1
|
Dang VB, Alsherbiny MA, Lin R, Gao Y, Li C, Bhuyan DJ. Impact of a Functional Dairy Powder and Its Primary Component on the Growth of Pathogenic and Probiotic Gut Bacteria and Human Coronavirus 229E. Int J Mol Sci 2024; 25:9353. [PMID: 39273301 PMCID: PMC11394815 DOI: 10.3390/ijms25179353] [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: 07/16/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Milk boasts an array of potent bioactive compounds, such as lactoferrin (Lf), immunoglobulins, and functional proteins, all delivering substantial therapeutic benefits. In this study, Immune Powder (a functional dairy formulation) and its primary component called Fractionated Milk Protein (FMP) containing Lf, zinc, and immunoglobulins and formulated by Ausnutria Pty Ltd. were evaluated for their potential broad-spectrum pharmacological activity. In particular, this study investigated the antibacterial (against pathogenic Escherichia coli), prebiotic (promoting Lactobacillus delbrueckii growth), anti-inflammatory (inhibition of NO production in RAW264.7 macrophages), and antiviral (against human coronavirus 229E) effects of the samples. In addition, the impact of simulated gastric digestion on the efficacy of the samples was explored. LCMS-based proteomics was implemented to unveil cellular and molecular mechanisms underlying antiviral activity. The Immune Powder demonstrated antibacterial activity against E. coli (up to 99.74 ± 11.47% inhibition), coupled with prebiotic action (10.84 ± 2.2 viability fold-change), albeit these activities diminished post-digestion (p < 0.01). The Immune Powder effectively mitigated NO production in lipopolysaccharide-stimulated RAW264.7 macrophages, with declining efficacy post-digestion (p < 0.0001). The Immune Powder showed similar antiviral activity before and after digestion (p > 0.05) with up to 3-fold improvement. Likewise, FMP exhibited antibacterial potency pre-digestion at high concentrations (95.56 ± 1.23% inhibition at 125 mg/mL) and post-digestion at lower doses (61.82 ± 5.58% inhibition at 3906.25 µg/mL). FMP also showed enhanced prebiotic activity post-digestion (p < 0.0001), NO inhibition pre-digestion, and significant antiviral activity. The proteomics study suggested that the formulation and its primary component shared similar antiviral mechanisms by inhibiting scavenger receptor binding and extracellular matrix interaction.
Collapse
Affiliation(s)
- Vu Bao Dang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia
| | | | - Ruohui Lin
- Australian Dairy Park Pty Ltd., 120 Frankston Gardens Drive, Carrum Downs, VIC 3201, Australia
- Ausnutria Pty Ltd., 25-27 Keysborough Avenue, Keysborough, VIC 3173, Australia
| | - Yumei Gao
- Ausnutria Pty Ltd., 25-27 Keysborough Avenue, Keysborough, VIC 3173, Australia
| | - Chunguang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia
| | - Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia
- School of Science, Western Sydney University, Penrith, NSW 2751, Australia
| |
Collapse
|
2
|
Atemin A, Ivanova A, Peppel W, Stamatov R, Gallegos R, Durden H, Uzunova S, Vershinin MD, Saffarian S, Stoynov SS. Kinetic Landscape of Single Virus-like Particles Highlights the Efficacy of SARS-CoV-2 Internalization. Viruses 2024; 16:1341. [PMID: 39205315 PMCID: PMC11359012 DOI: 10.3390/v16081341] [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: 06/26/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The efficiency of virus internalization into target cells is a major determinant of infectivity. SARS-CoV-2 internalization occurs via S-protein-mediated cell binding followed either by direct fusion with the plasma membrane or endocytosis and subsequent fusion with the endosomal membrane. Despite the crucial role of virus internalization, the precise kinetics of the processes involved remains elusive. We developed a pipeline, which combines live-cell microscopy and advanced image analysis, for measuring the rates of multiple internalization-associated molecular events of single SARS-CoV-2-virus-like particles (VLPs), including endosome ingression and pH change. Our live-cell imaging experiments demonstrate that only a few minutes after binding to the plasma membrane, VLPs ingress into RAP5-negative endosomes via dynamin-dependent scission. Less than two minutes later, VLP speed increases in parallel with a pH drop below 5, yet these two events are not interrelated. By co-imaging fluorescently labeled nucleocapsid proteins, we show that nucleocapsid release occurs with similar kinetics to VLP acidification. Neither Omicron mutations nor abrogation of the S protein polybasic cleavage site affected the rate of VLP internalization, indicating that they do not confer any significant advantages or disadvantages during this process. Finally, we observe that VLP internalization occurs two to three times faster in VeroE6 than in A549 cells, which may contribute to the greater susceptibility of the former cell line to SARS-CoV-2 infection. Taken together, our precise measurements of the kinetics of VLP internalization-associated processes shed light on their contribution to the effectiveness of SARS-CoV-2 propagation in cells.
Collapse
Affiliation(s)
- Aleksandar Atemin
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Aneliya Ivanova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Wiley Peppel
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Rumen Stamatov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Rodrigo Gallegos
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Haley Durden
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Sonya Uzunova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Michael D. Vershinin
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Saveez Saffarian
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Stoyno S. Stoynov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| |
Collapse
|
3
|
Hudák A, Pusztai D, Letoha A, Letoha T. Mutual Inhibition of Antithrombin III and SARS-CoV-2 Cellular Attachment to Syndecans: Implications for COVID-19 Treatment and Vaccination. Int J Mol Sci 2024; 25:7534. [PMID: 39062776 PMCID: PMC11277105 DOI: 10.3390/ijms25147534] [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: 06/18/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Antithrombin III (ATIII) is a potent endogenous anticoagulant that binds to heparan sulfate proteoglycans (HSPGs) on endothelial cells' surfaces. Among these HSPGs, syndecans (SDCs) are crucial as transmembrane receptors bridging extracellular ligands with intracellular signaling pathways. Specifically, syndecan-4 (SDC4) has been identified as a key receptor on endothelial cells for transmitting the signaling effects of ATIII. Meanwhile, SDCs have been implicated in facilitating the cellular internalization of SARS-CoV-2. Given the complex interactions between ATIII and SDC4, our study analyzed the impact of ATIII on the virus entry into host cells. While ATIII binds to all SDC isoforms, it shows the strongest affinity for SDC4. SDCs' heparan sulfate chains primarily influence ATIII's SDC attachment, although other parts might also play a role in ATIII's dominant affinity toward SDC4. ATIII significantly reduces SARS-CoV-2's cellular entry into cell lines expressing SDCs, suggesting a competitive inhibition mechanism at the SDC binding sites, particularly SDC4. Conversely, the virus or its spike protein decreases the availability of SDCs on the cell surface, reducing ATIII's cellular attachment and hence contributing to a procoagulant environment characteristic of COVID-19.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (D.P.)
| | - Dávid Pusztai
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (D.P.)
| | - Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (D.P.)
| |
Collapse
|
4
|
Shouman S, El-Kholy N, Hussien AE, El-Derby AM, Magdy S, Abou-Shanab AM, Elmehrath AO, Abdelwaly A, Helal M, El-Badri N. SARS-CoV-2-associated lymphopenia: possible mechanisms and the role of CD147. Cell Commun Signal 2024; 22:349. [PMID: 38965547 PMCID: PMC11223399 DOI: 10.1186/s12964-024-01718-3] [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: 02/24/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.
