1
|
Calandria JM, Bazan HEP, Bhattacharjee S, Kautzmann MI, Maness NJ, Bazan NG. ELV-N34, RvD6-Isomer, or NPD1 Halt Replication of SARS-CoV-2 Omicron BA.5 Virus in Human Lung and Nasal Cells. FASEB J 2025; 39:e70563. [PMID: 40407038 PMCID: PMC12100679 DOI: 10.1096/fj.202403197r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025]
Abstract
Current vaccines rely on the sequence of Spike (S) protein to induce immunity against the severe acute respiratory coronavirus-2 (SARS-CoV-2) virus. Because of the high mutation rate of the viral S protein, new mutant strains are developed to generate new infectivity profiles. Bioactive lipid mediators (LMs) derived from docosahexaenoic acid (DHA) are synthesized on demand to sustain homeostasis. The purpose of this study was to determine the action of selected LMs in the viral replication of SARS-CoV-2 Omicron BA.5 variant in human lung and nasal epithelial cells. Cells from healthy donors were infected with Omicron BA.5 for one hour and treated with 500 nM Elovanoid (ELV)-N32, ELV-N34, Resolvin D6 isomer (RvD6i), Neuroprotection D1 (NPD1), or vehicle before and after infection. Impedance was recorded to determine cell death by infectivity. Cells were then immunostained for nucleocapsid (N) protein, microtubule-associated protein 1B-light chain 3 (LC3B), and autophagic proteins. N and S RNA were measured to assess the synthesis of viral components. The addition of ELV-N34 or RvD6i decreased the synthesis of N RNA by 76.7% and 96.9%, respectively, in lung primary culture, while NPD1 exerted the same effect in nasal epithelial cells (61.7% reduction). In lung cells, transcription of autophagy-related gene-3 (ATG3) and Sequestosome 1 (SQSTM1/p62), components of the autophagy initiation process, decreased compared to the non-treated infected cells. The results suggest that specific LMs prevent viral autophagy machinery hijacking, leading to a decrease in BA.5 replication. This novel effect of the bioactive LMs as antivirals, regardless of the protein sequence, would potentially complement vaccination and other prevention and treatment therapeutics.
Collapse
Affiliation(s)
- Jorgelina M. Calandria
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | - Haydee E. P. Bazan
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | - Marie‐Audrey I. Kautzmann
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | | | - Nicolas G. Bazan
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| |
Collapse
|
2
|
Negi V, Kuhn RJ. A BSL-2 chimeric system designed to screen SARS-CoV-2 E protein ion channel inhibitors. J Virol 2025; 99:e0225224. [PMID: 40304492 PMCID: PMC12090776 DOI: 10.1128/jvi.02252-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/12/2025] [Indexed: 05/02/2025] Open
Abstract
A major hindrance to the identification of new drug targets and the large-scale testing of new or existing compound libraries against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is that research on the virus is restricted to biosafety level 3 (BSL-3) laboratories. In such cases, BSL-2 surrogate systems or chimeric and attenuated versions of the virus are developed for safer, faster, and cheaper examination of the stages of the virus life cycle and specific drug targets. In this study, we describe a BSL-2 chimeric viral system utilizing a Sindbis virus background as a tool to study one such target, the SARS-CoV-2 Envelope (E) protein channel activity. This protein is fully conserved between SARS-CoV and SARS-CoV-2 variants of concern (VOCs), except for a threonine to isoleucine mutation in the Omicron variant, making the E ion channel domain an attractive antiviral target for combination therapy. Using a BSL-2-chimeric system, we have been able to show similar inhibition profiles using channel inhibitors as previously reported for E-channel inhibition in authentic SARS-CoV-2. This system has the potential to allow faster initial screening of E-channel inhibitors and can be useful in developing broad-spectrum antivirals against viral channel proteins.IMPORTANCEDespite its importance in viral infections, no antivirals exist against the ion channel activity of the SARS-CoV-2 Envelope (E) protein. The E protein is highly conserved among SARS-CoV-2 variants, making it an attractive target for antiviral therapies. Research on SARS-CoV-2 is restricted to BSL-3 laboratories, creating a bottleneck for screening potential antiviral compounds. This study presents a BSL-2 chimeric system using a Sindbis virus background to study the ion channel activity of the E protein. This novel BSL-2 system bypasses this limitation, offering a safer and faster approach for the initial screening of ion channel inhibitors. By replicating the channel inhibition profiles of authentic SARS-CoV-2 in a more accessible system, this research paves the way for the development of broad-spectrum antivirals against viral channel proteins, potentially expediting the discovery of life-saving treatments for COVID-19 and other viral diseases.
Collapse
Affiliation(s)
- Vashi Negi
- Department of Biological Sicences, Purdue University, West Lafayette, Indiana, USA
| | - Richard J. Kuhn
- Department of Biological Sicences, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
3
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
4
|
Dai J, Feng Y, Long H, Liao Y, Tan L, Sun Y, Song C, Qiu X, Ding C. Dexamethasone disrupts intracellular pH homeostasis to delay coronavirus infectious bronchitis virus cell entry via sodium hydrogen exchanger 3 activation. J Virol 2025:e0189424. [PMID: 40340398 DOI: 10.1128/jvi.01894-24] [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: 10/31/2024] [Accepted: 04/14/2025] [Indexed: 05/10/2025] Open
Abstract
Coronavirus entry into host cells enables the virus to initiate its replication cycle efficiently while evading host immune response. Cell entry is intricately associated with pH levels in the cytoplasm or endosomes. In this study, we observed that the sodium hydrogen exchanger 3 (Na+/H+ exchanger 3 or NHE3), which is strongly activated by dexamethasone (Dex) to promote cell membrane Na+/H+ exchange, was critical for cytoplasmic and endosomal acidification. Dex activates NHE3, which increases intracellular pH and blocks the initiation of coronavirus infectious bronchitis virus (IBV) negative-stranded genomic RNA synthesis. Also, Dex antiviral effects are relieved by the glucocorticoid receptor (GR) antagonist RU486 and the NHE3 selective inhibitor tenapanor. These results show that Dex antiviral effects depend on GR and NHE3 activities. Furthermore, Dex exhibits remarkable dose-dependent inhibition of IBV replication, although its antiviral effects are constrained by specific virus and cell types. To our knowledge, this is the first report to show that Dex helps suppress the entry of coronavirus IBV into cells by promoting proton leak pathways, as well as by precisely tuning luminal pH levels mediated by NHE3. Disrupted cytoplasmic pH homeostasis, triggered by Dex and NHE3, plays a crucial role in impeding coronavirus IBV replication. Therefore, cytoplasmic pH plays an essential role during IBV cell entry, probably assisting viruses at the fusion and/or uncoating stages. The strategic modulation of NHE3 activity to regulate intracellular pH could provide a compelling mechanism when developing potent anti-coronavirus drugs.IMPORTANCESince the outbreak of coronavirus disease 2019, dexamethasone (Dex) has been proven to be the first drug that can reduce the mortality rate of coronavirus patients to a certain extent, but its antiviral effect is limited and its underlying mechanism has not been fully clarified. Here, we comprehensively evaluated the effect of Dex on coronavirus infectious bronchitis virus (IBV) replication and found that the antiviral effect of Dex is achieved by regulating sodium hydrogen exchanger 3 (NHE3) activity through the influence of glucocorticoid receptor on cytoplasmic pH or endosome pH. Dex activates NHE3, leading to an increase in intracellular pH and blocking the initiation of negative-stranded genomic RNA synthesis of coronavirus IBV. In this study, we identified the mechanism by which glucocorticoids counteract coronaviruses in cell models, laying the foundation for the development of novel antiviral drugs.
Collapse
Affiliation(s)
- Jun Dai
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Experimental Animal Center, Zunyi Medical University, Zunyi, China
| | - Yiyi Feng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Hong Long
- Experimental Animal Center, Zunyi Medical University, Zunyi, China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lei Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yingjie Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Cuiping Song
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
5
|
Herrock O, Campbell N, Deer E, Amaral LM, Whitney D, Morris R, Wallace K, Turner T, Cleveland EH, Belk S, Booz GW, Cornelius DC, LaMarca B. Preeclamptic Placental CD19+ B Cells Are Causal to Hypertension During Pregnancy. Hypertension 2025; 82:894-903. [PMID: 40171666 PMCID: PMC12003061 DOI: 10.1161/hypertensionaha.124.24552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Patients with preeclampsia exhibit hypertension and chronic inflammation characterized by CD (cluster determinant) 4+T cells, B cells secreting AT1-AA (agonistic autoantibody against the angiotensin II type 1 receptor), inflammatory cytokines, and complement activation. Importantly, a history of COVID-19 during pregnancy is associated with an increased incidence of a preeclampsia-like phenotype and is partly mediated by CD4+T cells. We recently showed pregnant patients with a history of COVID-19 with or without preeclampsia produce AT1-AA, indicating the importance of B lymphocytes in the progression of preeclampsia and possibly of COVID-19. Therefore, we hypothesize that B cells from patients with preeclampsia with or without COVID-19 history induce the preeclampsia phenotype through AT1-AA. METHODS Placental B cells were isolated from normal pregnant, patients with preeclampsia, normotensive COVID-19 history, or preeclampsia COVID-19 history at delivery. Then, 3×105 B cells were transferred intraperitoneally into pregnant athymic rats at gestational day 12. On gestational day 18, carotid catheters were inserted. On gestational day 19, mean arterial pressure was measured, and tissues were collected. RESULTS Preeclampsia B-cell recipients had significantly increased mean arterial pressure, AT1-AA, inflammatory cytokines, and complement activation compared with normal pregnant B-cell recipients. Recipients of B cells with COVID-19 history had markers of inflammation and hypertension but not to the level of significance as recipients of preeclampsia B cells. Inhibition of AT1-AA attenuated the hypertension that occurred in response to preeclampsia or preeclampsia B cells with COVID-19 history. CONCLUSIONS This study demonstrates the important role of B cells in contributing to hypertension and chronic inflammation during preeclampsia with or without COVID-19 history through secretion of AT1-AA.
Collapse
Affiliation(s)
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Lorena M. Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Darby Whitney
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Rachael Morris
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| | - Kedra Wallace
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Ty Turner
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - E. Hawthorne Cleveland
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Sheila Belk
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - George W. Booz
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Denise C. Cornelius
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
6
|
Ying Q, Zhang X, Gu T, Zhang J, Dong Y, Feng W, Li D, Wu X, Wang F. Apatinib inhibits HTNV by stimulating TFEB-driven lysosome biogenesis to degrade viral protein. Antiviral Res 2025; 237:106124. [PMID: 40020878 DOI: 10.1016/j.antiviral.2025.106124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Hantaan Orthohantavirus (Hantaan virus, HTNV) infection causes hemorrhagic fever with renal syndrome (HFRS) in humans, posing a significant health threat. Currently, there are no long-lasting protective vaccines or specific antivirals available, creating an urgent need for effective antiviral treatments in the clinical management of HFRS. Given that viruses exploit multiple host factors for their replication, host-oriented inhibitors could offer promising therapeutic options. In our study, we screened a library of 2570 drugs and identified apatinib, a kinase inhibitor, as a potent suppressor of HTNV infection both in vitro and in vivo. Mechanistic studies revealed that apatinib exerts its antiviral effect by targeting transcription factor EB (TFEB). Specifically, apatinib inhibits the PI3K-Akt signaling pathway and reduces mTOR phosphorylation, which in turn downregulates TFEB phosphorylation. This facilitates the nuclear translocation of TFEB and enhances lysosomal function by upregulating the expression of lysosome-associated genes and promoting lysosome biogenesis. Consequently, there is an increase in lysosome-mediated viral nucleocapsid protein degradation. The ability of apatinib to stimulate this lysosome-driven antiviral mechanism presents a potential new therapeutic approach for viral infections and offers valuable insights into virus-host interactions during HTNV replication.
Collapse
Affiliation(s)
- Qikang Ying
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Xiaoxiao Zhang
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Tianle Gu
- Department of Pathogen Biology, College of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Junmei Zhang
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Yuhang Dong
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Wenjie Feng
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Dongjing Li
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China.
| | - Fang Wang
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China.
| |
Collapse
|
7
|
Bird LE, Xu B, Hobbs AD, Ziegler AR, Scott NE, Newton P, Thomas DR, Edgington-Mitchell LE, Newton HJ. Coxiella burnetii manipulates the lysosomal protease cathepsin B to facilitate intracellular success. Nat Commun 2025; 16:3844. [PMID: 40274809 PMCID: PMC12022341 DOI: 10.1038/s41467-025-59283-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
The obligate intracellular bacterium Coxiella burnetii establishes an intracellular replicative niche termed the Coxiella-containing vacuole (CCV), which has been characterised as a bacterially modified phagolysosome. How C. burnetii withstands the acidic and degradative properties of this compartment is not well understood. We demonstrate that the key lysosomal protease cathepsin B is actively and selectively removed from C. burnetii-infected cells through a mechanism involving the Dot/Icm type IV-B secretion system effector CvpB. Overexpression of cathepsin B leads to defects in CCV biogenesis and bacterial replication, indicating that removal of this protein represents a strategy to reduce the hostility of the intracellular niche. In addition, we show that C. burnetii infection of mammalian cells induces the secretion of a wider cohort of lysosomal proteins, including cathepsin B, to the extracellular milieu via a mechanism dependent on retrograde traffic. This study reveals that C. burnetii is actively modulating the hydrolase cohort of its replicative niche to promote intracellular success and demonstrates that infection incites the secretory pathway to maintain lysosomal homoeostasis.
