chen
- Dartsbio Pharmaceuticals, Zhongshan, Guangdong, China
| | - Qi Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Zhou
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, and Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-ScienceChinese Academy of Sciences, Wuhan, Hubei, China. .,Drug Discovery Center for Infectious Disease, Nankai University, Tianjin, China.
| | - Chunhe Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China. .,Dartsbio Pharmaceuticals, Zhongshan, Guangdong, China. .,Fudan University, School of Pharmacy, Shanghai, China.
| |
Collapse
|
6
|
Shukla SP, Cho KB, Rustagi V, Gao X, Fu X, Zhang SX, Guo B, Udugamasooriya DG. "Molecular Masks" for ACE2 to Effectively and Safely Block SARS-CoV-2 Virus Entry. Int J Mol Sci 2021; 22:ijms22168963. [PMID: 34445669 PMCID: PMC8396575 DOI: 10.3390/ijms22168963] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19) remains a global health crisis, despite the development and success of vaccines in certain countries. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, uses its spike protein to bind to the human cell surface receptor angiotensin-converting enzyme 2 (ACE2), which allows the virus to enter the human body. Using our unique cell screening technology, we identified two ACE2-binding peptoid compounds and developed dimeric derivatives (ACE2P1D1 and ACE2P2D1) that effectively blocked spike protein-ACE2 interaction, resulting in the inhibition of SARS-CoV-2 pseudovirus entry into human cells. ACE2P1D1 and ACE2P2D1 also blocked infection by a D614G mutant pseudovirus. More importantly, these compounds do not decrease ACE2 expression nor its enzyme activity (which is important in normal blood pressure regulation), suggesting safe applicability in humans
Collapse
Affiliation(s)
- Satya Prakash Shukla
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX 77204-5037, USA; (S.P.S.); (K.B.C.); (V.R.); (X.G.)
| | - Kwang Bog Cho
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX 77204-5037, USA; (S.P.S.); (K.B.C.); (V.R.); (X.G.)
| | - Vineeta Rustagi
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX 77204-5037, USA; (S.P.S.); (K.B.C.); (V.R.); (X.G.)
| | - Xiang Gao
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX 77204-5037, USA; (S.P.S.); (K.B.C.); (V.R.); (X.G.)
| | - Xinping Fu
- Department of Biology and Biochemistry, University of Houston, 3455 Cullen Blvd, Houston, TX 77204-5037, USA; (X.F.); (S.X.Z.)
| | - Shaun Xiaoliu Zhang
- Department of Biology and Biochemistry, University of Houston, 3455 Cullen Blvd, Houston, TX 77204-5037, USA; (X.F.); (S.X.Z.)
| | - Bin Guo
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX 77204-5037, USA; (S.P.S.); (K.B.C.); (V.R.); (X.G.)
- Correspondence: (B.G.); (D.G.U.)
| | - D. Gomika Udugamasooriya
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd, Houston, TX 77204-5037, USA; (S.P.S.); (K.B.C.); (V.R.); (X.G.)
- MD Anderson Cancer Center, Department of Cancer Systems Imaging, 1881 East Road, Houston, TX 77030-4009, USA
- Correspondence: (B.G.); (D.G.U.)
| |
Collapse
|
7
|
Najafi Fard S, Petrone L, Petruccioli E, Alonzi T, Matusali G, Colavita F, Castilletti C, Capobianchi MR, Goletti D. In Vitro Models for Studying Entry, Tissue Tropism, and Therapeutic Approaches of Highly Pathogenic Coronaviruses. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8856018. [PMID: 34239932 PMCID: PMC8221881 DOI: 10.1155/2021/8856018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/27/2021] [Accepted: 06/05/2021] [Indexed: 12/31/2022]
Abstract
Coronaviruses (CoVs) are enveloped nonsegmented positive-sense RNA viruses belonging to the family Coronaviridae that contain the largest genome among RNA viruses. Their genome encodes 4 major structural proteins, and among them, the Spike (S) protein plays a crucial role in determining the viral tropism. It mediates viral attachment to the host cell, fusion to the membranes, and cell entry using cellular proteases as activators. Several in vitro models have been developed to study the CoVs entry, pathogenesis, and possible therapeutic approaches. This article is aimed at summarizing the current knowledge about the use of relevant methodologies and cell lines permissive for CoV life cycle studies. The synthesis of this information can be useful for setting up specific experimental procedures. We also discuss different strategies for inhibiting the binding of the S protein to the cell receptors and the fusion process which may offer opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Saeid Najafi Fard
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Linda Petrone
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Tonino Alonzi
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Francesca Colavita
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| |
Collapse
|
8
|
MRC5 cells engineered to express ACE2 serve as a model system for the discovery of antivirals targeting SARS-CoV-2. Sci Rep 2021; 11:5376. [PMID: 33686154 PMCID: PMC7940632 DOI: 10.1038/s41598-021-84882-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
Although the spread of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has resulted in a worldwide pandemic, there are currently no virus-specific drugs that are fully effective against SARS-CoV-2. Only a limited number of human-derived cells are capable of supporting SARS-CoV-2 replication and the infectivity of SARS-CoV-2 in these cells remains poor. In contrast, monkey-derived Vero cells are highly susceptibility to infection with SARS-CoV-2, although they are not suitable for the study of antiviral effects by small molecules due to their limited capacity to metabolize drugs compared to human-derived cells. In this study, our goal was to generate a virus-susceptible human cell line that would be useful for the identification and testing of candidate drugs. Towards this end, we stably transfected human lung-derived MRC5 cells with a lentiviral vector encoding angiotensin-converting enzyme 2 (ACE2), the cellular receptor for SARS-CoV-2. Our results revealed that SARS-CoV-2 replicates efficiently in MRC5/ACE2 cells. Furthermore, viral RNA replication and progeny virus production were significantly reduced in response to administration of the replication inhibitor, remdesivir, in MRC5/ACE2 cells compared with Vero cells. We conclude that the MRC5/ACE2 cells will be important in developing specific anti-viral therapeutics and will assist in vaccine development to combat SARS-CoV-2 infections.