Collapse
Affiliation(s)
- Shaimaa Shouman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Nada El-Kholy
- Department of Drug Discovery, H. Lee Moffit Cancer Center& Research Institute, Tampa, FL, 33612, USA
- Cancer Chemical Biology Ph.D. Program, University of South Florida, Tampa, FL, 33620, USA
| | - Alaa E Hussien
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | | | - Ahmad Abdelwaly
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, PA, 19122, USA
| | - Mohamed Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt.
| |
Collapse
|
5
|
Fredolini C, Dodig-Crnković T, Bendes A, Dahl L, Dale M, Albrecht V, Mattsson C, Thomas CE, Torinsson Naluai Å, Gisslen M, Beck O, Roxhed N, Schwenk JM. Proteome profiling of home-sampled dried blood spots reveals proteins of SARS-CoV-2 infections. COMMUNICATIONS MEDICINE 2024; 4:55. [PMID: 38565620 PMCID: PMC10987641 DOI: 10.1038/s43856-024-00480-4] [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: 10/12/2022] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Self-sampling of dried blood spots (DBS) offers new routes to gather valuable health-related information from the general population. Yet, the utility of using deep proteome profiling from home-sampled DBS to obtain clinically relevant insights about SARS-CoV-2 infections remains largely unexplored. METHODS Our study involved 228 individuals from the general Swedish population who used a volumetric DBS sampling device and completed questionnaires at home during spring 2020 and summer 2021. Using multi-analyte COVID-19 serology, we stratified the donors by their response phenotypes, divided them into three study sets, and analyzed 276 proteins by proximity extension assays (PEA). After normalizing the data to account for variances in layman-collected samples, we investigated the association of DBS proteomes with serology and self-reported information. RESULTS Our three studies display highly consistent variance of protein levels and share associations of proteins with sex (e.g., MMP3) and age (e.g., GDF-15). Studying seropositive (IgG+) and seronegative (IgG-) donors from the first pandemic wave reveals a network of proteins reflecting immunity, inflammation, coagulation, and stress response. A comparison of the early-infection phase (IgM+IgG-) with the post-infection phase (IgM-IgG+) indicates several proteins from the respiratory system. In DBS from the later pandemic wave, we find that levels of a virus receptor on B-cells differ between seropositive (IgG+) and seronegative (IgG-) donors. CONCLUSIONS Proteome analysis of volumetric self-sampled DBS facilitates precise analysis of clinically relevant proteins, including those secreted into the circulation or found on blood cells, augmenting previous COVID-19 reports with clinical blood collections. Our population surveys support the usefulness of DBS, underscoring the role of timing the sample collection to complement clinical and precision health monitoring initiatives.
Collapse
Affiliation(s)
- Claudia Fredolini
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
- Affinity Proteomics Unit, SciLifeLab Infrastructure, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Tea Dodig-Crnković
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Annika Bendes
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Leo Dahl
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Matilda Dale
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
- Affinity Proteomics Unit, SciLifeLab Infrastructure, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Vincent Albrecht
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Cecilia Mattsson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
- Affinity Proteomics Unit, SciLifeLab Infrastructure, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Cecilia E Thomas
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Åsa Torinsson Naluai
- Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Magnus Gisslen
- Department of Infectious Diseases, The Sahlgrenska Academy at University of Gothenburg, 405 30, Gothenburg, Sweden
- Sahlgrenska University Hospital, 413 45, Gothenburg, Sweden
- Public Health Agency of Sweden, 171 65, Solna, Sweden
| | - Olof Beck
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Niclas Roxhed
- MedTechLabs, BioClinicum, Karolinska University Hospital, 171 64, Solna, Sweden.
- Department of Micro and Nanosystems, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology Stockholm, 100 44, Stockholm, Sweden.
| | - Jochen M Schwenk
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden.
| |
Collapse
|
6
|
Giunta S, Giordani C, De Luca M, Olivieri F. Long-COVID-19 autonomic dysfunction: An integrated view in the framework of inflammaging. Mech Ageing Dev 2024; 218:111915. [PMID: 38354789 DOI: 10.1016/j.mad.2024.111915] [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: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
The recently identified syndrome known as Long COVID (LC) is characterized by a constellation of debilitating conditions that impair both physical and cognitive functions, thus reducing the quality of life and increasing the risk of developing the most common age-related diseases. These conditions are linked to the presence of symptoms of autonomic dysfunction, in association with low cortisol levels, suggestive of reduced hypothalamic-pituitary-adrenal (HPA) axis activity, and with increased pro-inflammatory condition. Alterations of dopamine and serotonin neurotransmitter levels were also recently observed in LC. Interestingly, at least some of the proposed mechanisms of LC development overlap with mechanisms of Autonomic Nervous System (ANS) imbalance, previously detailed in the framework of the aging process. ANS imbalance is characterized by a proinflammatory sympathetic overdrive, and a concomitant decreased anti-inflammatory vagal parasympathetic activity, associated with reduced anti-inflammatory effects of the HPA axis and cholinergic anti-inflammatory pathway (CAP). These neuro-immune-endocrine system imbalanced activities fuel the vicious circle of chronic inflammation, i.e. inflammaging. Here, we refine our original hypothesis that ANS dysfunction fuels inflammaging and propose that biomarkers of ANS imbalance could also be considered biomarkers of inflammaging, recognized as the main risk factor for developing age-related diseases and the sequelae of viral infections, i.e. LC.
Collapse
Affiliation(s)
- Sergio Giunta
- Casa di Cura Prof. Nobili (Gruppo Garofalo (GHC) Castiglione dei Pepoli -Bologna), Italy
| | - Chiara Giordani
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy.
| | - Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fabiola Olivieri
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy; Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
7
|
Tomasicchio M, Jaumdally S, Wilson L, Kotze A, Semple L, Meier S, Pooran A, Esmail A, Pillay K, Roberts R, Kriel R, Meldau R, Oelofse S, Mandviwala C, Burns J, Londt R, Davids M, van der Merwe C, Roomaney A, Kühn L, Perumal T, Scott AJ, Hale MJ, Baillie V, Mahtab S, Williamson C, Joseph R, Sigal A, Joubert I, Piercy J, Thomson D, Fredericks DL, Miller MGA, Nunes MC, Madhi SA, Dheda K. SARS-CoV-2 Viral Replication Persists in the Human Lung for Several Weeks after Symptom Onset. Am J Respir Crit Care Med 2024; 209:840-851. [PMID: 38226855 PMCID: PMC10995573 DOI: 10.1164/rccm.202308-1438oc] [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: 08/17/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024] Open
Abstract
Rationale: In the upper respiratory tract, replicating (culturable) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is recoverable for ∼4-8 days after symptom onset, but there is a paucity of data about the frequency and duration of replicating virus in the lower respiratory tract (i.e., the human lung).Objectives: We undertook lung tissue sampling (needle biopsy) shortly after death in 42 mechanically ventilated decedents during the Beta and Delta waves. An independent group of 18 ambulatory patients served as a control group.Methods: Lung biopsy cores from decedents underwent viral culture, histopathological analysis, electron microscopy, transcriptomic profiling, and immunohistochemistry.Measurements and Main Results: Thirty-eight percent (16 of 42) of mechanically ventilated decedents had culturable virus in the lung for a median of 15 days (persisting for up to 4 wk) after symptom onset. Lung viral culture positivity was not associated with comorbidities or steroid use. Delta but not Beta variant lung culture positivity was associated with accelerated death and secondary bacterial infection (P < 0.05). Nasopharyngeal culture was negative in 23.1% (6 of 26) of decedents despite lung culture positivity. This hitherto undescribed biophenotype of lung-specific persisting viral replication was associated with an enhanced transcriptomic pulmonary proinflammatory response but with concurrent viral culture positivity.Conclusions: Concurrent rather than sequential active viral replication continues to drive a heightened proinflammatory response in the human lung beyond the second week of illness and was associated with variant-specific increased mortality and morbidity. These findings have potential implications for the design of interventional strategies and clinical management of patients with severe coronavirus disease (COVID-19).