Collapse
Affiliation(s)
- Lauren E Bird
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Bangyan Xu
- Department of Biochemistry and Pharmacology at the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew D Hobbs
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Alexander R Ziegler
- Department of Biochemistry and Pharmacology at the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Patrice Newton
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - David R Thomas
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology at the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia.
| | - Hayley J Newton
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.
- Infection Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
8
|
Labrecque M, Brunet-Ratnasingham E, Hamilton LK, Auld D, Montpetit A, Richards B, Durand M, Rousseau S, Finzi A, Kaufmann DE, Tetreault M. Transcriptomic profiling of severe and critical COVID-19 patients reveals alterations in expression, splicing and polyadenylation. Sci Rep 2025; 15:13469. [PMID: 40251257 PMCID: PMC12008264 DOI: 10.1038/s41598-025-95905-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/25/2025] [Indexed: 04/20/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a multi-systemic illness that became a pandemic in March 2020. Although environmental factors and comorbidities can influence disease progression, there is a lack of prognostic markers to predict the severity of COVID-19 illness. Identifying these markers is crucial for improving patient outcomes and appropriately allocating scarce resources. Here, an RNA-sequencing study was conducted on blood samples from unvaccinated, hospitalized patients divided by disease severity; 367 moderate, 173 severe, and 199 critical. Using a bioinformatics approach, we identified differentially expressed genes (DEGs), alternative splicing (AS) and alternative polyadenylation (APA) events that were severity-dependent. In the severe group, we observed a higher expression of kappa immunoglobulins compared to the moderate group. In the critical cohort, a majority of AS events were mutually exclusive exons and APA genes mostly had longer 3'UTRs. Interestingly, multiple genes associated with cytoskeleton, TUBA4A, NRGN, BSG, and CD300A, were differentially expressed, alternatively spliced and polyadenylated in the critical group. Furthermore, several inflammation-related pathways were observed predominantly in critical vs. moderate. We demonstrate that integrating multiple downstream analyses of transcriptomics, from moderate, severe, and critical patients confers a significant advantage in identifying relevant dysregulated genes and pathways.
Collapse
Affiliation(s)
- Marjorie Labrecque
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | | | - Laura K Hamilton
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Daniel Auld
- Department of Human Genetics, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill Genome Centre, McGill University, Montreal, QC, Canada
| | | | - Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Epidemiology, Department of Human Genetics, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Madeleine Durand
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Simon Rousseau
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Andrés Finzi
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Daniel E Kaufmann
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Division of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Martine Tetreault
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
9
|
Stephens EB, Kunec D, Henke W, Vidal RM, Greishaber B, Saud R, Kalamvoki M, Singh G, Kafle S, Trujillo JD, Ferreyra FM, Morozov I, Richt JA. The Role of the Tyrosine-Based Sorting Signals of the ORF3a Protein of SARS-CoV-2 in Intracellular Trafficking and Pathogenesis. Viruses 2025; 17:522. [PMID: 40284965 PMCID: PMC12031507 DOI: 10.3390/v17040522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
The open reading frame 3a (ORF3a) is a protein important to the pathogenicity of SARS-CoV-2. The cytoplasmic domain of ORF3a has three canonical tyrosine-based sorting signals (160YNSV163, 211YYQL213, and 233YNKI236), and a previous study has indicated that mutation of the 160YNSV163 motif abrogated plasma membrane expression and inhibited ORF3a-induced apoptosis. Here, we have systematically removed all three tyrosine-based motifs and assessed the importance of each motif or combination of motifs in trafficking to the cell surface. Our results indicate that the 160YNSV163 motif alone was insufficient for ORF3a cell-surface trafficking, while the 211YYQL213 motif was the most important. Additionally, an ORF3a with all three YxxΦ motifs disrupted (ORF3a-[ΔYxxΦ]) was not transported to the cell surface, and LysoIP studies indicate that ORF3a but not ORF3a-[ΔYxxΦ] was present in late endosome/lysosome fractions. A growth-curve analysis of different SARS-CoV-2 viruses expressing the different mutant ORF3a proteins revealed no significant differences in virus replication. Finally, the inoculation of K18hACE-2 mice indicated that the SARS-CoV-2 lacking the three YxxΦ motifs was less pathogenic than the unmodified SARS-CoV-2. These results indicate that the tyrosine motifs of ORF3a contribute to cell-surface expression and SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Edward B. Stephens
- Department of Microbiology, Molecular Genetics, and Immunology 2000 Cates Hall, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Dusan Kunec
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany; (D.K.)
| | - Wyatt Henke
- Department of Microbiology, Molecular Genetics, and Immunology 2000 Cates Hall, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | | | - Brandon Greishaber
- Department of Microbiology, Molecular Genetics, and Immunology 2000 Cates Hall, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Rabina Saud
- Department of Microbiology, Molecular Genetics, and Immunology 2000 Cates Hall, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Maria Kalamvoki
- Department of Microbiology, Molecular Genetics, and Immunology 2000 Cates Hall, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Gagandeep Singh
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (G.S.); (J.D.T.)
| | - Sujan Kafle
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (G.S.); (J.D.T.)
| | - Jessie D. Trujillo
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (G.S.); (J.D.T.)
| | - Franco Matias Ferreyra
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (G.S.); (J.D.T.)
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (G.S.); (J.D.T.)
| |
Collapse
|
10
|
Bare Y, Defourny K, Bretou M, Van Niel G, Nolte-'t Hoen E, Gaudin R. The endoplasmic reticulum as a cradle for virus and extracellular vesicle secretion. Trends Cell Biol 2025; 35:282-293. [PMID: 39730274 DOI: 10.1016/j.tcb.2024.11.008] [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: 08/23/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024]
Abstract
Extracellular vesicles (EVs) are small membranous carriers of protein, lipid, and nucleic acid cargoes and play a key role in intercellular communication. Recent work has revealed the previously under-recognized participation of endoplasmic reticulum (ER)-associated proteins (ERAPs) during EV secretion, using pathways reminiscent of viral replication and secretion. Here, we present highlights of the literature involving ER/ERAPs in EV biogenesis and propose mechanistic parallels with ERAPs exploited during viral infections. We propose that ERAPs play an active role in the release of EVs and viral particles, and we present views on whether viruses hijack or enhance pre-existing ERAP-dependent secretory machineries or whether they repurpose ERAPs to create new secretory pathways.
Collapse
Affiliation(s)
- Yonis Bare
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS UMR9004, Université Montpellier, Montpellier, France.
| | - Kyra Defourny
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marine Bretou
- Université Paris Cité, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France
| | - Guillaume Van Niel
- CRCI2NA, Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France; GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Esther Nolte-'t Hoen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS UMR9004, Université Montpellier, Montpellier, France.
| |
Collapse
|
11
|
Subramanian SP, Wojtkiewicz M, Yu F, Castro C, Schuette EN, Rodriguez-Paar J, Churko J, Renavikar P, Anderson D, Mahr C, Gundry RL. Integrated Multiomics Reveals Alterations in Paucimannose and Complex Type N-Glycans in Cardiac Tissue of Patients with COVID-19. Mol Cell Proteomics 2025; 24:100929. [PMID: 39988192 DOI: 10.1016/j.mcpro.2025.100929] [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: 10/07/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025] Open
Abstract
Coronavirus infectious disease of 2019 (COVID-19) can lead to cardiac complications, yet the molecular mechanisms driving these effects remain unclear. Protein glycosylation is crucial for viral replication, immune response, and organ function and has been found to change in the lungs and liver of patients with COVID-19. However, how COVID-19 impacts cardiac protein glycosylation has not been defined. Our study combined single nuclei transcriptomics, mass spectrometry (MS)-based glycomics, and lectin-based tissue imaging to investigate alterations in N-glycosylation in the human heart post-COVID-19. We identified significant expression differences in glycogenes involved in N-glycan biosynthesis and MS analysis revealed a reduction in high mannose and isomers of paucimannose structures post-infection, with changes in paucimannose directly correlating with COVID-19 independent of comorbidities. Our observations suggest that COVID-19 primes cardiac tissues to alter the glycome at all levels, namely, metabolism, nucleotide sugar transport, and glycosyltransferase activity. Given the role of N-glycosylation in cardiac function, this study provides a basis for understanding the molecular events leading to cardiac damage post-COVID-19 and informing future therapeutic strategies to treat cardiac complications resulting from coronavirus infections.
Collapse
Affiliation(s)
- Sabarinath Peruvemba Subramanian
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Melinda Wojtkiewicz
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chase Castro
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Erin N Schuette
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jocelyn Rodriguez-Paar
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jared Churko
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Pranav Renavikar
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Daniel Anderson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Claudius Mahr
- Institute for Advanced Cardiac Care, Medical City Healthcare, Dallas, Texas, USA
| | - Rebekah L Gundry
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
12
|
Denus M, Filaquier A, Fargues W, Néel E, Stewart SE, Colladant M, Curel T, Mezghrani A, Marin P, Claeysen S, Rubinsztein DC, Parmentier M, Villeneuve J. A Sensitive and Versatile Cell-Based Assay Combines Luminescence and Trapping Approaches to Monitor Unconventional Protein Secretion. Traffic 2025; 26:e70009. [PMID: 40396394 PMCID: PMC12093449 DOI: 10.1111/tra.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/16/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
In addition to the conventional endoplasmic reticulum (ER)-Golgi secretory pathway, alternative routes are increasingly recognized for their critical roles in exporting a growing number of secreted factors. These alternative processes, collectively referred to as unconventional protein secretion (UcPS), challenge traditional views of protein and membrane trafficking. Unlike the well-characterized molecular machinery of the conventional secretory pathway, the mechanisms underlying UcPS remain poorly understood. Various UcPS pathways may involve direct transport of cytosolic proteins across the plasma membrane or the incorporation of cargo proteins into intracellular compartments redirected for secretion. Identifying the specific chaperones, transporters and fusion machinery involved in UcPS cargo recognition, selection and transport is crucial to decipher how cargo proteins are selectively or synergistically directed through multiple secretory routes. These processes can vary depending on cell type and in response to particular stress conditions or cellular demands, underscoring the need for standardized tools and methods to study UcPS. Here, we combine the sensitivity of split NanoLuc Binary Technology with the versatility of the Retention Using Selective Hooks (RUSH) system to develop a straightforward and reliable cell-based assay for investigating both conventional and unconventional protein secretion. This system allows for the identification of intracellular compartments involved in UcPS cargo trafficking. Additionally, its sensitivity enabled us to demonstrate that disease-associated mutants or variants of Tau and superoxide dismutase-1 (SOD1) show altered secretion via UcPS. Finally, we leveraged this assay to screen for Alzheimer's disease risk factors, revealing a functional link between amyloid-beta production and Tau UcPS. This robust assay provides a powerful tool for increasing our knowledge of protein secretion mechanisms in physiological and pathological contexts.