Collapse
|
9
|
Beltrán-Pavez C, Riquelme-Barrios S, Oyarzún-Arrau A, Gaete-Argel A, González-Stegmaier R, Cereceda-Solis K, Aguirre A, Travisany D, Palma-Vejares R, Barriga GP, Gaggero A, Martínez-Valdebenito C, Corre NL, Ferrés M, Balcells ME, Fernandez J, Ramírez E, Villarroel F, Valiente-Echeverría F, Soto-Rifo R. Insights into neutralizing antibody responses in individuals exposed to SARS-CoV-2 in Chile. SCIENCE ADVANCES 2021; 7:eabe6855. [PMID: 33579701 PMCID: PMC7880587 DOI: 10.1126/sciadv.abe6855] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/23/2020] [Indexed: 05/08/2023]
Abstract
Chile has one of the worst numbers worldwide in terms of SARS-CoV-2 positive cases and COVID-19-related deaths per million inhabitants; thus, characterization of neutralizing antibody (NAb) responses in the general population is critical to understanding of immunity at the local level. Given our inability to perform massive classical neutralization assays due to the scarce availability of BSL-3 facilities in the country, we developed and fully characterized an HIV-based SARS-CoV-2 pseudotype, which was used in a 96-well plate format to investigate NAb responses in samples from individuals exposed to SARS-CoV-2 or treated with convalescent plasma. We also identified samples with decreased or enhanced neutralization activity against the D614G spike variant compared with the wild type, indicating the relevance of this variant in host immunity. The data presented here represent the first insights into NAb responses in individuals from Chile, serving as a guide for future studies in the country.
Collapse
Affiliation(s)
- Carolina Beltrán-Pavez
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Sebastián Riquelme-Barrios
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aarón Oyarzún-Arrau
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aracelly Gaete-Argel
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Karina Cereceda-Solis
- Translational Medicine Laboratory, Fundación Arturo López Pérez Cancer Center, Santiago, Chile
| | - Adam Aguirre
- Translational Medicine Laboratory, Fundación Arturo López Pérez Cancer Center, Santiago, Chile
| | - Dante Travisany
- Centro de Modelamiento Matemático UMI-CNRS 2807, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile and Fondap Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- INRIA Chile Research Center, Santiago, Chile
| | - Ricardo Palma-Vejares
- Centro de Modelamiento Matemático UMI-CNRS 2807, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile and Fondap Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gonzalo P Barriga
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Emerging Viruses, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Aldo Gaggero
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Environmental Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Constanza Martínez-Valdebenito
- Departamento de Enfermedades Infecciosas e Inmunología Pediátricas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Infectología y Virología Molecular, Laboratorio de Bioseguridad Nivel 3, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicole Le Corre
- Departamento de Enfermedades Infecciosas e Inmunología Pediátricas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Infectología y Virología Molecular, Laboratorio de Bioseguridad Nivel 3, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcela Ferrés
- Departamento de Enfermedades Infecciosas e Inmunología Pediátricas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratorio de Infectología y Virología Molecular, Laboratorio de Bioseguridad Nivel 3, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Elvira Balcells
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Fernandez
- Subdepartamento de Genética Molecular, Instituto de Salud Pública, Santiago, Chile
| | - Eugenio Ramírez
- Sección Virus Oncogénicos, Subdepartamento de Enfermedades Virales, Instituto de Salud Pública, Santiago, Chile
| | - Franz Villarroel
- Translational Medicine Laboratory, Fundación Arturo López Pérez Cancer Center, Santiago, Chile
| | - Fernando Valiente-Echeverría
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ricardo Soto-Rifo
- SARS-CoV-2 Research Group, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
10
|