Collapse
Affiliation(s)
- Michele Tomasicchio
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Shameem Jaumdally
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Lindsay Wilson
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Andrea Kotze
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Lynn Semple
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Stuart Meier
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Aliasgar Esmail
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Komala Pillay
- Division of Anatomical Pathology, Department of Pathology, and
| | - Riyaadh Roberts
- Division of Anatomical Pathology, Department of Pathology, and
| | - Raymond Kriel
- Division of Anatomical Pathology, Department of Pathology, and
| | - Richard Meldau
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Suzette Oelofse
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Carley Mandviwala
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Jessica Burns
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Rolanda Londt
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Malika Davids
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Charnay van der Merwe
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Aqeedah Roomaney
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Louié Kühn
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Tahlia Perumal
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Alex J. Scott
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | | | - Vicky Baillie
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sana Mahtab
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa; and
| | - Ivan Joubert
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - Jenna Piercy
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - David Thomson
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - David L. Fredericks
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - Malcolm G. A. Miller
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - Marta C. Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre of Excellence in Respiratory Pathogens, Hospices Civils de Lyon and Centre International de Recherche en Infectiologie, Équipe Santé Publique, Épidémiologie et Écologie Évolutive des Maladies Infectieuses, Inserm U1111, CNRS UMR5308, ENS de Lyon, Université Claude Bernard – Lyon 1, Lyon, France
| | - Shabir A. Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
- Institute of Infectious Disease and Molecular Medicine
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
8
|
Gaurav R. SARS-CoV-2 Lurking and Lingering in the Depths of Lungs. Am J Respir Crit Care Med 2024; 209:779-780. [PMID: 38324719 PMCID: PMC10995574 DOI: 10.1164/rccm.202401-0067ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/07/2024] [Indexed: 02/09/2024] Open
Affiliation(s)
- Rohit Gaurav
- Biomedical Research Novartis Cambridge, Massachusetts
| |
Collapse
|
9
|
Lu J, Ren J, Liu J, Lu M, Cui Y, Liao Y, Zhou Y, Gao Y, Tang F, Wang J, Wang S, Wen L, Song L. High-resolution single-cell transcriptomic survey of cardiomyocytes from patients with hypertrophic cardiomyopathy. Cell Prolif 2024; 57:e13557. [PMID: 37766635 PMCID: PMC10905351 DOI: 10.1111/cpr.13557] [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: 06/02/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited cardiovascular disease, which can cause heart failure and lead to death. In this study, we performed high-resolution single-cell RNA-sequencing of 2115 individual cardiomyocytes obtained from HCM patients and normal controls. Signature up- and down-regulated genes in HCM were identified by integrative analysis across 37 patients and 41 controls from our data and published human single-cell and single-nucleus RNA-seq datasets, which were further classified into gene modules by single-cell co-expression analysis. Using our high-resolution dataset, we also investigated the heterogeneity among individual cardiomyocytes and revealed five distinct clusters within HCM cardiomyocytes. Interestingly, we showed that some extracellular matrix (ECM) genes were up-regulated in the HCM cardiomyocytes, suggesting that they play a role in cardiac remodelling. Taken together, our study comprehensively profiled the transcriptomic programs of HCM cardiomyocytes and provided insights into molecular mechanisms underlying the pathogenesis of HCM.
Collapse
Affiliation(s)
- Jiansen Lu
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
| | - Jie Ren
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary StudiesPeking UniversityBeijingChina
| | - Jie Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Minjie Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yueli Cui
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
| | - Yuhan Liao
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
| | - Yuan Zhou
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
| | - Yun Gao
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
| | - Fuchou Tang
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary StudiesPeking UniversityBeijingChina
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shuiyun Wang
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lu Wen
- College of Life Sciences, Biomedical Pioneering Innovation CenterMinistry of Education Key Laboratory of Cell Proliferation and DifferentiationBeijingChina
- Beijing Advanced Innovation Center for Genomics, College of Life SciencesPeking UniversityBeijingChina
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Cardiomyopathy ward, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
10
|
Harriott NC, Ryan AL. Proteomic profiling identifies biomarkers of COVID-19 severity. Heliyon 2024; 10:e23320. [PMID: 38163173 PMCID: PMC10755324 DOI: 10.1016/j.heliyon.2023.e23320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
SARS-CoV-2 infection remains a major public health concern, particularly for the aged and those individuals with co-morbidities at risk for developing severe COVID-19. Understanding the pathogenesis and biomarkers associated with responses to SARS-CoV-2 infection remain critical components in developing effective therapeutic approaches, especially in cases of severe and long-COVID-19. In this study blood plasma protein expression was compared in subjects with mild, moderate, and severe COVID-19 disease. Evaluation of an inflammatory protein panel confirms upregulation of proteins including TNFβ, IL-6, IL-8, IL-12, already associated with severe cytokine storm and progression to severe COVID-19. Importantly, we identify several proteins not yet associated with COVID-19 disease, including mesothelin (MSLN), that are expressed at significantly higher levels in severe COVID-19 subjects. In addition, we find a subset of markers associated with T-cell and dendritic cell responses to viral infection that are significantly higher in mild cases and decrease in expression as severity of COVID-19 increases, suggesting that an immediate and effective activation of T-cells is critical in modulating disease progression. Together, our findings identify new targets for further investigation as therapeutic approaches for the treatment of SARS-CoV-2 infection and prevention of complications of severe COVID-19.
Collapse
Affiliation(s)
- Noa C. Harriott
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| |
Collapse
|
11
|
Letoha A, Hudák A, Letoha T. Exploring the Syndecan-Mediated Cellular Internalization of the SARS-CoV-2 Omicron Variant. Int J Mol Sci 2023; 24:14140. [PMID: 37762442 PMCID: PMC10531417 DOI: 10.3390/ijms241814140] [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: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
SARS-CoV-2 variants evolve to rely more on heparan sulfate (HS) for viral attachment and subsequent infection. In our earlier work, we demonstrated that the Delta variant's spike protein binds more strongly to HS compared to WT SARS-CoV-2, leading to enhanced cell internalization via syndecans (SDCs), a family of transmembrane HS proteoglycans (HSPGs) facilitating the cellular entry of the original strain. Using our previously established ACE2- or SDC-overexpressing cellular models, we now compare the ACE2- and SDC-dependent cellular uptake of heat-inactivated WT SARS-CoV-2 with the Delta and Omicron variants. Internalization studies with inactivated virus particles showed that ACE2 overexpression could not compensate for the loss of HS in Omicron's internalization, suggesting that this variant primarily uses HSPGs to enter cells. Although SDCs increased the internalization of all three viruses, subtle differences could be detected between their SDC isoform preferences. The Delta variant particularly benefitted from SDC1, 2, and 4 overexpression for cellular entry, while SDC4 had the most prominent effect on Omicron internalization. The SDC4 knockdown (KD) in Calu-3 cells reduced the cellular uptake of all three viruses, but the inhibition was the most pronounced for Omicron. The polyanionic heparin also hindered the cellular internalization of all three viruses with a dominant inhibitory effect on Omicron. Omicron's predominant HSPG affinity, combined with its preference for the universally expressed SDC4, might account for its efficient transmission yet reduced pathogenicity.
Collapse
Affiliation(s)
- Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | | | | |
Collapse
|
12
|
Chen F, Ke Q, Wei W, Cui L, Wang Y. Apolipoprotein E and viral infection: Risks and Mechanisms. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:529-542. [PMID: 37588688 PMCID: PMC10425688 DOI: 10.1016/j.omtn.2023.07.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Apolipoprotein E (ApoE) is a multifunctional protein critical for lipid metabolism and cholesterol homeostasis. In addition to being a well known genetic determinant of both neurodegenerative and cardiovascular diseases, ApoE is frequently involved in various viral infection-related diseases. Human ApoE protein is functionally polymorphic with three isoforms, namely, ApoE2, ApoE3, and ApoE4, with markedly altered protein structures and functions. ApoE4 is associated with increased susceptibility to infection with herpes simplex virus type-1 and HIV. Conversely, ApoE4 protects against hepatitis C virus and hepatitis B virus infection. With the outbreak of coronavirus disease 2019, ApoE4 has been shown to determine the incidence and progression of severe acute respiratory syndrome coronavirus 2 infection. These findings clearly indicate the critical role of ApoE in viral infection. Furthermore, ApoE polymorphism has various or even opposite effects in these infection processes, which are partly related to the structural features that distinguish the different ApoE statuses. In the current review, we summarize the emerging relationship between ApoE and viral infection, discuss the potential mechanisms, and identify future directions that may help to advance our understanding of the link between ApoE and viral infection.