Collapse
Affiliation(s)
- Morgane Denus
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Aurore Filaquier
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - William Fargues
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Eloïse Néel
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Sarah E. Stewart
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and EnvironmentLa Trobe Institute for Molecular Science, La Trobe UniversityMelbourneVictoriaAustralia
| | - Maëlle Colladant
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Thomas Curel
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Alexandre Mezghrani
- Centre de Biologie Structurale (CBS)University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Philippe Marin
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Sylvie Claeysen
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - David C. Rubinsztein
- Department of Medical GeneticsUniversity of Cambridge, Cambridge Institute for Medical ResearchCambridgeUK
- UK Dementia Research InstituteCambridgeUK
| | - Marie‐Laure Parmentier
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Julien Villeneuve
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERMMontpellierFrance
| |
Collapse
|
13
|
Soultsioti M, de Jong AWM, Blomberg N, Tas A, Giera M, Snijder EJ, Bárcena M. Perturbation of de novo lipogenesis hinders MERS-CoV assembly and release, but not the biogenesis of viral replication organelles. J Virol 2025; 99:e0228224. [PMID: 39976449 PMCID: PMC11915874 DOI: 10.1128/jvi.02282-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Coronaviruses hijack host cell metabolic pathways and resources to support their replication. They induce extensive host endomembrane remodeling to generate viral replication organelles and exploit host membranes for assembly and budding of their enveloped progeny virions. Because of the overall significance of host membranes, we sought to gain insight into the role of host factors involved in lipid metabolism in cells infected with Middle East respiratory syndrome coronavirus (MERS-CoV). We employed a single-cycle infection approach in combination with pharmacological inhibitors, biochemical assays, lipidomics, and light and electron microscopy. Pharmacological inhibition of acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN), key host factors in de novo fatty acid biosynthesis, led to pronounced inhibition of MERS-CoV particle release. Inhibition of ACC led to a profound metabolic switch in Huh7 cells, altering their lipidomic profile and inducing lipolysis. However, despite the extensive changes induced by the ACC inhibitor, the biogenesis of viral replication organelles remained unaffected. Instead, ACC inhibition appeared to affect the trafficking and post-translational modifications of the MERS-CoV envelope proteins. Electron microscopy revealed an accumulation of nucleocapsids in early budding stages, indicating that MERS-CoV assembly is adversely impacted by ACC inhibition. Notably, inhibition of palmitoylation resulted in similar effects, while supplementation of exogenous palmitic acid reversed the compound's inhibitory effects, possibly reflecting a crucial need for palmitoylation of the MERS-CoV spike and envelope proteins for their role in virus particle assembly.IMPORTANCEMiddle East respiratory syndrome coronavirus (MERS-CoV) is the etiological agent of a zoonotic respiratory disease of limited transmissibility between humans. However, MERS-CoV is still considered a high-priority pathogen and is closely monitored by WHO due to its high lethality rate of around 35% of laboratory-confirmed infections. Like other positive-strand RNA viruses, MERS-CoV relies on the host cell's endomembranes to support various stages of its replication cycle. However, in spite of this general reliance of MERS-CoV replication on host cell lipid metabolism, mechanistic insights are still very limited. In our study, we show that pharmacological inhibition of acetyl-CoA carboxylase (ACC), a key enzyme in the host cell's fatty acid biosynthesis pathway, significantly disrupts MERS-CoV particle assembly without exerting a negative effect on the biogenesis of viral replication organelles. Furthermore, our study highlights the potential of ACC as a target for the development of host-directed antiviral therapeutics against coronaviruses.
Collapse
Affiliation(s)
- M. Soultsioti
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - A. W. M. de Jong
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - N. Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - A. Tas
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - M. Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - E. J. Snijder
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - M. Bárcena
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
14
|
Wang Q, Wang R, Hu H, Huo X, Wang F. Lysosomes' fallback strategies: more than just survival or death. Front Cell Dev Biol 2025; 13:1559504. [PMID: 40134576 PMCID: PMC11933002 DOI: 10.3389/fcell.2025.1559504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Lysosomes are heterogeneous, acidic organelles whose proper functionality is critically dependent on maintaining the integrity of their membranes and the acidity within their lumen. When subjected to stress, the lysosomal membrane can become permeabilized, posing a significant risk to the organelle's survival and necessitating prompt repair. Although numerous mechanisms for lysosomal repair have been identified in recent years, the progression of lysosome-related diseases is more closely linked to the organelle's alternative strategies when repair mechanisms fail, particularly in the contexts of aging and pathogen infection. This review explores lysosomal responses to damage, including the secretion of lysosomal contents and the interactions with lysosome-associated organelles in the endolysosomal system. Furthermore, it examines the role of organelles outside this system, such as the endoplasmic reticulum (ER) and Golgi apparatus, as auxiliary organelles of the endolysosomal system. These alternative strategies are crucial to understanding disease progression. For instance, the secretion and spread of misfolded proteins play key roles in neurodegenerative disease advancement, while pathogen escape via lysosomal secretion and lysosomotropic drug expulsion underlie cancer treatment resistance. Reexamining these lysosomal fallback strategies could provide new perspectives on lysosomal biology and their contribution to disease progression.
Collapse
Affiliation(s)
- Quan Wang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Ruolin Wang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Haihui Hu
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xiaoqing Huo
- Huaian Maternity and Child Healthcare Hospital of JiangSu Province, Huaian, China
| | - Fulong Wang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Ren WW, Kawahara R, Suzuki KG, Dipta P, Yang G, Thaysen-Andersen M, Fujita M. MYO18B promotes lysosomal exocytosis by facilitating focal adhesion maturation. J Cell Biol 2025; 224:e202407068. [PMID: 39751400 PMCID: PMC11697975 DOI: 10.1083/jcb.202407068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/17/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Many cancer cells exhibit increased amounts of paucimannose glycans, which are truncated N-glycan structures rarely found in mammals. Paucimannosidic proteins are proposedly generated within lysosomes and exposed on the cell surface through a yet uncertain mechanism. In this study, we revealed that paucimannosidic proteins are produced by lysosomal glycosidases and secreted via lysosomal exocytosis. Interestingly, lysosomal exocytosis preferentially occurred in the vicinity of focal adhesions, protein complexes connecting the actin cytoskeleton to the extracellular matrix. Through genome-wide knockout screening, we identified that MYO18B, an actin crosslinker, is required for focal adhesion maturation, facilitating lysosomal exocytosis and the release of paucimannosidic lysosomal proteins to the extracellular milieu. Moreover, a mechanosensitive cation channel PIEZO1 locally activated at focal adhesions imports Ca2+ necessary for lysosome-plasma membrane fusion. Collectively, our study unveiled an intimate relationship between lysosomal exocytosis and focal adhesion, shedding light on the unexpected interplay between lysosomal activities and cellular mechanosensing.
Collapse
Affiliation(s)
- Wei-Wei Ren
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Rebeca Kawahara
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
| | - Kenichi G.N. Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Division of Advanced Bioimaging, National Cancer Center Research Institute, Tokyo, Japan
| | - Priya Dipta
- School of Natural Sciences, Macquarie University, Sydney, Australia
| | - Ganglong Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Morten Thaysen-Andersen
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
- School of Natural Sciences, Macquarie University, Sydney, Australia
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| |
Collapse
|
16
|
Hinkle JJ, Trychta KA, Wires ES, Osborn RM, Leach JR, Faraz ZF, Svarcbahs R, Richie CT, Dewhurst S, Harvey BK. Subcellular localization of SARS-CoV-2 E and 3a proteins along the secretory pathway. J Mol Histol 2025; 56:98. [PMID: 40025386 PMCID: PMC11872775 DOI: 10.1007/s10735-025-10375-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
SARS-CoV-2 E and 3a proteins are important for the assembly, budding, and release of viral particles. These two transmembrane proteins have been implicated in forming channels in the membrane that allow the transport of ions to favor viral replication. During an active infection, both proteins generally localize to the endoplasmic reticulum (ER), ER-Golgi intermediate compartment (ERGIC), and the Golgi where viral assembly occurs. The ER and Golgi are critical for the proper packaging and trafficking of cellular proteins along the secretory pathways which determine a protein's final destination inside or outside of the cell. The SARS-CoV-2 virus primarily infects epithelial cells that are highly secretory in nature such as those in the lung and gut. Here we quantified the distribution of SARS-CoV-2 E and 3a proteins along the secretory pathways in a human intestinal epithelial cell line. We used NaturePatternMatch to demonstrate that epitope-tagged E and 3a proteins expressed alone via transient transfection have a similar immunoreactivity pattern as E and 3a proteins expressed by wild-type viral infection. While E and 3a proteins localized with all selected cellular markers to varying degrees, 3a protein displayed a higher correlation coefficient with the Golgi, early/late endosome, lysosome, and plasma membrane when compared to E protein. This work is the first to provide quantification of the subcellular distribution of E and 3a proteins along the multiple components of the secretory pathway and serves as a basis to develop models for examining how E and 3a alter proteostasis within these structures and affect their function.
Collapse
Affiliation(s)
- Joshua J Hinkle
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| | - Kathleen A Trychta
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Emily S Wires
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Raven M Osborn
- School of Medicine & Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Justin R Leach
- School of Medicine & Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Zoha F Faraz
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Reinis Svarcbahs
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Stephen Dewhurst
- School of Medicine & Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
17
|
Li MY, Deng K, Cheng XH, Siu LYL, Gao ZR, Naik TS, Stancheva VG, Cheung PPH, Teo QW, van Leur SW, Wong HH, Lan Y, Lam TTY, Sun MX, Zhang NN, Zhang Y, Cao TS, Yang F, Deng YQ, Sanyal S, Qin CF. ARF4-mediated intracellular transport as a broad-spectrum antiviral target. Nat Microbiol 2025; 10:710-723. [PMID: 39972062 DOI: 10.1038/s41564-025-01940-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025]
Abstract
Host factors that are involved in modulating cellular vesicular trafficking of virus progeny could be potential antiviral drug targets. ADP-ribosylation factors (ARFs) are GTPases that regulate intracellular vesicular transport upon GTP binding. Here we demonstrate that genetic depletion of ARF4 suppresses viral infection by multiple pathogenic RNA viruses including Zika virus (ZIKV), influenza A virus (IAV) and SARS-CoV-2. Viral infection leads to ARF4 activation and virus production is rescued upon complementation with active ARF4, but not with inactive mutants. Mechanistically, ARF4 deletion disrupts translocation of virus progeny into the Golgi complex and redirects them for lysosomal degradation, thereby blocking virus release. More importantly, peptides targeting ARF4 show therapeutic efficacy against ZIKV and IAV challenge in mice by inhibiting ARF4 activation. Our findings highlight the role of ARF4 during viral infection and its potential as a broad-spectrum antiviral target for further development.
Collapse
Affiliation(s)
- Ming-Yuan Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kao Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-He Cheng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Lewis Yu-Lam Siu
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhuo-Ran Gao
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Trupti Shivaprasad Naik
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Peter Pak-Hang Cheung
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi-Wen Teo
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie W van Leur
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Ho-Him Wong
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yun Lan
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Tommy Tsan-Yuk Lam
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Meng-Xu Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Na-Na Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yue Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tian-Shu Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Fan Yang
- Institute of Pathogenic Biology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China.
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
18
|
Bahojb Mahdavi SZ, Jebelli A, Aghbash PS, Baradaran B, Amini M, Oroojalian F, Pouladi N, Baghi HB, de la Guardia M, Mokhtarzadeh AA. A comprehensive overview on the crosstalk between microRNAs and viral pathogenesis and infection. Med Res Rev 2025; 45:349-425. [PMID: 39185567 PMCID: PMC11796338 DOI: 10.1002/med.22073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 04/11/2023] [Accepted: 08/04/2024] [Indexed: 08/27/2024]
Abstract
Infections caused by viruses as the smallest infectious agents, pose a major threat to global public health. Viral infections utilize different host mechanisms to facilitate their own propagation and pathogenesis. MicroRNAs (miRNAs), as small noncoding RNA molecules, play important regulatory roles in different diseases, including viral infections. They can promote or inhibit viral infection and have a pro-viral or antiviral role. Also, viral infections can modulate the expression of host miRNAs. Furthermore, viruses from different families evade the host immune response by producing their own miRNAs called viral miRNAs (v-miRNAs). Understanding the replication cycle of viruses and their relation with host miRNAs and v-miRNAs can help to find new treatments against viral infections. In this review, we aim to outline the structure, genome, and replication cycle of various viruses including hepatitis B, hepatitis C, influenza A virus, coronavirus, human immunodeficiency virus, human papillomavirus, herpes simplex virus, Epstein-Barr virus, Dengue virus, Zika virus, and Ebola virus. We also discuss the role of different host miRNAs and v-miRNAs and their role in the pathogenesis of these viral infections.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Department of Biology, Faculty of Basic SciencesAzarbaijan Shahid Madani UniversityTabrizIran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic ScienceHigher Education Institute of Rab‐RashidTabrizIran
- Tuberculosis and Lung Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohammad Amini
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic SciencesAzarbaijan Shahid Madani UniversityTabrizIran
| | | | | | | |
Collapse
|
19
|
Hein MY, Peng D, Todorova V, McCarthy F, Kim K, Liu C, Savy L, Januel C, Baltazar-Nunez R, Sekhar M, Vaid S, Bax S, Vangipuram M, Burgess J, Njoya L, Wang E, Ivanov IE, Byrum JR, Pradeep S, Gonzalez CG, Aniseia Y, Creery JS, McMorrow AH, Sunshine S, Yeung-Levy S, DeFelice BC, Mehta SB, Itzhak DN, Elias JE, Leonetti MD. Global organelle profiling reveals subcellular localization and remodeling at proteome scale. Cell 2025; 188:1137-1155.e20. [PMID: 39742809 DOI: 10.1016/j.cell.2024.11.028] [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: 06/14/2024] [Revised: 10/05/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025]
Abstract
Defining the subcellular distribution of all human proteins and their remodeling across cellular states remains a central goal in cell biology. Here, we present a high-resolution strategy to map subcellular organization using organelle immunocapture coupled to mass spectrometry. We apply this workflow to a cell-wide collection of membranous and membraneless compartments. A graph-based analysis assigns the subcellular localization of over 7,600 proteins, defines spatial networks, and uncovers interconnections between cellular compartments. Our approach can be deployed to comprehensively profile proteome remodeling during cellular perturbation. By characterizing the cellular landscape following HCoV-OC43 viral infection, we discover that many proteins are regulated by changes in their spatial distribution rather than by changes in abundance. Our results establish that proteome-wide analysis of subcellular remodeling provides key insights for elucidating cellular responses, uncovering an essential role for ferroptosis in OC43 infection. Our dataset can be explored at organelles.czbiohub.org.