Sasaki M, Uemura K, Sato A, Toba S, Sanaki T, Maenaka K, Hall WW, Orba Y, Sawa H. SARS-CoV-2 variants with mutations at the S1/S2 cleavage site are generated in vitro during propagation in TMPRSS2-deficient cells. PLoS Pathog 2021; 17:e1009233. [PMID: 33476327 PMCID: PMC7853460 DOI: 10.1371/journal.ppat.1009233] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/02/2021] [Accepted: 12/08/2020] [Indexed: 01/28/2023] Open
Abstract
The spike (S) protein of Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) binds to a host cell receptor which facilitates viral entry. A polybasic motif detected at the cleavage site of the S protein has been shown to broaden the cell tropism and transmissibility of the virus. Here we examine the properties of SARS-CoV-2 variants with mutations at the S protein cleavage site that undergo inefficient proteolytic cleavage. Virus variants with S gene mutations generated smaller plaques and exhibited a more limited range of cell tropism compared to the wild-type strain. These alterations were shown to result from their inability to utilize the entry pathway involving direct fusion mediated by the host type II transmembrane serine protease, TMPRSS2. Notably, viruses with S gene mutations emerged rapidly and became the dominant SARS-CoV-2 variants in TMPRSS2-deficient cells including Vero cells. Our study demonstrated that the S protein polybasic cleavage motif is a critical factor underlying SARS-CoV-2 entry and cell tropism. As such, researchers should be alert to the possibility of de novo S gene mutations emerging in tissue-culture propagated virus strains.
Collapse
Affiliation(s)
- Michihito Sasaki
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kentaro Uemura
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Shionogi & Co., Ltd., Osaka, Japan
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Science, Hokkaido University, Sapporo, Japan
| | - Akihiko Sato
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Shionogi & Co., Ltd., Osaka, Japan
| | - Shinsuke Toba
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Shionogi & Co., Ltd., Osaka, Japan
| | - Takao Sanaki
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Shionogi & Co., Ltd., Osaka, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Science, Hokkaido University, Sapporo, Japan
- Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - William W Hall
- International Collaboration Unit, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- National Virus Reference Laboratory, School of Medicine, University College of Dublin, Ireland
- Global Virus Network, Baltimore, Maryland, United States of America
| | - Yasuko Orba
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Global Virus Network, Baltimore, Maryland, United States of America
| |
Collapse
|
11
|
Wu CY, Lin YS, Yang YH, Shu LH, Cheng YC, Liu HT. Potential Simultaneous Inhibitors of Angiotensin-Converting Enzyme 2 and Transmembrane Protease, Serine 2. Front Pharmacol 2020; 11:584158. [PMID: 33390952 PMCID: PMC7773841 DOI: 10.3389/fphar.2020.584158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/12/2020] [Indexed: 01/09/2023] Open
Abstract
Outbreak of coronavirus disease 2019 occurred in Wuhan and has rapidly spread to almost all parts of world. GB-1, the herbal formula from Tian Shang Sheng Mu of Chiayi Puzi Peitian Temple, is used for the prophylaxis of SARS-CoV-2 in Taiwan. In this study, we investigated that the effect of GB-1 and the index compounds of GB-1 on the ACE2 and TMPRSS2 expression through in vitro and in vivo study. In our result, GB-1 can inhibit ACE2 and TMPRSS2 protein expression in HepG2 cells, 293T cells, and Caco-2 cells without cytotoxicity. For the mouse model, GB-1 treatment could decrease ACE2 and TMPRSS2 expression levels of the lung and kidney tissue without adverse effects, including nephrotoxicity and hepatotoxicity. In the compositions of GB-1, 0.5-1 mg/ml of Glycyrrhiza uralensis Fisch. ex DC. extract could not inhibit ACE2 mRNA and protein expression in HepG2 cells. In addition, theaflavin-3-gallate could inhibit protein expression of ACE2 and TMPRSS2 without significant cytotoxicity. Our results suggest that GB-1 and theaflavin-3-gallate could act as potential candidates for prophylaxis or treatment of SARS-CoV-2 infection through inhibiting protein expression of ACE2 and TMPRSS2 for the further study.