Collapse
Affiliation(s)
- Feng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Qiongwei Ke
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Wenyan Wei
- Department of Gerontology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| |
Collapse
|
13
|
Lv B, Huang S, Huang H, Niu N, Liu J. Endothelial Glycocalyx Injury in SARS-CoV-2 Infection: Molecular Mechanisms and Potential Targeted Therapy. Mediators Inflamm 2023; 2023:6685251. [PMID: 37674786 PMCID: PMC10480029 DOI: 10.1155/2023/6685251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 07/05/2023] [Accepted: 08/17/2023] [Indexed: 09/08/2023] Open
Abstract
This review aims at summarizing state-of-the-art knowledge on glycocalyx and SARS-CoV-2. The endothelial glycocalyx is a dynamic grid overlying the surface of the endothelial cell (EC) lumen and consists of membrane-bound proteoglycans and glycoproteins. The role of glycocalyx has been determined in the regulation of EC permeability, adhesion, and coagulation. SARS-CoV-2 is an enveloped, single-stranded RNA virus belonging to β-coronavirus that causes the outbreak and the pandemic of COVID-19. Through the respiratory tract, SARS-CoV-2 enters blood circulation and interacts with ECs possessing angiotensin-converting enzyme 2 (ACE2). Intact glycolyx prevents SARS-CoV-2 invasion of ECs. When the glycocalyx is incomplete, virus spike protein of SARS-CoV-2 binds with ACE2 and enters ECs for replication. In addition, cytokine storm targets glycocalyx, leading to subsequent coagulation disorder. Therefore, it is intriguing to develop a novel treatment for SARS-CoV-2 infection through the maintenance of the integrity of glycocalyx. This review aims to summarize state-of-the-art knowledge of glycocalyx and its potential function in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Bingxuan Lv
- The Second Hospital of Shandong University, Shandong University, 247 Beiyuan Street, Jinan 250033, China
| | - Shengshi Huang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China
| | - Hong Huang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China
| | - Na Niu
- Department of Pediatrics, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwu Road, Jinan 250021, China
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan 250014, China
| |
Collapse
|
14
|
Zhu Z, Ling X, Zhou H, Xie J. Syndecan-4 is the key proteoglycan involved in mediating sepsis-associated lung injury. Heliyon 2023; 9:e18600. [PMID: 37576224 PMCID: PMC10413080 DOI: 10.1016/j.heliyon.2023.e18600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023] Open
Abstract
Vascular endothelial cell dysfunction involving syndecan (SDC) proteoglycans contributes to acute sepsis-associated lung injury (ALI), but the exact SDC isoform involved is unclear. We aimed to clarify which SDCs are involved in ALI. A relevant gene expression dataset (GSE5883) was analysed for differentially expressed genes (DEGs) between lipopolysaccharide (LPS)-treated and control lung endothelial cells and for SDC isoform expression. Bioinformatic analyses to predict DEG function were conducted using R language, Gene Ontology, and the Kyoto Encyclopedia of Genes and Genomes. SDC2 and SDC4 expression profiles were examined under inflammatory conditions in human lung vascular endothelial cell and mouse sepsis-associated ALI models. Transcription factors regulating SDC2/4 were predicted to indirectly assess SDC involvement in septic inflammation. Of the DEGs, 224 and 102 genes were up- and downregulated, respectively. Functional analysis indicated that DEGs were involved in modulating receptor ligand and signalling receptor activator activities, cytokine receptor binding, responses to LPS and molecules of bacterial origin, regulation of cell adhesion, tumour necrosis factor signalling, and other functions. DEGs were also enriched for cytoplasmic ribonucleoprotein granules, transcription regulator complexes, and membrane raft cellular components. SDC4 gene expression was 4.5-fold higher in the LPS group than in the control group, while SDC2 levels were similar in both groups. SDC4 mRNA and protein expression was markedly upregulated in response to inflammatory injury, and SDC4 downregulation severely exacerbated inflammatory responses in both in vivo and in vitro models. Overall, our data demonstrate that SDC4, rather than SDC2, is involved in LPS-induced sepsis-associated ALI.
Collapse
Affiliation(s)
- Zhipeng Zhu
- Department of Anaesthesiology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, 314000, China
| | - Xiaoyan Ling
- Department of Outpatient Nursing, The Second Affiliated Hospital of Jiaxing University, Zhejiang, 314000, China
| | - Hongmei Zhou
- Department of Anaesthesiology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, 314000, China
| | - Junran Xie
- Department of Anaesthesiology, Run Xia Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, 314000, China
| |
Collapse
|
15
|
Ricard-Blum S, Couchman JR. Conformations, interactions and functions of intrinsically disordered syndecans. Biochem Soc Trans 2023:BST20221085. [PMID: 37334846 DOI: 10.1042/bst20221085] [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: 02/19/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans present on most mammalian cell surfaces. They have a long evolutionary history, a single syndecan gene being expressed in bilaterian invertebrates. Syndecans have attracted interest because of their potential roles in development and disease, including vascular diseases, inflammation and various cancers. Recent structural data is providing important insights into their functions, which are complex, involving both intrinsic signaling through cytoplasmic binding partners and co-operative mechanisms where syndecans form a signaling nexus with other receptors such as integrins and tyrosine kinase growth factor receptors. While the cytoplasmic domain of syndecan-4 has a well-defined dimeric structure, the syndecan ectodomains are intrinsically disordered, which is linked to a capacity to interact with multiple partners. However, it remains to fully establish the impact of glycanation and partner proteins on syndecan core protein conformations. Genetic models indicate that a conserved property of syndecans links the cytoskeleton to calcium channels of the transient receptor potential class, compatible with roles as mechanosensors. In turn, syndecans influence actin cytoskeleton organization to impact motility, adhesion and the extracellular matrix environment. Syndecan clustering with other cell surface receptors into signaling microdomains has relevance to tissue differentiation in development, for example in stem cells, but also in disease where syndecan expression can be markedly up-regulated. Since syndecans have potential as diagnostic and prognostic markers as well as possible targets in some forms of cancer, it remains important to unravel structure/function relationships in the four mammalian syndecans.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- ICBMS, UMR 5246 CNRS, Universite Claude Bernard Lyon 1, F-69622 Villeurbanne, France
| | - John R Couchman
- Biotech Research & Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
16
|
Kummarapurugu AB, Hawkridge AM, Ma J, Osei S, Martin RK, Zheng S, Voynow JA. Neutrophil Elastase decreases SARS-CoV-2 Spike protein binding to human bronchial epithelia by clipping ACE-2 ectodomian from the epithelial surface. J Biol Chem 2023:104820. [PMID: 37187291 PMCID: PMC10181948 DOI: 10.1016/j.jbc.2023.104820] [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: 03/28/2022] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
Patients with cystic fibrosis (CF) have decreased severity of SARS-CoV-2 infections, but the underlying cause is unknown. Patients with CF have high levels of neutrophil elastase (NE) in the airway. We examined whether respiratory epithelial angiotensin converting enzyme 2 (ACE-2), the receptor for the SARS-CoV-2 spike protein, is a proteolytic target of NE. Soluble ACE-2 (sACE-2) levels were quantified by ELISA in airway secretions and serum from patients with and without CF, and the association between sACE-2 levels and NE activity levels was evaluated in CF sputum. We determined that NE activity was directly correlated with increased ACE-2 in CF sputum. Additionally, primary human bronchial epithelial (HBE) cells, exposed to NE or control vehicle, were evaluated by western analysis for the release of cleaved ACE-2 ectodomain fragment into conditioned media, and by flow cytometry for the loss of cell surface ACE-2, its impact on SARS-CoV-2 spike protein binding. We found that NE treatment released ACE-2 ectodomain fragment from HBE and decreased spike protein binding to HBE. Furthermore, we performed NE treatment of recombinant ACE-2-Fc tagged protein in vitro to assess whether NE was sufficient to cleave recombinant ACE-2-Fc protein. Proteomic analysis identified, specific NE cleavage sites in the ACE-2 ectodomain that would result in loss of the putative N-terminal spike binding domain. Collectively, data support that NE plays a disruptive role in SARS-CoV-2 infection by catalyzing ACE-2 ectodomain shedding from the airway epithelia. This mechanism may reduce SARS-CoV-2 virus binding to respiratory epithelial cells and decrease severity of COVID19 infection.