Collapse
Affiliation(s)
| | - Duo Peng
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | | | | | - Kibeom Kim
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Chad Liu
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Laura Savy
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | | | | | | | - Sophie Bax
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - James Burgess
- Institute for Computational & Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Leila Njoya
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Eileen Wang
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | | | | | | | | | | | | | - Sara Sunshine
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Serena Yeung-Levy
- Chan Zuckerberg Biohub, San Francisco, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | | | | | | | | |
Collapse
|
20
|
Suhag K, Borkotoky S, Siddiqui SI, Kumar J, Kumar CS, Tatiya P, Ghosh S, Banerjee M. Mechanistic Insights into the Divergent Membrane Activities of a Viroporin from Chikungunya Virus and Its Transframe Variant. ACS Infect Dis 2025; 11:430-441. [PMID: 39745175 DOI: 10.1021/acsinfecdis.4c00562] [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] [Indexed: 05/01/2025]
Abstract
Alphaviruses, a genus of vector-borne viruses in the Togaviridae family, encode a small ion-channel-forming protein, 6K, and its transframe variant (TF) during infections. Although 6K/TF have vital roles in glycoprotein transport, virus assembly, and budding, there is no mechanistic explanation for these functions. We investigated the distinct biochemical functionalities of 6K and TF from the mosquito-borne alphavirus, Chikungunya Virus. We show that like 6K, TF is also capable of forming ion channels in bilayer membranes. The assemblies formed by 6K in membranes are structurally more complex and potentially more ion-restrictive than those formed by TF. Both 6K and TF show strong affinity toward the ER membranes, indicating that the localization of these components at the plasma membrane, as previously reported, is either linked to post-translational modification or mediated through interaction with binding partners. These structural and functional insights may elucidate the distinct roles of 6K and TF in the alphavirus life cycle.
Collapse
Affiliation(s)
- Kirti Suhag
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Subhomoi Borkotoky
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shumaila Iqbal Siddiqui
- Department of Biophysics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Jitender Kumar
- Department of Physics and Astrophysics, Delhi University, New Delhi 110007, India
| | - Chandra Shekhar Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Pushkar Tatiya
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Subhendu Ghosh
- Department of Biophysics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Manidipa Banerjee
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
21
|
Heffner AL, Rouault TA. A Comparison of Conserved Features in the Human Coronavirus Family Shows That Studies of Viruses Less Pathogenic than SARS-CoV-2, Such as HCoV-OC43, Are Good Model Systems for Elucidating Basic Mechanisms of Infection and Replication in Standard Laboratories. Viruses 2025; 17:256. [PMID: 40007010 PMCID: PMC11860170 DOI: 10.3390/v17020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
In 2021, at the height of the COVID-19 pandemic, coronavirus research spiked, with over 83,000 original research articles related to the word "coronavirus" added to the online resource PubMed. Just 2 years later, in 2023, only 30,900 original research articles related to the word "coronavirus" were added. While, irrefutably, the funding of coronavirus research drastically decreased, a possible explanation for the decrease in interest in coronavirus research is that projects on SARS-CoV-2, the causative agent of COVID-19, halted due to the challenge of establishing a good cellular or animal model system. Most laboratories do not have the capabilities to culture SARS-CoV-2 'in house' as this requires a Biosafety Level (BSL) 3 laboratory. Until recently, BSL 2 laboratory research on endemic coronaviruses was arduous due to the low cytopathic effect in isolated cell culture infection models and the lack of means to quantify viral loads. The purpose of this review article is to compare the human coronaviruses and provide an assessment of the latest techniques that use the endemic coronaviruses-HCoV-229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1-as lower-biosafety-risk models for the more pathogenic coronaviruses-SARS-CoV-2, SARS-CoV, and MERS-CoV.
Collapse
Affiliation(s)
- Audrey L. Heffner
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tracey A. Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Saha T, Masum ZU, Biswas A, Mou MA, Ahmed S, Saha T. Inhaled Dry Powder of Antiviral Agents: A Promising Approach to Treating Respiratory Viral Pathogens. Viruses 2025; 17:252. [PMID: 40007007 PMCID: PMC11860668 DOI: 10.3390/v17020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Inhaled dry powder formulations of antiviral agents represent a novel and potentially transformative approach to managing respiratory viral infections. Traditional antiviral therapies in the form of tablets or capsules often face limitations in terms of therapeutic activity, systemic side effects, and delayed onset of action. Dry powder inhalers (DPIs) provide a targeted delivery system, ensuring the direct administration of antivirals to the infection site, the respiratory tract, which potentially enhance therapeutic efficacy and minimize systemic exposure. This review explores the current state of inhaled dry powder antiviral agents, their advantages over traditional routes, and specific formulations under development. We discuss the benefits of targeted delivery, such as improved drug deposition in the lungs and reduced side effects, alongside considerations related to the formulation preparation. In addition, we summarize the developed (published and marketed) inhaled dry powders of antiviral agents.
Collapse
Affiliation(s)
- Tushar Saha
- Mastaplex Ltd., Centre for Innovation, University of Otago, Dunedin 9016, New Zealand
| | - Zia Uddin Masum
- College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA;
| | - Anik Biswas
- Materials and Nanotechnology, North Dakota State University, Fargo, ND 58105, USA;
| | - Moushumi Afroza Mou
- Department of Biological Science, St. John’s University, Queens, New York, NY 11439, USA;
| | - Sohag Ahmed
- Department of Chemistry, West Virginia University, Morgantown, WV 26506, USA;
| | - Tamal Saha
- International Centre for Diarrheal Disease Research, Bangladesh, Dhaka 1212, Bangladesh;
| |
Collapse
|
23
|
Del Sorbo L, Giugliano R, Cerracchio C, Iovane V, Salvatore MM, Serra F, Amoroso MG, Pellegrini F, Levante M, Capozza P, Diakoudi G, Galdiero M, Fusco G, Pratelli A, Andolfi A, Fiorito F. In Vitro Evaluation of Aryl Hydrocarbon Receptor Involvement in Feline Coronavirus Infection. Viruses 2025; 17:227. [PMID: 40006982 PMCID: PMC11860311 DOI: 10.3390/v17020227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Feline coronavirus (FCoV) is an alphacoronavirus (αCoV) that causes moderate or chronic asymptomatic infection in cats. However, in a single infected cat, FCoV can modify its cellular tropism by acquiring the ability to infect macrophages, resulting in the development of feline infectious peritonitis (FIP). In this context, to restrain the impact of FCoV infection, scientific research has focused attention on the development of antiviral therapies involving novel mechanisms of action. Recent studies have demonstrated that aryl hydrocarbon receptor (AhR) signaling regulates the host response to different human and animal CoVs. Hence, the mechanism of action of AhR was evaluated upon FCoV infection in Crandell Feline Kidney (CRFK) and in canine fibrosarcoma (A72) cells. Following infection with feline enteric CoV (FECV), strain "München", a significant activation of AhR and of its target CYP1A1, was observed. The selective AhR antagonist CH223191 provoked a reduction in FCoV replication and in the levels of viral nucleocapsid protein (NP). Furthermore, the effect of the AhR inhibitor on the acidity of lysosomes in infected cells was observed. Our findings indicate that FCoV acts on viral replication that upregulates AhR. CH223191 repressed virus yield through the inhibition of AhR. In this respect, for counteracting FCoV, AhR represents a new target useful for identifying antiviral drugs. Moreover, in the presence of CH223191, the alkalinization of lysosomes in FCoV-infected CRFK cells was detected, outlining their involvement in antiviral activity.
Collapse
Affiliation(s)
- Luca Del Sorbo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
| | - Rosa Giugliano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
| | - Valentina Iovane
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy; (V.I.); (A.A.)
| | - Maria Michela Salvatore
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Francesco Serra
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Maria Grazia Amoroso
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Francesco Pellegrini
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Martina Levante
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Paolo Capozza
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Georgia Diakoudi
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Giovanna Fusco
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Annamaria Pratelli
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Anna Andolfi
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy; (V.I.); (A.A.)
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, Italy
| |
Collapse
|
24
|
Prasad V. Transmission of unfolded protein response-a regulator of disease progression, severity, and spread in virus infections. mBio 2025; 16:e0352224. [PMID: 39772778 PMCID: PMC11796368 DOI: 10.1128/mbio.03522-24] [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] [Indexed: 01/11/2025] Open
Abstract
The unfolded protein response (UPR) is a cell-autonomous stress response aimed at restoring homeostasis due to the accumulation of misfolded proteins in the endoplasmic reticulum (ER). Viruses often hijack the host cell machinery, leading to an accumulation of misfolded proteins in the ER. The cell-autonomous UPR is the immediate response of an infected cell to this stress, aiming to restore normal function by halting protein translation, degrading misfolded proteins, and activating signaling pathways that increase the production of molecular chaperones. The cell-non-autonomous UPR involves the spreading of UPR signals from initially stressed cells to neighboring unstressed cells that lack the stressor. Though viruses are known modulators of cell-autonomous UPR, recent advancements have highlighted that cell-non-autonomous UPR plays a critical role in elucidating how local infections cause systemic effects, thereby contributing to disease symptoms and progression. Additionally, by utilizing cell-non-autonomous UPR, viruses have devised novel strategies to establish a pro-viral state, promoting virus spread. This review discusses examples that have broadened the understanding of the role of UPR in virus infections and disease progression by looking beyond cell-autonomous to non-autonomous processes and mechanistic details of the inducers, spreaders, and receivers of UPR signals.
Collapse
Affiliation(s)
- Vibhu Prasad
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
25
|
Michelucci A, Sforna L, Focaia R, Leonardi MV, Di Battista A, Rastelli G, Vespa S, Boncompagni S, Di Cristina M, Catacuzzeno L. SARS-CoV-2 ORF3a accessory protein is a water-permeable channel that induces lysosome swelling. Commun Biol 2025; 8:170. [PMID: 39905220 PMCID: PMC11794868 DOI: 10.1038/s42003-024-07442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/23/2024] [Indexed: 02/06/2025] Open
Abstract
ORF3a, the most abundantly expressed accessory protein of SARS-CoV-2, plays an essential role in virus egress by inactivating lysosomes through their deacidification. However, the mechanism underlying this process remains unclear. While seminal studies suggested ORF3a being a cation-selective channel (i.e., viroporin), recent works disproved this conclusion. To unravel the potential function of ORF3a, here we employed a multidisciplinary approach including patch-clamp electrophysiology, videoimaging, molecular dynamics (MD) simulations, and electron microscopy. Preliminary structural analyses and patch-clamp recordings in HEK293 cells rule out ORF3a functioning as either viroporin or proton (H+) channel. Conversely, videoimaging experiments demonstrate that ORF3a mediates the transmembrane transport of water. MD simulations identify the tetrameric assembly of ORF3a as the functional water transporter, with a putative selectivity filter for water permeation that includes two essential asparagines, N82 and N119. Consistent with this, N82L and N82W mutations abolish ORF3a-mediated water permeation. Finally, ORF3a expression in HEK293 cells leads to lysosomal volume increase, mitochondrial damage, and accumulation of intracellular membranes, all alterations reverted by the N82W mutation. We propose a novel function for ORF3a as a lysosomal water-permeable channel, essential for lysosome deacidification and inactivation, key steps to promote virus egress.