Collapse
Affiliation(s)
- Ching-Yuan Wu
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,School of Chinese Medicine, College of Medicine, Chang Gung University, Chiayi, Taiwan
| | - Yu-Shih Lin
- Department of Pharmacy, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yao-Hsu Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,School of Chinese Medicine, College of Medicine, Chang Gung University, Chiayi, Taiwan.,Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Li-Hsin Shu
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Ching Cheng
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Hung Te Liu
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| |
Collapse
|
12
|
Zamorano Cuervo N, Grandvaux N. ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. eLife 2020; 9:e61390. [PMID: 33164751 PMCID: PMC7652413 DOI: 10.7554/elife.61390] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
Pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus 19 disease (COVID-19) which presents a large spectrum of manifestations with fatal outcomes in vulnerable people over 70-years-old and with hypertension, diabetes, obesity, cardiovascular disease, COPD, and smoking status. Knowledge of the entry receptor is key to understand SARS-CoV-2 tropism, transmission and pathogenesis. Early evidence pointed to angiotensin-converting enzyme 2 (ACE2) as SARS-CoV-2 entry receptor. Here, we provide a critical summary of the current knowledge highlighting the limitations and remaining gaps that need to be addressed to fully characterize ACE2 function in SARS-CoV-2 infection and associated pathogenesis. We also discuss ACE2 expression and potential role in the context of comorbidities associated with poor COVID-19 outcomes. Finally, we discuss the potential co-receptors/attachment factors such as neuropilins, heparan sulfate and sialic acids and the putative alternative receptors, such as CD147 and GRP78.
Collapse
Affiliation(s)
| | - Nathalie Grandvaux
- CRCHUM - Centre Hospitalier de l’Université de MontréalQuébecCanada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de MontréalQuébecCanada
| |
Collapse
|
13
|
Cantuti-Castelvetri L, Ojha R, Pedro LD, Djannatian M, Franz J, Kuivanen S, van der Meer F, Kallio K, Kaya T, Anastasina M, Smura T, Levanov L, Szirovicza L, Tobi A, Kallio-Kokko H, Österlund P, Joensuu M, Meunier FA, Butcher SJ, Winkler MS, Mollenhauer B, Helenius A, Gokce O, Teesalu T, Hepojoki J, Vapalahti O, Stadelmann C, Balistreri G, Simons M. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 2020; 370:856-860. [PMID: 33082293 PMCID: PMC7857391 DOI: 10.1126/science.abd2985] [Citation(s) in RCA: 1301] [Impact Index Per Article: 325.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Virus-host interactions determine cellular entry and spreading in tissues. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the earlier SARS-CoV use angiotensin-converting enzyme 2 (ACE2) as a receptor; however, their tissue tropism differs, raising the possibility that additional host factors are involved. The spike protein of SARS-CoV-2 contains a cleavage site for the protease furin that is absent from SARS-CoV (see the Perspective by Kielian). Cantuti-Castelvetri et al. now show that neuropilin-1 (NRP1), which is known to bind furin-cleaved substrates, potentiates SARS-CoV-2 infectivity. NRP1 is abundantly expressed in the respiratory and olfactory epithelium, with highest expression in endothelial and epithelial cells. Daly et al. found that the furin-cleaved S1 fragment of the spike protein binds directly to cell surface NRP1 and blocking this interaction with a small-molecule inhibitor or monoclonal antibodies reduced viral infection in cell culture. Understanding the role of NRP1 in SARS-CoV-2 infection may suggest potential targets for future antiviral therapeutics. Science, this issue p. 856, p. 861; see also p. 765 The causative agent of coronavirus disease 2019 (COVID-19) is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). For many viruses, tissue tropism is determined by the availability of virus receptors and entry cofactors on the surface of host cells. In this study, we found that neuropilin-1 (NRP1), known to bind furin-cleaved substrates, significantly potentiates SARS-CoV-2 infectivity, an effect blocked by a monoclonal blocking antibody against NRP1. A SARS-CoV-2 mutant with an altered furin cleavage site did not depend on NRP1 for infectivity. Pathological analysis of olfactory epithelium obtained from human COVID-19 autopsies revealed that SARS-CoV-2 infected NRP1-positive cells facing the nasal cavity. Our data provide insight into SARS-CoV-2 cell infectivity and define a potential target for antiviral intervention.
Collapse
Affiliation(s)
- Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Ravi Ojha
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Liliana D Pedro
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Minou Djannatian
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jonas Franz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Suvi Kuivanen
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | | | - Katri Kallio
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Tuğberk Kaya
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Maria Anastasina
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Teemu Smura
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Lev Levanov
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Leonora Szirovicza
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Allan Tobi
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Hannimari Kallio-Kokko
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pamela Österlund
- Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Sarah J Butcher
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Martin Sebastian Winkler
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.,Paracelsus-Elena-Klinik Kassel, Kassel, Germany
| | - Ari Helenius
- Institute of Biochemistry, ETH Zürich, Zürich, Switzerland
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Tambet Teesalu
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Center for Nanomedicine and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jussi Hepojoki
- Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany.,Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Olli Vapalahti
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland.,Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Giuseppe Balistreri
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland. .,The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|