Collapse
Affiliation(s)
- Apparao B Kummarapurugu
- Department of Pediatric Pulmonary Medicine, Children's Hospital of Richmond at Virginia Commonwealth University.
| | - Adam M Hawkridge
- School of Pharmacy at Virginia Commonwealth University, Richmond VA
| | - Jonathan Ma
- Department of Pediatric Pulmonary Medicine, Children's Hospital of Richmond at Virginia Commonwealth University
| | | | - Rebecca K Martin
- Department of Microbiology and Immunology at Virginia Commonwealth University, Richmond VA
| | - Shuo Zheng
- Department of Pediatric Pulmonary Medicine, Children's Hospital of Richmond at Virginia Commonwealth University
| | - Judith A Voynow
- Department of Pediatric Pulmonary Medicine, Children's Hospital of Richmond at Virginia Commonwealth University
| |
Collapse
|
17
|
Syndecan-4 Mediates the Cellular Entry of Adeno-Associated Virus 9. Int J Mol Sci 2023; 24:ijms24043141. [PMID: 36834552 PMCID: PMC9963952 DOI: 10.3390/ijms24043141] [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: 01/07/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Due to their low pathogenicity, immunogenicity, and long-term gene expression, adeno-associated virus (AAV) vectors emerged as safe and efficient gene delivery tools, over-coming setbacks experienced with other viral gene delivery systems in early gene therapy trials. Among AAVs, AAV9 can translocate through the blood-brain barrier (BBB), making it a promising gene delivery tool for transducing the central nervous system (CNS) via systemic administration. Recent reports on the shortcomings of AAV9-mediated gene delivery into the CNS require reviewing the molecular base of AAV9 cellular biology. A more detailed understanding of AAV9's cellular entry would eradicate current hurdles and enable more efficient AAV9-based gene therapy approaches. Syndecans, the transmembrane family of heparan-sulfate proteoglycans, facilitate the cellular uptake of various viruses and drug delivery systems. Utilizing human cell lines and syndecan-specific cellular assays, we assessed the involvement of syndecans in AAV9's cellular entry. The ubiquitously expressed isoform, syndecan-4 proved its superiority in facilitating AAV9 internalization among syndecans. Introducing syndecan-4 into poorly transducible cell lines enabled robust AAV9-dependent gene transduction, while its knockdown reduced AAV9's cellular entry. Attachment of AAV9 to syndecan-4 is mediated not just by the polyanionic heparan-sulfate chains but also by the cell-binding domain of the extracellular syndecan-4 core protein. Co-immunoprecipitation assays and affinity proteomics also confirmed the role of syndecan-4 in the cellular entry of AAV9. Overall, our findings highlight the universally expressed syndecan-4 as a significant contributor to the cellular internalization of AAV9 and provide a molecular-based, rational explanation for the low gene delivery potential of AAV9 into the CNS.
Collapse
|
18
|
Stocker N, Radzikowska U, Wawrzyniak P, Tan G, Huang M, Ding M, Akdis CA, Sokolowska M. Regulation of angiotensin-converting enzyme 2 isoforms by type 2 inflammation and viral infection in human airway epithelium. Mucosal Immunol 2023; 16:5-16. [PMID: 36642382 PMCID: PMC9836991 DOI: 10.1016/j.mucimm.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/06/2022] [Indexed: 01/15/2023]
Abstract
SARS-CoV-2 enters human cells through its main receptor, angiotensin-converting enzyme 2 (ACE2), which constitutes a limiting factor of infection. Recent findings demonstrating novel ACE2 isoforms implicate that this receptor is regulated in a more complex way than previously anticipated. However, it remains unknown how various inflammatory conditions influence the abundance of these ACE2 variants. Hence, we studied expression of ACE2 messenger RNA (mRNA) and protein isoforms, together with its glycosylation and spatial localization in primary human airway epithelium upon allergic inflammation and viral infection. We found that interleukin-13, the main type 2 cytokine, decreased expression of long ACE2 mRNA and reduced glycosylation of full-length ACE2 protein via alteration of N-linked glycosylation process, limiting its availability on the apical side of ciliated cells. House dust mite allergen did not affect the expression of ACE2. Rhinovirus infection increased short ACE2 mRNA, but it did not influence its protein expression. In addition, by screening other SARS-CoV-2 related host molecules, we found that interleukin-13 and rhinovirus significantly regulated mRNA, but not protein of transmembrane serine protease 2 and neuropilin 1. Regulation of ACE2 and other host proteins was comparable in healthy and asthmatic epithelium, underlining the lack of intrinsic differences but dependence on the inflammatory milieu in the airways.
Collapse
Affiliation(s)
- Nino Stocker
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Department of Gastroenterology and Hepatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| |
Collapse
|
19
|
Cerezo-Magaña M, Bång-Rudenstam A, Belting M. Proteoglycans: a common portal for SARS-CoV-2 and extracellular vesicle uptake. Am J Physiol Cell Physiol 2023; 324:C76-C84. [PMID: 36458979 PMCID: PMC9799137 DOI: 10.1152/ajpcell.00453.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
As structural components of the glycocalyx, heparan sulfate proteoglycans (HSPGs) are involved in multiple pathophysiological processes at the apex of cell signaling cascades, and as endocytosis receptors for particle structures, such as lipoproteins, extracellular vesicles, and enveloped viruses, including SARS-CoV-2. Given their diversity and complex biogenesis regulation, HSPGs remain understudied. Here we compile some of the latest studies focusing on HSPGs as internalizing receptors of extracellular vesicles ("endogenous virus") and SARS-CoV-2 lipid-enclosed particles and highlight similarities in their biophysical and structural characteristics. Specifically, the similarities in their biogenesis, size, and lipid composition may explain a common dependence on HSPGs for efficient cell-surface attachment and uptake. We further discuss the relative complexity of extracellular vesicle composition and the viral mechanisms that evolve towards increased infectivity that complicate therapeutic strategies addressing blockade of their uptake.
Collapse
Affiliation(s)
| | - Anna Bång-Rudenstam
- 1Department of Clinical Sciences Lund, Oncology, Lund University, Lund, Sweden
| | - Mattias Belting
- 1Department of Clinical Sciences Lund, Oncology, Lund University, Lund, Sweden,2Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden,3Department of Hematology, Oncology, and Radiophysics, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
20
|
Jasim SA, Mahdi RS, Bokov DO, Najm MAA, Sobirova GN, Bafoyeva ZO, Taifi A, Alkadir OKA, Mustafa YF, Mirzaei R, Karampoor S. The deciphering of the immune cells and marker signature in COVID-19 pathogenesis: An update. J Med Virol 2022; 94:5128-5148. [PMID: 35835586 PMCID: PMC9350195 DOI: 10.1002/jmv.28000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/28/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022]
Abstract
The precise interaction between the immune system and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical in deciphering the pathogenesis of coronavirus disease 2019 (COVID-19) and is also vital for developing novel therapeutic tools, including monoclonal antibodies, antivirals drugs, and vaccines. Viral infections need innate and adaptive immune reactions since the various immune components, such as neutrophils, macrophages, CD4+ T, CD8+ T, and B lymphocytes, play different roles in various infections. Consequently, the characterization of innate and adaptive immune reactions toward SARS-CoV-2 is crucial for defining the pathogenicity of COVID-19. In this study, we explain what is currently understood concerning the conventional immune reactions to SARS-CoV-2 infection to shed light on the protective and pathogenic role of immune response in this case. Also, in particular, we investigate the in-depth roles of other immune mediators, including neutrophil elastase, serum amyloid A, and syndecan, in the immunopathogenesis of COVID-19.