Collapse
Affiliation(s)
- Antonio Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy.
| | - Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Riccardo Focaia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | | | - Angela Di Battista
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Giorgia Rastelli
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Simone Vespa
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Simona Boncompagni
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Manlio Di Cristina
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy.
| |
Collapse
|
26
|
Song MS, Sim HJ, Eun SH, Jung MK, Hwang SJ, Ham MH, Kwak K, Lee HJ, Kim JY, Jang DG, Chung HC, Shin DH, Kim YJ, Noh SH, Mun JY, Lee JM, Lee MG. Tubular ER structures shaped by ER-phagy receptors engage in stress-induced Golgi bypass. Dev Cell 2025:S1534-5807(25)00031-0. [PMID: 39919755 DOI: 10.1016/j.devcel.2025.01.011] [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: 12/15/2023] [Revised: 10/04/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025]
Abstract
Cellular stresses, particularly endoplasmic reticulum (ER) stress induced by ER-to-Golgi transport blockade, trigger Golgi-independent secretion of cytosolic and transmembrane proteins. However, the molecular mechanisms underlying this unconventional protein secretion (UPS) remain largely elusive. Here, we report that an ER tubulovesicular structure (ER tubular body [ER-TB]), shaped by the tubular ER-phagy receptors ATL3 and RTN3L, plays an important role in stress-induced UPS of transmembrane proteins such as cystic fibrosis transmembrane conductance regulator (CFTR) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Correlative light-electron microscopy analyses demonstrate the formation of ER-TB under UPS-inducing conditions in HEK293 and HeLa cells. Individual gene knockdowns of ATL3 and RTN3 inhibit ER-TB formation and the UPS of trafficking-deficient ΔF508-CFTR. Combined supplementation of ATL3 and RTN3L induces ER-TB formation and UPS. ATL3 also participates in the SARS-CoV-2-associated convoluted membrane formation and Golgi-independent trafficking of SARS-CoV-2 spike protein. These findings suggest that ER-TB serves a common function in mediating stress-induced UPS, which participates in various physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Min Seok Song
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Physiology, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Hun Ju Sim
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sung Ho Eun
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Gastroenterology, National Health Insurance Service Ilsan Hospital, Goyang 10444, Republic of Korea
| | - Min Kyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu 41068, Republic of Korea
| | - Su Jin Hwang
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Hee Ham
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kihyuck Kwak
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hea Ji Lee
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Republic of Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Republic of Korea; Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Dong Geon Jang
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hee Chun Chung
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Hoon Shin
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ye Jin Kim
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Shin Hye Noh
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu 41068, Republic of Korea
| | - Jae Myun Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Min Goo Lee
- Department of Pharmacology, Woo Choo Lee Institute for Precision Drug Development, Graduate School of Medical Science Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
27
|
Zhu Y, Hu H, Guo X, Zhang H, Li D, Dela Cruz CS, Xie W, Xie L, Sharma L, Chang D. Early antiviral use and supplemental oxygen decrease the risk of secondary bacterial infections: a multi-centre, nested, case-control study. J Hosp Infect 2025; 156:87-95. [PMID: 39701496 DOI: 10.1016/j.jhin.2024.12.005] [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: 08/21/2024] [Revised: 11/14/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The purpose of this study was to evaluate the treatment strategies that dictate the host susceptibility to secondary bacterial infections during coronavirus disease 2019 (COVID-19). METHODS This nested, case-control study was conducted in three general hospitals in China between 1st December 2022 and 1st March 2023. A total of 456 confirmed COVID-19 patients matched 1:2 (152 cases and 304 controls) based on age, sex, disease severity and age-adjusted Charlson Comorbidity Index (aCCI) using propensity-score matching (PSM) were included. Association of secondary bacterial infections with treatment strategies including the supportive measures, antiviral, and antibacterial therapies were the main outcome measures. FINDINGS Conditional logistic regression analyses demonstrated that among categorical variables, use of antibiotics, antivirals, intravenous injection of human immunoglobulin, glucocorticoids or anticoagulants were not associated with the risk of secondary bacterial infections in the COVID-19 patients. The use of supplemental oxygen by either low (odds ratio (OR): 0.18, P<0.001) or high flow (OR: 0.06, P<0.001), but not through ventilators were associated with significant protection against secondary bacterial infection. In contrast, feeding through gastric tube (OR: 10.97, P<0.001) or parenteral nutrition (OR: 3.97, P=0.002) was associated with significant increase in the risk of secondary bacterial infections. Similar data were obtained when data were analysed using continuous variables. Further, the early (<5 days post symptom onset, OR: 0.09, P<0.001), but not the late use of antivirals was associated with protection against secondary bacterial infections. CONCLUSIONS Oxygen supplementation in non-ventilator settings and early use of antivirals were associated with decreased incidences of secondary bacterial infections, while parenteral nutrition or tube feedings were associated with increased incidences of secondary bacterial infections.
Collapse
Affiliation(s)
- Y Zhu
- Department of Pulmonary and Critical Care Medicine at the Seventh Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine at the First Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - H Hu
- Department of Pulmonary and Critical Care Medicine at the Seventh Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine at the First Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - X Guo
- Department of Pulmonary and Critical Care Medicine at the Third Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - H Zhang
- Department of Pulmonary and Critical Care Medicine at the Seventh Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - D Li
- Department of Pulmonary and Critical Care Medicine at the Seventh Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - C S Dela Cruz
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - W Xie
- Peking University Clinical Research Institute, Beijing, China
| | - L Xie
- Department of Pulmonary and Critical Care Medicine at the Seventh Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine at the First Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine at the Third Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; State Key Laboratory of Kidney Disease, Beijing, China
| | - L Sharma
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - D Chang
- Department of Pulmonary and Critical Care Medicine at the Seventh Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine at the First Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine at the Third Medical Center, College of Pulmonary and Critical Care Medicine of the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China; State Key Laboratory of Kidney Disease, Beijing, China.
| |
Collapse
|
28
|
Hirabayashi A, Muramoto Y, Takenaga T, Tsunoda Y, Wakazaki M, Sato M, Fujita-Fujiharu Y, Nomura N, Yamauchi K, Onishi C, Nakano M, Toyooka K, Noda T. Coatomer complex I is required for the transport of SARS-CoV-2 progeny virions from the endoplasmic reticulum-Golgi intermediate compartment. mBio 2025; 16:e0333124. [PMID: 39611845 PMCID: PMC11708035 DOI: 10.1128/mbio.03331-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024] Open
Abstract
SARS-CoV-2 undergoes budding within the lumen of the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), and the progeny virions are delivered to the cell surface via vesicular transport. However, the molecular mechanisms remain poorly understood. Using three-dimensional electron microscopic analysis, such as array tomography and electron tomography, we found that virion-transporting vesicles possessed protein coats on their membrane and demonstrated that the protein coat was coatomer complex I (COPI). During the later stages of SARS-CoV-2 infection, we observed a notable alteration in the distribution of COPI and ERGIC throughout the cytoplasm, suggesting their potential involvement in virus replication. Depletion of COPB2, a key component of COPI, led to the confinement of SARS-CoV-2 progeny virions within the ERGIC at the perinuclear region. While the expression levels of viral proteins within cells were comparable, this depletion significantly reduced the efficiency of virion release, leading to the significant reduction of viral replication. Hence, our findings suggest COPI as a critical player in facilitating the transport of SARS-CoV-2 progeny virions from the ERGIC. Thus, COPI could be a promising target for the development of antivirals against SARS-CoV-2. IMPORTANCE SARS-CoV-2 virions are synthesized within the ERGIC and are transported to the cell surface via vesicular transport for release. However, the precise mechanisms remain unclear. Through various electron microscopic techniques, we identified the presence of COPI on virion-transporting vesicles. Alterations in the distribution of COPI and ERGIC in SARS-CoV-2 infected cells are evident, suggesting their involvement in virus replication. When COPB2, a component of COPI, is depleted, progeny virions become trapped within the ERGIC, leading to a reduction in the efficiency of virion release. These findings highlight COPI's crucial role in mediating SARS-CoV-2 vesicular transport from the ERGIC and suggest it as a potential antiviral target.
Collapse
Affiliation(s)
- Ai Hirabayashi
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
| | - Yukiko Muramoto
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto Prefecture, Japan
| | - Toru Takenaga
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
| | - Yugo Tsunoda
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto Prefecture, Japan
| | - Mayumi Wakazaki
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa Prefecture, Japan
| | - Mayuko Sato
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa Prefecture, Japan
| | - Yoko Fujita-Fujiharu
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto Prefecture, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, , Kyoto, Kyoto Prefecture, Japan
| | - Koji Yamauchi
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto Prefecture, Japan
| | - Chiho Onishi
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
| | - Masahiro Nakano
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto Prefecture, Japan
| | - Kiminori Toyooka
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa Prefecture, Japan
| | - Takeshi Noda
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama Prefecture, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto, Kyoto Prefecture, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Kyoto Prefecture, Japan
| |
Collapse
|
29
|
dos Santos F, Vindel-Alfageme J, Ciordia S, Castro V, Orera I, Garaigorta U, Gastaminza P, Corrales F. Dynamic Cellular Proteome Remodeling during SARS-CoV-2 Infection. Identification of Plasma Protein Readouts. J Proteome Res 2025; 24:171-188. [PMID: 39593238 PMCID: PMC11705369 DOI: 10.1021/acs.jproteome.4c00566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
The outbreak of COVID-19, led to an ongoing pandemic with devastating consequences for the global economy and human health. With the global spread of SARS-CoV-2, multidisciplinary initiatives were launched to explore new diagnostic, therapeutic, and vaccination strategies. From this perspective, proteomics could help to understand the mechanisms associated with SARS-CoV-2 infection and to identify new therapeutic options. A TMT-based quantitative proteomics and phosphoproteomics analysis was performed to study the proteome remodeling of human lung alveolar cells expressing human ACE2 (A549-ACE2) after infection with SARS-CoV-2. Detectability and the prognostic value of selected proteins was analyzed by targeted PRM. A total of 6802 proteins and 6428 phospho-sites were identified in A549-ACE2 cells after infection with SARS-CoV-2. The differential proteins here identified revealed that A549-ACE2 cells undergo a time-dependent regulation of essential processes, delineating the precise intervention of the cellular machinery by the viral proteins. From this mechanistic background and by applying machine learning modeling, 29 differential proteins were selected and detected in the serum of COVID-19 patients, 14 of which showed promising prognostic capacity. Targeting these proteins and the protein kinases responsible for the reported phosphorylation changes may provide efficient alternative strategies for the clinical management of COVID-19.
Collapse
Affiliation(s)
- Fátima
Milhano dos Santos
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Jorge Vindel-Alfageme
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Sergio Ciordia
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Victoria Castro
- Department
of Molecular and Cell Biology, National
Center for Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Irene Orera
- Proteomics
Research Core Facility, Instituto Aragonés
de Ciencias de la Salud (IACS), Zaragoza 50009, Spain
| | - Urtzi Garaigorta
- Department
of Molecular and Cell Biology, National
Center for Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Pablo Gastaminza
- Department
of Molecular and Cell Biology, National
Center for Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| | - Fernando Corrales
- Functional
Proteomics Laboratory, National Center for
Biotechnology (CNB-CSIC), Darwin 3, Madrid 28049, Spain
| |
Collapse
|
30
|
Solár P, Šerý O, Vojtíšek T, Krajsa J, Srník M, Dziedzinská R, Králík P, Kessler M, Dubový P, Joukal A, Balcar VJ, Joukal M. The Blood-Cerebrospinal Fluid Barrier as a Potential Entry Site for the SARS-CoV-2 Virus. J Med Virol 2025; 97:e70184. [PMID: 39835622 DOI: 10.1002/jmv.70184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is an RNA virus responsible for coronavirus disease 2019 (COVID-19). While SARS-CoV-2 primarily targets the lungs and airways, it can also infect other organs, including the central nervous system (CNS). The aim of this study was to investigate whether the choroid plexus could serve as a potential entry site for SARS-CoV-2 into the brain. Tissue samples from 24 deceased COVID-19-positive individuals were analyzed. Reverse transcription real-time PCR (RT-qPCR) was performed on selected brain regions, including the choroid plexus, to detect SARS-CoV-2 viral RNA. Additionally, immunofluorescence staining and confocal microscopy were used to detect and localize two characteristic proteins of SARS-CoV-2: the spike protein S1 and the nucleocapsid protein. RT-qPCR analysis confirmed the presence of SARS-CoV-2 viral RNA in the choroid plexus. Immunohistochemical staining revealed viral particles localized in the epithelial cells of the choroid plexus, with the spike protein S1 detected in the late endosomes. Our findings suggest that the blood-cerebrospinal fluid (B-CSF) barrier in the choroid plexus serves as a route of entry for SARS-CoV-2 into the CNS. This study contributes to the understanding of the mechanisms underlying CNS involvement in COVID-19 and highlights the importance of further research to explore potential therapeutic strategies targeting this entry pathway.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Omar Šerý
- Department of Forensic Medicine, St. Anne's Faculty Hospital, Brno, Czech Republic
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Tomáš Vojtíšek
- Department of Forensic Medicine, St. Anne's Faculty Hospital, Brno, Czech Republic
- Department of Forensic Medicine, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Krajsa
- Department of Forensic Medicine, St. Anne's Faculty Hospital, Brno, Czech Republic
- Department of Forensic Medicine, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michal Srník
- Department of Forensic Medicine, St. Anne's Faculty Hospital, Brno, Czech Republic
- Department of Forensic Medicine, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Radka Dziedzinská
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petr Králík
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Markéta Kessler
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petr Dubový
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Andrea Joukal
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vladimir J Balcar
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
- Neuroscience Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney NSW, Sydney, New South Wales, Australia
| | - Marek Joukal
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
31
|
McCaig CD. SARS-CoV-2 Is an Electricity-Driven Virus. Rev Physiol Biochem Pharmacol 2025; 187:361-410. [PMID: 39838019 DOI: 10.1007/978-3-031-68827-0_18] [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] [Indexed: 01/23/2025]
Abstract
One of the most important and challenging biological events of recent times has been the pandemic caused by SARS-CoV-2. Since the underpinning argument behind this book is the ubiquity of electrical forces driving multiple disparate biological events, consideration of key aspects of the SARS-CoV-2 structural proteins is included. Electrical regulation of spike protein, nucleocapsid protein, membrane protein, and envelope protein is included, with several of their activities regulated by LLPS and the multivalent and π-cation and π-π electrical forces that drive phase separation.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
32
|
Sultana R, Stahelin RV. Strengths and limitations of SARS-CoV-2 virus-like particle systems. Virology 2025; 601:110285. [PMID: 39536645 PMCID: PMC11624109 DOI: 10.1016/j.virol.2024.110285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Virus-like particles (VLPs) resemble the parent virus but lack the viral genome, providing a safe and efficient platform for the analysis of virus assembly and budding as well as the development of vaccines and drugs. During the COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the formation of SARS-CoV-2 VLPs was investigated as an alternative to authentic virions because the latter requires biosafety level 3 (BSL-3) facilities. This allowed researchers to model its assembly and budding processes, examine the role of mutations in variants of concern, and determine how the structural proteins interact with each other. Also, the absence of viral genome in VLPs circumvents worries of gains in infectivity via mutagenesis. This review summarizes the strengths and limitations of several SARS-CoV-2 VLP systems and details some of the strides that have been made in using these systems to study virus assembly and budding, viral entry, and antibody and vaccine development.