Collapse
Affiliation(s)
| | - Roaa Salih Mahdi
- Department of Pathology, College of MedicineUniversity of BabylonHillaIraq
| | - Dmitry Olegovich Bokov
- Institute of PharmacySechenov First Moscow State Medical UniversityMoscowRussian Federation,Laboratory of Food ChemistryFederal Research Center of Nutrition, Biotechnology and Food SafetyMoscowRussian Federation
| | - Mazin A. A. Najm
- Pharmaceutical Chemistry Department, College of PharmacyAl‐Ayen UniversityThi‐QarIraq
| | - Guzal N. Sobirova
- Department of Rehabilitation, Folk Medicine and Physical EducationTashkent Medical AcademyTashkentUzbekistan
| | - Zarnigor O. Bafoyeva
- Department of Rehabilitation, Folk Medicine and Physical EducationTashkent Medical AcademyTashkentUzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of PharmacyUniversity of MosulMosulIraq
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
21
|
Heinl ES, Lorenz S, Schmidt B, Nasser M Laqtom N, Mazzulli JR, Francelle L, Yu TW, Greenberg B, Storch S, Tegtmeier I, Othmen H, Maurer K, Steinfurth M, Witzgall R, Milenkovic V, Wetzel CH, Reichold M. CLN7/MFSD8 may be an important factor for SARS-CoV-2 cell entry. iScience 2022; 25:105082. [PMID: 36093380 PMCID: PMC9444308 DOI: 10.1016/j.isci.2022.105082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 05/12/2022] [Accepted: 08/31/2022] [Indexed: 11/12/2022] Open
Abstract
The SARS-CoV-2 virus has triggered a worldwide pandemic. According to the BioGrid database, CLN7 (MFSD8) is thought to interact with several viral proteins. The aim of this work was to investigate a possible involvement of CLN7 in the infection process. Experiments on a CLN7-deficient HEK293T cell line exhibited a 90% reduced viral load compared to wild-type cells. This observation may be linked to the finding that CLN7 ko cells have a significantly reduced GM1 content in their cell membrane. GM1 is found highly enriched in lipid rafts, which are thought to play an important role in SARS-CoV-2 infection. In contrast, overexpression of CLN7 led to an increase in viral load. This study provides evidence that CLN7 is involved in SARS-CoV-2 infection. This makes it a potential pharmacological target for drug development against COVID-19. Furthermore, it provides insights into the physiological function of CLN7 where still only little is known about.
Collapse
Affiliation(s)
- Elena-Sofia Heinl
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Sebastian Lorenz
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Barbara Schmidt
- Institute of Clinical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | - Nouf Nasser M Laqtom
- Departments of Chemical Engineering and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Joseph R. Mazzulli
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Laetitia Francelle
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Timothy W. Yu
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Benjamin Greenberg
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Stephan Storch
- Children’s Hospital Biochemistry, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Ines Tegtmeier
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Helga Othmen
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
- Institute for Molecular and Cellular Anatomy, University Regensburg, 93053 Regensburg, Germany
| | - Katja Maurer
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Malin Steinfurth
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University Regensburg, 93053 Regensburg, Germany
| | - Vladimir Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Christian H. Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany
| | - Markus Reichold
- Medical Cell Biology, University Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
22
|
Lindqvist R, Benz C, Sereikaite V, Maassen L, Laursen L, Jemth P, Strømgaard K, Ivarsson Y, Överby AK. A Syntenin Inhibitor Blocks Endosomal Entry of SARS-CoV-2 and a Panel of RNA Viruses. Viruses 2022; 14:v14102202. [PMID: 36298757 PMCID: PMC9610207 DOI: 10.3390/v14102202] [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: 08/18/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
Viruses are dependent on host factors in order to efficiently establish an infection and replicate. Targeting the interactions of such host factors provides an attractive strategy to develop novel antivirals. Syntenin is a protein known to regulate the architecture of cellular membranes by its involvement in protein trafficking and has previously been shown to be important for human papilloma virus (HPV) infection. Here, we show that a highly potent and metabolically stable peptide inhibitor that binds to the PDZ1 domain of syntenin inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection by blocking the endosomal entry of the virus. Furthermore, we found that the inhibitor also hampered chikungunya infection and strongly reduced flavivirus infection, which is completely dependent on receptor-mediated endocytosis for their entry. In conclusion, we have identified a novel broad spectrum antiviral inhibitor that efficiently targets a broad range of RNA viruses.
Collapse
Affiliation(s)
- Richard Lindqvist
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90186 Umeå, Sweden
| | - Caroline Benz
- Department of Chemistry—BMC, Uppsala University, Box 576, Husargatan 3, 75123 Uppsala, Sweden
| | - Vita Sereikaite
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Lars Maassen
- Department of Chemistry—BMC, Uppsala University, Box 576, Husargatan 3, 75123 Uppsala, Sweden
| | - Louise Laursen
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, Husargatan 3, 75123 Uppsala, Sweden
| | - Per Jemth
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, Husargatan 3, 75123 Uppsala, Sweden
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ylva Ivarsson
- Department of Chemistry—BMC, Uppsala University, Box 576, Husargatan 3, 75123 Uppsala, Sweden
- Correspondence: (Y.I.); (A.K.Ö.)
| | - Anna K. Överby
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90186 Umeå, Sweden
- Correspondence: (Y.I.); (A.K.Ö.)
| |
Collapse
|
23
|
Crozier TW, Greenwood EJ, Williamson JC, Guo W, Porter LM, Gabaev I, Teixeira-Silva A, Grice GL, Wickenhagen A, Stanton RJ, Wang ECY, Wilson SJ, Matheson NJ, Nathan JA, McCaughan F, Lehner PJ. Quantitative proteomic analysis of SARS-CoV-2 infection of primary human airway ciliated cells and lung epithelial cells demonstrates the effectiveness of SARS-CoV-2 innate immune evasion. Wellcome Open Res 2022; 7:224. [PMID: 36483314 PMCID: PMC9706147 DOI: 10.12688/wellcomeopenres.17946.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 02/02/2023] Open
Abstract
Background: Quantitative proteomics is able to provide a comprehensive, unbiased description of changes to cells caused by viral infection, but interpretation may be complicated by differential changes in infected and uninfected 'bystander' cells, or the use of non-physiological cellular models. Methods: In this paper, we use fluorescence-activated cell sorting (FACS) and quantitative proteomics to analyse cell-autonomous changes caused by authentic SARS-CoV-2 infection of respiratory epithelial cells, the main target of viral infection in vivo. First, we determine the relative abundance of proteins in primary human airway epithelial cells differentiated at the air-liquid interface (basal, secretory and ciliated cells). Next, we specifically characterise changes caused by SARS-CoV-2 infection of ciliated cells. Finally, we compare temporal proteomic changes in infected and uninfected 'bystander' Calu-3 lung epithelial cells and compare infection with B.29 and B.1.1.7 (Alpha) variants. Results: Amongst 5,709 quantified proteins in primary human airway ciliated cells, the abundance of 226 changed significantly in the presence of SARS-CoV-2 infection (q <0.05 and >1.5-fold). Notably, viral replication proceeded without inducing a type-I interferon response. Amongst 6,996 quantified proteins in Calu-3 cells, the abundance of 645 proteins changed significantly in the presence of SARS-CoV-2 infection (q < 0.05 and > 1.5-fold). In contrast to the primary cell model, a clear type I interferon (IFN) response was observed. Nonetheless, induction of IFN-inducible proteins was markedly attenuated in infected cells, compared with uninfected 'bystander' cells. Infection with B.29 and B.1.1.7 (Alpha) variants gave similar results. Conclusions: Taken together, our data provide a detailed proteomic map of changes in SARS-CoV-2-infected respiratory epithelial cells in two widely used, physiologically relevant models of infection. As well as identifying dysregulated cellular proteins and processes, the effectiveness of strategies employed by SARS-CoV-2 to avoid the type I IFN response is illustrated in both models.
Collapse
Affiliation(s)
- Thomas W.M. Crozier
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Edward J.D. Greenwood
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - James C. Williamson
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Wenrui Guo
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Linsey M. Porter
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ildar Gabaev
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Ana Teixeira-Silva
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Guinevere L. Grice
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Arthur Wickenhagen
- MRC - University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, UK
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Eddie C. Y. Wang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sam J. Wilson
- MRC - University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, UK
| | - Nicholas J. Matheson
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
- NHS Blood and Transplant, Cambridge, CB2 0PT, UK
| | - James A. Nathan
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Frank McCaughan
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Paul J. Lehner
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| |
Collapse
|
24
|
Hudák A, Morgan G, Bacovsky J, Patai R, Polgár TF, Letoha A, Pettko-Szandtner A, Vizler C, Szilák L, Letoha T. Biodistribution and Cellular Internalization of Inactivated SARS-CoV-2 in Wild-Type Mice. Int J Mol Sci 2022; 23:ijms23147609. [PMID: 35886958 PMCID: PMC9316427 DOI: 10.3390/ijms23147609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Despite the growing list of identified SARS-CoV-2 receptors, the human angiotensin-converting enzyme 2 (ACE2) is still viewed as the main cell entry receptor mediating SARS-CoV-2 internalization. It has been reported that wild-type mice, like other rodent species of the Muridae family, cannot be infected with SARS-CoV-2 due to differences in their ACE2 receptors. On the other hand, the consensus heparin-binding motif of SARS-CoV-2’s spike protein, PRRAR, enables the attachment to rodent heparan sulfate proteoglycans (HSPGs), including syndecans, a transmembrane HSPG family with a well-established role in clathrin- and caveolin-independent endocytosis. As mammalian syndecans possess a relatively conserved structure, we analyzed the cellular uptake of inactivated SARS-CoV-2 particles in in vitro and in vivo mice models. Cellular studies revealed efficient uptake into murine cell lines with established syndecan-4 expression. After intravenous administration, inactivated SARS-CoV-2 was taken up by several organs in vivo and could also be detected in the brain. Internalized by various tissues, inactivated SARS-CoV-2 raised tissue TNF-α levels, especially in the heart, reflecting the onset of inflammation. Our studies on in vitro and in vivo mice models thus shed light on unknown details of SARS-CoV-2 internalization and help broaden the understanding of the molecular interactions of SARS-CoV-2.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
| | | | | | - Roland Patai
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (R.P.); (T.F.P.)
| | - Tamás F. Polgár
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (R.P.); (T.F.P.)
- Theoretical Medicine Doctoral School, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary;
| | | | - Csaba Vizler
- Institute of Biochemistry, Biological Research Centre, H-6726 Szeged, Hungary;
| | - László Szilák
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
| | - Tamás Letoha
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
- Correspondence: ; Tel.: +36-30-2577393
| |
Collapse
|
25
|
Faraji SN, Raee MJ, Hashemi SMA, Daryabor G, Tabrizi R, Dashti FS, Behboudi E, Heidarnejad K, Nowrouzi-Sohrabi P, Hatam G. Human interaction targets of SARS-COV-2 spike protein: A systematic review. EUR J INFLAMM 2022. [PMCID: PMC9160582 DOI: 10.1177/1721727x221095382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objectives: The development of effective targeted therapy and drug-design approaches against the SARS-CoV-2 is a universal health priority. Therefore, it is important to assess possible therapeutic strategies against SARS-CoV-2 via its most interaction targets. The present study aimed to perform a systematic review on clinical and experimental investigations regarding SARS-COV-2 interaction targets for human cell entry. Methods: A systematic search using relevant MeSH terms and keywords was performed in PubMed, Scopus, Embase, and Web of Science (ISI) databases up to July 2021. Two reviewers independently assessed the eligibility of the studies, extracted the data, and evaluated the methodological quality of the included studies. Additionally, a narrative synthesis was done as a qualitative method for data gathering and synthesis of each outcome measure. Results: A total of 5610 studies were identified, and 128 articles were included in the systematic review. Based on the results, spike antigen was the only interaction protein from SARS-CoV-2. However, the interaction proteins from humans varied including different spike receptors and several cleavage enzymes. The most common interactions of the spike protein of SARS-CoV-2 for cell entry were ACE2 (entry receptor) and TMPRSS2 (for spike priming). A lot of published studies have mainly focused on the ACE2 receptor followed by the TMPRSS family and furin. Based on the results, ACE2 polymorphisms as well as spike RBD mutations affected the SARS-CoV-2 binding affinity. Conclusion: The included studies shed more light on SARS-CoV-2 cellular entry mechanisms and detailed interactions, which could enhance the understanding of SARS-CoV-2 pathogenesis and the development of new and comprehensive therapeutic approaches.
Collapse
Affiliation(s)
- Seyed Nooreddin Faraji
- School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohamad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohamad Ali Hashemi
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gholamreza Daryabor
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Tabrizi
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Fateme Sadat Dashti
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Emad Behboudi
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Kamran Heidarnejad
- Recombinant Antibody Laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Peyman Nowrouzi-Sohrabi
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Hatam
- Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Cheudjeu A. The SARS-CoV-2 Entry Inhibition Mechanisms of Serine Protease Inhibitors, OM-85, Heparin and Soluble HS Might Be Linked to HS Attachment Sites. Molecules 2022; 27:molecules27061947. [PMID: 35335311 PMCID: PMC8954261 DOI: 10.3390/molecules27061947] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
This article discusses the importance of D-xylose for fighting viruses (especially SARS-CoV-2) that use core proteins as receptors at the cell surface, by providing additional supporting facts that these viruses probably bind at HS/CS attachment sites (i.e., the hydroxyl groups of Ser/Thr residues of the core proteins intended to receive the D-xylose molecules to initiate the HS/CS chains). Essentially, the additional supporting facts, are: some anterior studies on the binding sites of exogenous heparin and soluble HS on the core proteins, the inhibition of the viral entry by pre-incubation of cells with heparin, and additionally, corroborating studies about the mechanism leading to type 2 diabetes during viral infection. We then discuss the mechanism by which serine protease inhibitors inhibit SARS-CoV-2 entry. The biosynthesis of heparan sulfate (HS), chondroitin sulfate (CS), dermatan sulfate (DS), and heparin (Hep) is initiated not only by D-xylose derived from uridine diphosphate (UDP)-xylose, but also bioactive D-xylose molecules, even in situations where cells were previously treated with GAG inhibitors. This property of D-xylose shown by previous anterior studies helped in the explanation of the mechanism leading to type 2 diabetes during SARS-CoV-2 infection. This explanation is completed here by a preliminary estimation of xyloside GAGs (HS/CS/DS/Hep) in the body, and with other previous studies helping to corroborate the mechanism by which the D-xylose exhibits its antiglycaemic properties and the mechanism leading to type 2 diabetes during SARS-CoV-2 infection. This paper also discusses the confirmatory studies of regarding the correlation between D-xylose and COVID-19 severity.
Collapse
|
27
|
Lima FB, Bezerra KC, Nascimento JCR, Meneses GC, Oriá RB. Risk Factors for Severe COVID-19 and Hepatitis C Infections: The Dual Role of Apolipoprotein E4. Front Immunol 2022; 13:721793. [PMID: 35359998 PMCID: PMC8962614 DOI: 10.3389/fimmu.2022.721793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 02/21/2022] [Indexed: 01/14/2023] Open
Affiliation(s)
- Felipe B. Lima
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
- Division of Anesthesiology, Hospital Geral de Fortaleza, Fortaleza, Brazil
| | - Karine C. Bezerra
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - José Carlos R. Nascimento
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
- Division of Anesthesiology, Hospital Geral de Fortaleza, Fortaleza, Brazil
| | - Gdayllon C. Meneses
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
- *Correspondence: Reinaldo B. Oriá,
| |
Collapse
|
28
|
Hudák A, Veres G, Letoha A, Szilák L, Letoha T. Syndecan-4 Is a Key Facilitator of the SARS-CoV-2 Delta Variant's Superior Transmission. Int J Mol Sci 2022; 23:796. [PMID: 35054983 PMCID: PMC8775852 DOI: 10.3390/ijms23020796] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
Emerging SARS-CoV-2 variants pose threats to vaccination campaigns against COVID-19. Being more transmissible than the original virus, the SARS-CoV-2 B.1.617 lineage, named the Delta variant, swept through the world in 2021. The mutations in the Delta's spike protein shift the protein towards a net positive electrostatic potential. To understand the key molecular drivers of the Delta infection, we investigate the cellular uptake of the Delta spike protein and Delta spike-bearing SARS-CoV-2 pseudoviruses. Specific in vitro modification of ACE2 and syndecan expression enabled us to demonstrate that syndecan-4, the syndecan isoform abundant in the lung, enhances the transmission of the Delta variant by attaching its mutated spike glycoprotein and facilitating its cellular entry. Compared to the wild-type spike, the Delta one shows a higher affinity towards heparan sulfate proteoglycans than towards ACE2. In addition to attachment to the polyanionic heparan sulfate chains, the Delta spike's molecular interactions with syndecan-4 also involve syndecan-4's cell-binding domain that mediates cell-to-cell adhesion. Regardless of the complexity of these interactions, exogenously added heparin blocks Delta's cellular entry as efficiently as syndecan-4 knockdown. Therefore, a profound understanding of the molecular mechanisms underlying Delta infections enables the development of molecularly targeted yet simple strategies to reduce the Delta variant's spread.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (G.V.); (L.S.)
| | - Gábor Veres
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (G.V.); (L.S.)
| | - Annamária Letoha
- Albert Szent-Györgyi Clinical Center, Department of Medicine, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - László Szilák
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (G.V.); (L.S.)