Collapse
Affiliation(s)
- Rokaia Sultana
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, 47907, West Lafayette, IN, USA
| | - Robert V Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, 47907, West Lafayette, IN, USA.
| |
Collapse
|
33
|
Marano V, Vlachová Š, Tiano SML, Cortese M. A portrait of the infected cell: how SARS-CoV-2 infection reshapes cellular processes and pathways. NPJ VIRUSES 2024; 2:66. [PMID: 40295886 PMCID: PMC11721117 DOI: 10.1038/s44298-024-00076-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/22/2024] [Indexed: 04/30/2025]
Abstract
Positive-sense single-stranded RNA (+ssRNA) viruses exert a profound influence on cellular organelles and metabolic pathway by usurping host processes to promote their replication. In this review, we present a portrait of selected cellular pathways perturbed in SARS-CoV-2 infection: the effect of viral translation, replication and assembly on the morphology and function of the ER, the remodelling of degradative pathways with a focus on the autophagic processes, and the alterations affecting cellular membranes and lipid metabolism. For each of these cellular processes, we highlight the specific viral and host factors involved and their interplay in this microscopic tug-of-war between pro-viral and anti-viral effects that ultimately tip the scale toward the propagation or the resolution of the infection.
Collapse
Affiliation(s)
- Valentina Marano
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- University of Rome Tor Vergata, Rome, Italy
| | - Štěpánka Vlachová
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Scuola Superiore Meridionale, Naples, Italy
| | - Sofia Maria Luigia Tiano
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Scuola Superiore Meridionale, Naples, Italy
| | - Mirko Cortese
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.
- University of Campania Luigi Vanvitelli, Caserta, Italy.
| |
Collapse
|
34
|
Neilsen G, Mathew AM, Castro JM, McFadden WM, Wen X, Ong YT, Tedbury PR, Lan S, Sarafianos SG. Dimming the corona: studying SARS-coronavirus-2 at reduced biocontainment level using replicons and virus-like particles. mBio 2024; 15:e0336823. [PMID: 39530689 PMCID: PMC11633226 DOI: 10.1128/mbio.03368-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
The coronavirus-induced disease 19 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections, has had a devastating impact on millions of lives globally, with severe mortality rates and catastrophic social implications. Developing tools for effective vaccine strategies and platforms is essential for controlling and preventing the recurrence of such pandemics. Moreover, molecular virology tools that facilitate the study of viral pathogens, impact of viral mutations, and interactions with various host proteins are essential. Viral replicon- and virus-like particle (VLP)-based systems are excellent examples of such tools. This review outlines the importance, advantages, and disadvantages of both the replicon- and VLP-based systems that have been developed for SARS-CoV-2 and have helped the scientific community in dimming the intensity of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Grace Neilsen
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Asha Maria Mathew
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jose M. Castro
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - William M. McFadden
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Xin Wen
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Yee T. Ong
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Philip R. Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Shuiyun Lan
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Stefan G. Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
35
|
Xiang Q, Wouters C, Chang P, Lu YN, Liu M, Wang H, Yang J, Pekosz A, Zhang Y, Wang J. Ubiquitin Ligase ITCH Regulates Life Cycle of SARS-CoV-2 Virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.624804. [PMID: 39677672 PMCID: PMC11642887 DOI: 10.1101/2024.12.04.624804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
SARS-CoV-2 infection poses a major threat to public health, and understanding the mechanism of viral replication and virion release would help identify therapeutic targets and effective drugs for combating the virus. Herein, we identified E3 ubiquitin-protein ligase Itchy homolog (ITCH) as a central regulator of SARS-CoV-2 at multiple steps and processes. ITCH enhances the ubiquitination of viral envelope and membrane proteins and mutual interactions of structural proteins, thereby aiding in virion assembly. ITCH-mediated ubiquitination also enhances the interaction of viral proteins to the autophagosome receptor p62, promoting their autophagosome-dependent secretion. Additionally, ITCH disrupts the trafficking of the protease furin and the maturation of cathepsin L, thereby suppressing their activities in cleaving and destabilizing the viral spike protein. Furthermore, ITCH exhibits robust activation during the SARS-CoV-2 replication stage, and SARS-CoV-2 replication is significantly decreased by genetic or pharmacological inhibition of ITCH. These findings provide new insights into the mechanisms of the SARS-CoV-2 life cycle and identify a potential target for developing treatments for the virus-related diseases.
Collapse
Affiliation(s)
- Qiwang Xiang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Camille Wouters
- Department of Molecular Microbiology & Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Peixi Chang
- Department of Veterinary Medicine, University of Maryland, College Park, MD, 20742, USA
| | - Yu-Ning Lu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mingming Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Haocheng Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Junqin Yang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology & Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, MD, 20742, USA
| | - Jiou Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| |
Collapse
|
36
|
Escalera A, Laporte M, Turner S, Karakus U, Gonzalez-Reiche AS, van de Guchte A, Farrugia K, Khalil Z, van Bakel H, Smith D, García-Sastre A, Aydillo T. The impact of S2 mutations on Omicron SARS-CoV-2 cell surface expression and fusogenicity. Emerg Microbes Infect 2024; 13:2297553. [PMID: 38112266 PMCID: PMC10866063 DOI: 10.1080/22221751.2023.2297553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/17/2023] [Indexed: 12/21/2023]
Abstract
SARS-CoV-2 Omicron subvariants are still emerging and spreading worldwide. These variants contain a high number of polymorphisms in the spike (S) glycoprotein that could potentially impact their pathogenicity and transmission. We have previously shown that the S:655Y and P681H mutations enhance S protein cleavage and syncytia formation. Interestingly, these polymorphisms are present in Omicron S protein. Here, we characterized the cleavage efficiency and fusogenicity of the S protein of different Omicron sublineages. Our results showed that Omicron BA.1 subvariant is efficiently cleaved but it is poorly fusogenic compared to previous SARS-CoV-2 strains. To understand the basis of this phenotype, we generated chimeric S protein using combinations of the S1 and S2 domains from WA1, Delta and Omicron BA.1 variants. We found that the S2 domain of Omicron BA.1 hindered efficient cell-cell fusion. Interestingly, this domain only contains six unique polymorphisms never detected before in ancestral SARS-CoV-2 variants. WA1614G S proteins containing the six individuals S2 Omicron mutations were assessed for their fusogenicity and S surface expression after transfection in cells. Results showed that the S:N856K and N969K substitutions decreased syncytia formation and impacted S protein cell surface levels. However, we observed that "first-generation" Omicron sublineages that emerged subsequently, had convergently evolved to an enhanced fusogenic activity and S expression on the surface of infected cells while "second-generation" Omicron variants have highly diverged and showed lineage-specific fusogenic properties. Importantly, our findings could have potential implications in the improvement and redesign of COVID-19 vaccines.
Collapse
Affiliation(s)
- Alba Escalera
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manon Laporte
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sam Turner
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Umut Karakus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana S. Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adriana van de Guchte
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keith Farrugia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zain Khalil
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harm van Bakel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Derek Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
37
|
Cao MM, Li YM, Ding X, Fang F, Yang LY. ARL8B promotes hepatocellular carcinoma progression and inhibits antitumor activity of lenvatinib via MAPK/ERK signaling by interacting with RAB2A. Cell Signal 2024; 124:111470. [PMID: 39413890 DOI: 10.1016/j.cellsig.2024.111470] [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: 07/01/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Tumor recurrence and metastasis are important factors affecting postoperative survival in hepatocellular carcinoma (HCC) patients. ADP Ribosylation factor-like GTPase 8B (ARL8B) plays a crucial role in many biological processes, including lysosomal function, immune response, and cellular communication, all of which are related to the occurrence and development of tumors. However, its role in HCC remains unclear. Herein, we revealed that ARL8B is consistently elevated in HCC tissues compared to normal liver tissues, suggesting an unfavorable outcome in HCC patients. Increased ARL8B levels promoted the malignant phenotype of HCC in vitro and in vivo. Notably, ARL8B also induced epithelial-to-mesenchymal transition (EMT) in HCC cells. Mechanistically, the results of bioinformatics analysis combined with mass spectrometry revealed the potential downstream target molecule RAB2A of ARL8B. ARL8B directly interacted with RAB2A and increased the levels of GTP-bound RAB2A, thereby contributing to the activation of the extracellular signal-regulated kinase (ERK) signaling pathway. Interestingly, knockout of ARL8B in Hep3B cells enhanced the antitumor activity of lenvatinib in vitro and in vivo. Furthermore, AAV-shARL8B enhanced the inhibition of HCC growth through lenvatinib, providing new insights into its mechanism of action in lenvatinib-insensitive patients. In conclusion, ARL8B promotes the malignant phenotype of HCC and EMT via RAB2A mediated activation of the MAPK/ERK signaling pathway and is expected to be a valuable prognostic indicator and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Mo-Mo Cao
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Ming Li
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Ding
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, China
| | - Feng Fang
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
38
|
Duran J, Salinas JE, Wheaton RP, Poolsup S, Allers L, Rosas-Lemus M, Chen L, Cheng Q, Pu J, Salemi M, Phinney B, Ivanov P, Lystad AH, Bhaskar K, Rajaiya J, Perkins DJ, Jia J. Calcium signaling from damaged lysosomes induces cytoprotective stress granules. EMBO J 2024; 43:6410-6443. [PMID: 39533058 PMCID: PMC11649789 DOI: 10.1038/s44318-024-00292-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/18/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Lysosomal damage induces stress granule (SG) formation. However, the importance of SGs in determining cell fate and the precise mechanisms that mediate SG formation in response to lysosomal damage remain unclear. Here, we describe a novel calcium-dependent pathway controlling SG formation, which promotes cell survival during lysosomal damage. Mechanistically, the calcium-activated protein ALIX transduces lysosomal damage signals to SG formation by controlling eIF2α phosphorylation after sensing calcium leakage. ALIX enhances eIF2α phosphorylation by promoting the association between PKR and its activator PACT, with galectin-3 inhibiting this interaction; these regulatory events occur on damaged lysosomes. We further find that SG formation plays a crucial role in promoting cell survival upon lysosomal damage caused by factors such as SARS-CoV-2ORF3a, adenovirus, malarial pigment, proteopathic tau, or environmental hazards. Collectively, these data provide insights into the mechanism of SG formation upon lysosomal damage and implicate it in diseases associated with damaged lysosomes and SGs.
Collapse
Affiliation(s)
- Jacob Duran
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Jay E Salinas
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Rui Ping Wheaton
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Suttinee Poolsup
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Monica Rosas-Lemus
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Li Chen
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Qiuying Cheng
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Jing Pu
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Michelle Salemi
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, CA, 95616, USA
| | - Brett Phinney
- Proteomics Core Facility, University of California Davis Genome Center, University of California, Davis, CA, 95616, USA
| | - Pavel Ivanov
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; HMS Initiative for RNA Medicine, Boston, MA, 02115, USA
| | - Alf Håkon Lystad
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Jaya Rajaiya
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Douglas J Perkins
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Jingyue Jia
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA.
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, Albuquerque, NM, 87106, USA.
| |
Collapse
|
39
|
Zhou CM, Jiang ZZ, Liu N, Yu XJ. Current insights into human pathogenic phenuiviruses and the host immune system. Virulence 2024; 15:2384563. [PMID: 39072499 PMCID: PMC11290763 DOI: 10.1080/21505594.2024.2384563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Phenuiviruses are a class of segmented negative-sense single-stranded RNA viruses, typically consisting of three RNA segments that encode four distinct proteins. The emergence of pathogenic phenuivirus strains, such as Rift Valley fever phlebovirus (RVFV) in sub-Saharan Africa, Severe Fever with Thrombocytopenia Syndrome Virus (SFTSV) in East and Southeast Asia, and Heartland Virus (HRTV) in the United States has presented considerable challenges to global public health in recent years. The innate immune system plays a crucial role as the initial defense mechanism of the host against invading pathogens. In addition to continued research aimed at elucidating the epidemiological characteristics of phenuivirus, significant advancements have been made in investigating its viral virulence factors (glycoprotein, non-structural protein, and nucleoprotein) and potential host-pathogen interactions. Specifically, efforts have focused on understanding mechanisms of viral immune evasion, viral assembly and egress, and host immune networks involving immune cells, programmed cell death, inflammation, nucleic acid receptors, etc. Furthermore, a plethora of technological advancements, including metagenomics, metabolomics, single-cell transcriptomics, proteomics, gene editing, monoclonal antibodies, and vaccines, have been utilized to further our understanding of phenuivirus pathogenesis and host immune responses. Hence, this review aims to provide a comprehensive overview of the current understanding of the mechanisms of host recognition, viral immune evasion, and potential therapeutic approaches during human pathogenic phenuivirus infections focusing particularly on RVFV and SFTSV.
Collapse
Affiliation(s)
- Chuan-Min Zhou
- Gastrointestinal Disease Diagnosis and Treatment Center, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ze-Zheng Jiang
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, China
| | - Ning Liu
- Department of Quality and Operations Management, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xue-Jie Yu
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, China
| |
Collapse
|
40
|
Hoste ACR, Smeralda W, Cugnet A, Brostaux Y, Deleu M, Garigliany M, Jacques P. The structure of lipopeptides impacts their antiviral activity and mode of action against SARS-CoV-2 in vitro. Appl Environ Microbiol 2024; 90:e0103624. [PMID: 39445780 DOI: 10.1128/aem.01036-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Microbial lipopeptides are synthesized by nonribosomal peptide synthetases and are composed of a hydrophobic fatty acid chain and a hydrophilic peptide moiety. These structurally diverse amphiphilic molecules can interact with biological membranes and possess various biological activities, including antiviral properties. This study aimed to evaluate the cytotoxicity and antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) of 15 diverse lipopeptides to understand their structure-activity relationships. Non-ionic lipopeptides were generally more cytotoxic than charged ones, with cationic lipopeptides being less cytotoxic than anionic and non-ionic variants. At 100 µg/mL, six lipopeptides reduced SARS-CoV-2 RNA to undetectable levels in infected Vero E6 cells, while six others achieved a 2.5- to 4.1-log reduction, and three had no significant effect. Surfactin, white line-inducing principle (WLIP), fengycin, and caspofungin emerged as the most promising anti-SARS-CoV-2 agents. Detailed analysis revealed that these four lipopeptides affected various stages of the viral life cycle involving the viral envelope. Surfactin and WLIP significantly reduced viral RNA levels in replication assays, comparable to neutralizing serum. Surfactin uniquely inhibited viral budding, while fengycin impacted viral binding after pre-infection treatment of the cells. Caspofungin demonstrated a lower antiviral effect compared to the others. Key structural traits of lipopeptides influencing their cytotoxic and antiviral activities were identified. Lipopeptides with a high number of amino acids, especially charged (preferentially anionic) amino acids, showed potent anti-SARS-CoV-2 activity. This research paves the way for designing new lipopeptides with low cytotoxicity and high antiviral efficacy, potentially leading to effective treatments. IMPORTANCE This study advances our understanding of how lipopeptides, which are molecules mostly produced by bacteria, with both fat and protein components, can be used to fight viruses like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). By analyzing 15 different lipopeptides, researchers identified key structural features that make some of these molecules particularly effective at reducing viral levels while being less harmful to cells. Specifically, lipopeptides with certain charged amino acids were found to have the strongest antiviral effects. This research lays the groundwork for developing new antiviral treatments that are both potent against viruses and safe for human cells, offering hope for better therapeutic options in the future.
Collapse
Affiliation(s)
- Alexis C R Hoste
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
- Veterinary Pathology, FARAH Research Centre, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Willy Smeralda
- LBMI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Aurélien Cugnet
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Yves Brostaux
- Applied Statistics, Computer Science and Modelling laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Magali Deleu
- LBMI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Mutien Garigliany
- Veterinary Pathology, FARAH Research Centre, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Philippe Jacques
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
41
|
Han H, Liu H, Steiner R, Zhao Z, Jin ZG. Inhibition of protein kinase D and its substrate phosphatidylinositol-4 kinase III beta blocks common human coronavirus replication. Microbiol Spectr 2024; 12:e0150124. [PMID: 39540754 PMCID: PMC11619529 DOI: 10.1128/spectrum.01501-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the infection of a coronavirus, named as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coronaviruses can be replicated in the infected host cells. Coronavirus replication involves various steps, including membrane fusion, peri-nuclear particle formation, and matrix vesicle transport to the cell membrane via the endoplasmic reticulum-Golgi-lysosome route. Recent studies have suggested that protein kinase D (PKD) plays a crucial role in regulation of vesicle formation and trafficking in the trans-Golgi network (TGN). Thus, we hypothesize that inhibiting PKD and its associated pathway could be an effective strategy to limit viral replication. Here, we report that molecular and pharmacological inhibition of PKD and its substrate phosphatidylinositol-4 kinase III beta (PI4KIIIβ) significantly diminishes the replication of common human coronaviruses. Specifically, we found that the PKD-silencing siRNA and the PKD inhibitor CRT0066101 have broad-spectrum antiviral activity against HCoV-OC43, HCoV-NL63, and HCoV-229E in cultured cells. Mechanistic studies revealed that the deactivation of PKD reduced the activation of PI4KIIIβ, thereby blocking the transport of viral particles in the host cells. Furthermore, the PI4KIIIβ inhibitor, BQR695, also exhibited antiviral activity against those coronaviruses. In conclusion, PKD and its substrate, PI4KIIIβ, may serve as novel antiviral targets for human coronaviruses and warrant further investigation. IMPORTANCE Human coronaviruses can lead to a range of clinical symptoms, from asymptomatic infection to severe illness and death, with a limited array of antiviral drugs available. Protein kinase D (PKD) is involved in various cellular processes, such as cell proliferation, apoptosis, and membrane fission of the Golgi apparatus. However, the specific role of PKD in the human coronavirus life cycle remains unclear. In this study, we found that PKD inhibitors effectively attenuated human coronavirus replication at the trans-Golgi network (TGN) stage in the viral life cycle. Furthermore, inhibiting PKD reduced PI4KIIIβ activation, thereby blocking viral replication in the host cells. Importantly, PI4KIIIβ inhibitors also blocked human coronavirus replication. Thus, PKD may represent a promising therapeutic target against both current circulating and future emerging coronaviruses.
Collapse
Affiliation(s)
- Huijuan Han
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Zhijun Zhao
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zheng-Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
42
|
Doyle A, Goodson BA, Kolaczkowski OM, Liu R, Jia J, Wang H, Han X, Ye C, Bradfute SB, Kell AM, Lemus MR, Pu J. Manipulation of Host Cholesterol by SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623299. [PMID: 39605369 PMCID: PMC11601339 DOI: 10.1101/2024.11.13.623299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
SARS-CoV-2 infection is associated with alterations in host lipid metabolism, including disruptions in cholesterol homeostasis. However, the specific mechanisms by which viral proteins influence cholesterol remain incompletely understood. Here, we report that SARS-CoV-2 infection induces cholesterol sequestration within lysosomes, with the viral protein ORF3a identified as the primary driver of this effect. Mechanistically, we found that ORF3a interacts directly with the HOPS complex subunit VPS39 through a hydrophobic interface formed by residues W193 and Y184. A W193A mutation in ORF3a significantly rescues cholesterol egress and corrects the mislocalization of the lysosomal cholesterol transporter NPC2, which is caused by defective trafficking of the trans-Golgi network (TGN) sorting receptor, the cation-independent mannose-6-phosphate receptor (CI-MPR). We further observed a marked reduction in bis(monoacylglycero)phosphate (BMP), a lipid essential for lysosomal cholesterol egress, in both SARS-CoV-2-infected cells and ORF3a-expressing cells, suggesting BMP reduction as an additional mechanism of SARS-CoV-2-caused cholesterol sequestration. Inhibition of lysosomal cholesterol egress using the compound U18666A significantly decreased SARS-CoV-2 infection, highlighting a potential viral strategy of manipulating lysosomal cholesterol to modulate host cell susceptibility. Our findings reveal that SARS-CoV-2 ORF3a disrupts cellular cholesterol transport by altering lysosomal protein trafficking and BMP levels, providing new insights into virus-host interactions that contribute to lipid dysregulation in infected cells.
Collapse
Affiliation(s)
- Aliza Doyle
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Baley A. Goodson
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Oralia M. Kolaczkowski
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Jingyue Jia
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Hu Wang
- Department of Medicine, UT Health San Antonio Long School of Medicine, San Antonio, Texas 78229, USA
| | - Xianlin Han
- Department of Medicine, UT Health San Antonio Long School of Medicine, San Antonio, Texas 78229, USA
| | - Chunyan Ye
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Steven B. Bradfute
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Alison M. Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Monica Rosas Lemus
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| | - Jing Pu
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
43
|
Yao M, Li M, Peng D, Wang Y, Li S, Zhang D, Yang B, Qiu HJ, Li LF. Unraveling Macrophage Polarization: Functions, Mechanisms, and "Double-Edged Sword" Roles in Host Antiviral Immune Responses. Int J Mol Sci 2024; 25:12078. [PMID: 39596148 PMCID: PMC11593441 DOI: 10.3390/ijms252212078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Numerous viruses that propagate through the respiratory tract may be initially engulfed by macrophages (Mφs) within the alveoli, where they complete their first replication cycle and subsequently infect the adjacent epithelial cells. This process can lead to significant pathological damage to tissues and organs, leading to various diseases. As essential components in host antiviral immune systems, Mφs can be polarized into pro-inflammatory M1 Mφs or anti-inflammatory M2 Mφs, a process involving multiple signaling pathways and molecular mechanisms that yield diverse phenotypic and functional features in response to various stimuli. In general, when infected by a virus, M1 macrophages secrete pro-inflammatory cytokines to play an antiviral role, while M2 macrophages play an anti-inflammatory role to promote the replication of the virus. However, recent studies have shown that some viruses may exhibit the opposite trend. Viruses have evolved various strategies to disrupt Mφ polarization for efficient replication and transmission. Notably, various factors, such as mechanical softness, the altered pH value of the endolysosomal system, and the homeostasis between M1/M2 Mφs populations, contribute to crucial events in the viral replication cycle. Here, we summarize the regulation of Mφ polarization, virus-induced alterations in Mφ polarization, and the antiviral mechanisms associated with these changes. Collectively, this review provides insights into recent advances regarding Mφ polarization in host antiviral immune responses, which will contribute to the development of precise prevention strategies as well as management approaches to disease incidence and transmission.
Collapse
Affiliation(s)
- Meng Yao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Meilin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Dingkun Peng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Yijing Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| |
Collapse
|
44
|
Dey S, Pahari P, Mukherjee S, Munro JB, Das DK. Conformational dynamics of SARS-CoV-2 Omicron spike trimers during fusion activation at single molecule resolution. Structure 2024; 32:1910-1925.e6. [PMID: 39366371 PMCID: PMC11560620 DOI: 10.1016/j.str.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/26/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron entry involves spike (S) glycoprotein-mediated fusion of viral and late endosomal membranes. Here, using single-molecule Förster resonance energy transfer (sm-FRET) imaging and biochemical measurements, we directly visualized conformational changes of individual spike trimers on the surface of SARS-CoV-2 Omicron pseudovirions during fusion activation. We observed that the S2 domain of the Omicron spike is a dynamic fusion machine. S2 reversibly interchanges between the pre-fusion conformation and two previously undescribed intermediate conformations. Acidic pH shifts the conformational equilibrium of S2 toward an intermediate conformation and promotes the membrane hemi-fusion reaction. Moreover, we captured conformational reversibility in the S2 domain, which suggests that spike can protect itself from pre-triggering. Furthermore, we determined that Ca2+ directly promotes the S2 conformational change from an intermediate conformation to post-fusion conformation. In the presence of a target membrane, low pH and Ca2+ stimulate the irreversible transition to S2 post-fusion state and promote membrane fusion.
Collapse
Affiliation(s)
- Shuvankar Dey
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Purba Pahari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Srija Mukherjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - James B Munro
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dibyendu Kumar Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India; Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
45
|
Gualdrón-López M, Ayllon-Hermida A, Cortes-Serra N, Resa-Infante P, Bech-Serra JJ, Aparici-Herraiz I, Nicolau-Fernandez M, Erkizia I, Gutierrez-Chamorro L, Marfil S, Pradenas E, Ávila Nieto C, Cucurull B, Montaner-Tarbés S, Muelas M, Sotil R, Ballana E, Urrea V, Fraile L, Montoya M, Vergara J, Segales J, Carrillo J, Izquierdo-Useros N, Blanco J, Fernandez-Becerra C, de La Torre C, Pinazo MJ, Martinez-Picado J, del Portillo HA. Proteomics of circulating extracellular vesicles reveals diverse clinical presentations of COVID-19 but fails to identify viral peptides. Front Cell Infect Microbiol 2024; 14:1442743. [PMID: 39569406 PMCID: PMC11576438 DOI: 10.3389/fcimb.2024.1442743] [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: 06/02/2024] [Accepted: 09/23/2024] [Indexed: 11/22/2024] Open
Abstract
Extracellular vesicles (EVs) released by virus-infected cells have the potential to encapsulate viral peptides, a characteristic that could facilitate vaccine development. Furthermore, plasma-derived EVs may elucidate pathological changes occurring in distal tissues during viral infections. We hypothesized that molecular characterization of EVs isolated from COVID-19 patients would reveal peptides suitable for vaccine development. Blood samples were collected from three cohorts: severe COVID-19 patients (G1), mild/asymptomatic cases (G2), and SARS-CoV-2-negative healthcare workers (G3). Samples were obtained at two time points: during the initial phase of the pandemic in early 2020 (m0) and eight months later (m8). Clinical data analysis revealed elevated inflammatory markers in G1. Notably, non-vaccinated individuals in G1 exhibited increased levels of neutralizing antibodies at m8, suggesting prolonged exposure to viral antigens. Proteomic profiling of EVs was performed using three distinct methods: immunocapture (targeting CD9), ganglioside-capture (utilizing Siglec-1) and size-exclusion chromatography (SEC). Contrary to our hypothesis, this analysis failed to identify viral peptides. These findings were subsequently validated through Western blot analysis targeting the RBD of the SARS-CoV-2 Spike protein's and comparative studies using samples from experimentally infected Syrian hamsters. Furthermore, analysis of the EV cargo revealed a diverse molecular profile, including components involved in the regulation of viral replication, systemic inflammation, antigen presentation, and stress responses. These findings underscore the potential significance of EVs in the pathogenesis and progression of COVID-19.
Collapse
Affiliation(s)
- Melisa Gualdrón-López
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Alberto Ayllon-Hermida
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Núria Cortes-Serra
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Patricia Resa-Infante
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
| | | | - Iris Aparici-Herraiz
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Marc Nicolau-Fernandez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | | | | | | | | | | | | | | | | | - Ruth Sotil
- International Health Service, Hospital Clínic, Barcelona, Spain
| | - Ester Ballana
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Lorenzo Fraile
- Agro tecnio Center, Department of Animal Science, University of Lleida, Lleida, Spain
| | - Maria Montoya
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Julia Vergara
- Unitat Mixta d’Investigació Institute of Agrifood Research and Technology UAB (IRTA-UAB) en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Spain
- Institute of Agrifood Research and Technology (IRTA) Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Spain
| | - Joaquim Segales
- Unitat Mixta d’Investigació Institute of Agrifood Research and Technology UAB (IRTA-UAB) en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Bellaterra, Spain
| | - Jorge Carrillo
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Julià Blanco
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Fernandez-Becerra
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Maria-Jesus Pinazo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Martinez-Picado
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Hernando A. del Portillo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
46
|
Peng C, Lu W, An R, Li X, Sun C, Fang Y. Resistant Starch Nanoparticles Induce Colitis through Lysosomal Exocytosis in Mice. ACS NANO 2024; 18:30749-30760. [PMID: 39442088 DOI: 10.1021/acsnano.4c10481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Resistant starch (RS) is present in various natural and processed foods as well as medications. It has garnered significant attention from both scientists and consumers due to its notable health benefits. However, there is a limited understanding of how RS particles are absorbed at the cellular level and their metabolic behavior, resulting in a lack of clarity regarding the intestinal safety implications of prolonged RS exposure. Here, we demonstrate that rice-derived RS nanoparticles (RSNs) can lead to colitis in mice by triggering lysosomal exocytosis. The research shows that RSNs enter the cells through macropinocytosis and clathrin- and caveolin-mediated endocytosis and activate TRPML1 thereafter, causing the release of lysosomal calcium ions. This, in turn, triggered the TFEB signaling pathway and thus upregulated the lysosomal exocytosis level, leading to lysosomal enzymes to be released to the intestinal lumen. As a result, a decreased number of intestinal goblet cells, diminished tight junction protein expression, and imbalanced intestinal flora in mice were observed. These damages to the intestinal barrier ultimately led to the occurrence of colitis. Our study offers important insights into the cellular bioeffects and detrimental effects on intestinal health caused by RS particles and emphasizes the need to re-evaluate the safety of long-term RS consumption.
Collapse
Affiliation(s)
- Chenglu Peng
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Lu
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ran An
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoyang Li
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cuixia Sun
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yapeng Fang
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
47
|
Kongsomros S, Boonyarattanasoonthorn T, Phongphaew W, Kasorndorkbua C, Sunyakumthorn P, Im-Erbsin R, Lugo-Roman LA, Kongratanapasert T, Paha J, Manopwisedjaroen S, Kwankhao P, Supannapan K, Ngamkhae N, Srimongkolpithak N, Vivithanaporn P, Hongeng S, Thitithanyanont A, Khemawoot P. In vivo evaluation of Andrographis paniculata and Boesenbergia rotunda extract activity against SARS-CoV-2 Delta variant in Golden Syrian hamsters: Potential herbal alternative for COVID-19 treatment. J Tradit Complement Med 2024; 14:598-610. [PMID: 39850600 PMCID: PMC11752117 DOI: 10.1016/j.jtcme.2024.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 01/25/2025] Open
Abstract
The ongoing COVID-19 pandemic has triggered extensive research, mainly focused on identifying effective therapeutic agents, specifically those targeting highly pathogenic SARS-CoV-2 variants. This study aimed to investigate the in vivo antiviral efficacy and anti-inflammatory activity of herbal extracts derived from Andrographis paniculata and Boesenbergia rotunda, using a Golden Syrian hamster model infected with Delta, a representative variant associated with severe COVID-19. Hamsters were intranasally inoculated with the SARS-CoV-2 Delta variant and orally administered either vehicle control, B. rotunda, or A. paniculata extract at a dosage of 1000 mg/kg/day. Euthanasia was conducted on days 1, 3, and 7 post-inoculation, with 4 animals per group. The results demonstrated that oral administration of A. paniculata extract significantly alleviated both lethality and infection severity compared with the vehicle control and B. rotunda extract. However, neither extract exhibited direct antiviral activity in terms of reducing viral load in the lungs. Nonetheless, A. paniculata extract treatment significantly reduced IL-6 protein levels in the lung tissue (7278 ± 868.4 pg/g tissue) compared to the control (12,495 ± 1118 pg/g tissue), indicating there was a decrease in local inflammation. This finding is evidenced by the ability of A. paniculata extract to reduce histological lesions in the lungs of infected hamsters. Furthermore, both extracts significantly decreased IL-6 and IP-10 mRNA expression in peripheral blood mononuclear cells of infected hamsters compared to the control group, suggesting systemic anti-inflammatory effects occurred. In conclusion, A. paniculata extract's potential therapeutic application for SARS-CoV-2 arises from its observed capacity to lessen inflammatory cytokine concentrations and mitigate lung pathology.
Collapse
Affiliation(s)
- Supasek Kongsomros
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| | - Tussapon Boonyarattanasoonthorn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| | - Wallaya Phongphaew
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Chaiyan Kasorndorkbua
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Piyanate Sunyakumthorn
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, 10400, Thailand
| | - Rawiwan Im-Erbsin
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, 10400, Thailand
| | - Luis A. Lugo-Roman
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, 10400, Thailand
| | - Teetat Kongratanapasert
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Jiraporn Paha
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Pakakrong Kwankhao
- Chao Phya Abhaibhubejhr Hospital Foundation, Prachinburi, 25000, Thailand
| | | | - Nittaya Ngamkhae
- Chao Phya Abhaibhubejhr Hospital Foundation, Prachinburi, 25000, Thailand
| | - Nitipol Srimongkolpithak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | | | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| |
Collapse
|
48
|
Hamdy NM, Basalious EB, El-Sisi MG, Nasr M, Kabel AM, Nossier ES, Abadi AH. Advancements in current one-size-fits-all therapies compared to future treatment innovations for better improved chemotherapeutic outcomes: a step-toward personalized medicine. Curr Med Res Opin 2024; 40:1943-1961. [PMID: 39412377 DOI: 10.1080/03007995.2024.2416985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/22/2024]
Abstract
The development of therapies followed a generalized approach for a long time, assuming that a single treatment could effectively address various patient populations. However, recent breakthroughs have revealed the limitations of this one-size-fits-all paradigm. More recently, the field of therapeutics has witnessed a shift toward other modules, including cell therapies, high molecular weight remedies, personalized medicines, and gene therapies. Such advancements in therapeutic modules have the potential to revolutionize healthcare and pave the way for medicines that are more efficient and with minimal side effects. Cell therapies have gained considerable attention in regenerative medicine. Stem cell-based therapies, for instance, hold promise for tissue repair and regeneration, with ongoing research focusing on enhancing their efficacy and safety. High molecular weight drugs like peptides and proteins emerged as promising therapeutics because of their high specificity and diverse biological functions. Engineered peptides and proteins are developed for targeted drug delivery, immunotherapy, and disease-modulation. In personalized medicine, tailored treatments to individuals based on specific genetic profiling, lifestyle, biomarkers, and disease characteristics are all implemented. Clinicians have tailored treatments to optimize outcomes and minimize adverse effects, using targeted therapies based on specific mutations, yielding remarkable results. Gene therapies have revolutionized the treatment of genetic disorders by directly targeting the underlying genetic abnormalities. Innovative techniques, such as CRISPR-Cas9 have allowed precise gene editing, opening up possibilities for curing previously incurable conditions. In conclusion, advancements in therapeutic modules have the potential to revolutionize healthcare and pave the way for medicines that are more efficient and with minimal side effects.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo, Egypt
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
| | - Emad B Basalious
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mona G El-Sisi
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo, Egypt
| | - Maha Nasr
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ahmed M Kabel
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman S Nossier
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Ashraf H Abadi
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| |
Collapse
|
49
|
Wu H, Fujioka Y, Sakaguchi S, Suzuki Y, Nakano T. Electron Tomography as a Tool to Study SARS-CoV-2 Morphology. Int J Mol Sci 2024; 25:11762. [PMID: 39519314 PMCID: PMC11547116 DOI: 10.3390/ijms252111762] [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: 09/30/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel betacoronavirus, is the causative agent of COVID-19, which has caused economic and social disruption worldwide. To date, many drugs and vaccines have been developed for the treatment and prevention of COVID-19 and have effectively controlled the global epidemic of SARS-CoV-2. However, SARS-CoV-2 is highly mutable, leading to the emergence of new variants that may counteract current therapeutic measures. Electron microscopy (EM) is a valuable technique for obtaining ultrastructural information about the intracellular process of virus replication. In particular, EM allows us to visualize the morphological and subcellular changes during virion formation, which would provide a promising avenue for the development of antiviral agents effective against new SARS-CoV-2 variants. In this review, we present our recent findings using transmission electron microscopy (TEM) combined with electron tomography (ET) to reveal the morphologically distinct types of SARS-CoV-2 particles, demonstrating that TEM and ET are valuable tools for visually understanding the maturation status of SARS-CoV-2 in infected cells. This review also discusses the application of EM analysis to the evaluation of genetically engineered RNA viruses.
Collapse
Affiliation(s)
- Hong Wu
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka 565-0871, Japan; (Y.F.); (S.S.); (T.N.)
| | | | | | - Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka 565-0871, Japan; (Y.F.); (S.S.); (T.N.)
| | | |
Collapse
|
50
|
Huang D, Li N, Dong X. Advances in mRNA vaccine research in the field of quality control. Biologicals 2024; 88:101799. [PMID: 39504797 DOI: 10.1016/j.biologicals.2024.101799] [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: 08/13/2024] [Revised: 10/12/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
In recent years, innovative research and development of mRNA vaccines have made remarkable achievements, especially in the context of pandemic infectious diseases such as the COVID-19 virus, and the need for rapid vaccine development has further fueled the rapid growth of this field. Nevertheless, there are still gaps in our understanding of the working mechanism of mRNA vaccines and their long-term safety, efficacy, and quality control. This article summarizes the development background and production process of mRNA vaccines, outlines existing reference guidelines, quality control projects, and testing methods at home and abroad, and also summarizes the difficulties and future prospects in research and development and quality control. It provides a reference for developing guidelines for mRNA vaccine production, quality control, and preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Daomiao Huang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Na Li
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai, 200444, China; Suzhou Innovation Center of Shanghai University, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|