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (G.V.); (L.S.)
| |
Collapse
|
29
|
Palacios-Rápalo SN, De Jesús-González LA, Cordero-Rivera CD, Farfan-Morales CN, Osuna-Ramos JF, Martínez-Mier G, Quistián-Galván J, Muñoz-Pérez A, Bernal-Dolores V, del Ángel RM, Reyes-Ruiz JM. Cholesterol-Rich Lipid Rafts as Platforms for SARS-CoV-2 Entry. Front Immunol 2021; 12:796855. [PMID: 34975904 PMCID: PMC8719300 DOI: 10.3389/fimmu.2021.796855] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Since its appearance, the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2), the causal agent of Coronavirus Disease 2019 (COVID-19), represents a global problem for human health that involves the host lipid homeostasis. Regarding, lipid rafts are functional membrane microdomains with highly and tightly packed lipid molecules. These regions enriched in sphingolipids and cholesterol recruit and concentrate several receptors and molecules involved in pathogen recognition and cellular signaling. Cholesterol-rich lipid rafts have multiple functions for viral replication; however, their role in SARS-CoV-2 infection remains unclear. In this review, we discussed the novel evidence on the cholesterol-rich lipid rafts as a platform for SARS-CoV-2 entry, where receptors such as the angiotensin-converting enzyme-2 (ACE-2), heparan sulfate proteoglycans (HSPGs), human Toll-like receptors (TLRs), transmembrane serine proteases (TMPRSS), CD-147 and HDL-scavenger receptor B type 1 (SR-B1) are recruited for their interaction with the viral spike protein. FDA-approved drugs such as statins, metformin, hydroxychloroquine, and cyclodextrins (methyl-β-cyclodextrin) can disrupt cholesterol-rich lipid rafts to regulate key molecules in the immune signaling pathways triggered by SARS-CoV-2 infection. Taken together, better knowledge on cholesterol-rich lipid rafts in the SARS-CoV-2-host interactions will provide valuable insights into pathogenesis and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Gustavo Martínez-Mier
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Judith Quistián-Galván
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Armando Muñoz-Pérez
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Víctor Bernal-Dolores
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Rosa María del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| |
Collapse
|
30
|
Ogata H, Mori M, Jingushi Y, Matsuzaki H, Katahira K, Ishimatsu A, Enokizu-Ogawa A, Taguchi K, Moriwaki A, Yoshida M. Impact of visceral fat on the prognosis of coronavirus disease 2019: an observational cohort study. BMC Infect Dis 2021; 21:1240. [PMID: 34893021 PMCID: PMC8660963 DOI: 10.1186/s12879-021-06958-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/07/2021] [Indexed: 01/08/2023] Open
Abstract
Background Clarification of the risk factors for coronavirus disease 2019 (COVID-19) severity is strongly warranted for global health. Recent studies have indicated that elevated body mass index (BMI) is associated with unfavorable progression of COVID-19. This is assumed to be due to excessive deposition of visceral adipose tissue (VAT); however, the evidence investigating the association between intra-abdominal fat and COVID-19 prognosis is sparse. We therefore investigated whether measuring the amount of intra-abdominal fat is useful to predict the prognosis of COVID-19. Methods The present study enrolled 53 consecutive cases of COVID-19 patients aged ≥ 20 years with chest computed tomography (CT) scans. The VAT area, total adipose tissue (TAT) area, and VAT/TAT ratio were estimated using axial CT images at the level of the upper pole of the right kidney. Severe COVID-19 was defined as death or acute respiratory failure demanding oxygen at ≥ 6 L per minute, a high-flow nasal cannula, or mechanical ventilation. The association of VAT/TAT with the incidence of progression to a severe state was estimated as a hazard ratio (HR) using Cox regression analysis. To compare the prediction ability for COVID-19 disease progression between BMI and VAT/TAT, the area under the receiver operating characteristic curve (AUC) of each was assessed. Results A total of 15 cases (28.3% of the whole study subjects) progressed to severe stages. The incidence of developing severe COVID-19 increased significantly with VAT/TAT (HR per 1% increase = 1.040 (95% CI 1.008–1.074), P = 0.01). After adjustment for potential confounders, the positive association of VAT/TAT with COVID-19 aggravation remained significant (multivariable-adjusted HR = 1.055 (95% CI 1.000–1.112) per 1% increase, P = 0.049). The predictive ability of VAT/TAT for COVID-19 becoming severe was significantly better than that of BMI (AUC of 0.73 for VAT/TAT and 0.50 for BMI; P = 0.0495 for the difference). Conclusions A higher ratio of VAT/TAT was an independent risk factor for disease progression among COVID-19 patients. VAT/TAT was superior to BMI in predicting COVID-19 morbidity. COVID-19 patients with high VAT/TAT levels should be carefully observed as high-risk individuals for morbidity and mortality. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06958-z.
Collapse
Affiliation(s)
- Hiroaki Ogata
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan.
| | - Masahiro Mori
- Department of Radiology, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Yujiro Jingushi
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Hiroshi Matsuzaki
- Department of Pediatrics, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Katsuyuki Katahira
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Akiko Ishimatsu
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Aimi Enokizu-Ogawa
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Kazuhito Taguchi
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Atsushi Moriwaki
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| | - Makoto Yoshida
- Department of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, 4-39-1 Yakatabaru, Minami-ku, Fukuoka, 811-1394, Japan
| |
Collapse
|
31
|
A spatial multi-scale fluorescence microscopy toolbox discloses entry checkpoints of SARS-CoV-2 variants in Vero E6 cells. Comput Struct Biotechnol J 2021; 19:6140-6156. [PMID: 34745450 PMCID: PMC8562013 DOI: 10.1016/j.csbj.2021.10.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/12/2023] Open
Abstract
We exploited a multi-scale microscopy imaging toolbox to address some major issues related to SARS-CoV-2 interactions with host cells. Our approach harnesses both conventional and super-resolution fluorescence microscopy and easily matches the spatial scale of single-virus/cell checkpoints. After its validation through the characterization of infected cells and virus morphology, we leveraged this toolbox to reveal subtle issues related to the entry phase of SARS-CoV-2 variants in Vero E6 cells. Our results show that in Vero E6 cells the B.1.1.7 strain (aka Alpha Variant of Concern) is associated with much faster kinetics of endocytic uptake compared to its ancestor B.1.177. Given the cell-entry scenario dominated by the endosomal “late pathway”, the faster internalization of B.1.1.7 could be directly related to the N501Y mutation in the S protein, which is known to strengthen the binding of Spike receptor binding domain with ACE2. Remarkably, we also directly observed the central role of clathrin as a mediator of endocytosis in the late pathway of entry. In keeping with the clathrin-mediated endocytosis, we highlighted the non-raft membrane localization of ACE2. Overall, we believe that our fluorescence microscopy-based approach represents a fertile strategy to investigate the molecular features of SARS-CoV-2 interactions with cells.
Collapse
|
32
|
McClelland RD, Culp TN, Marchant DJ. Imaging Flow Cytometry and Confocal Immunofluorescence Microscopy of Virus-Host Cell Interactions. Front Cell Infect Microbiol 2021; 11:749039. [PMID: 34712624 PMCID: PMC8546218 DOI: 10.3389/fcimb.2021.749039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/27/2021] [Indexed: 11/25/2022] Open
Abstract
Viruses are diverse pathogens that use host factors to enter cells and cause disease. Imaging the entry and replication phases of viruses and their interactions with host factors is key to fully understanding viral infections. This review will discuss how confocal microscopy and imaging flow cytometry are used to investigate virus entry and replication mechanisms in fixed and live cells. Quantification of viral images and the use of cryo-electron microscopy to gather structural information of viruses is also explored. Using imaging to understand how viruses replicate and interact with host factors, we gain insight into cellular processes and identify novel targets to develop antiviral therapeutics and vaccines.
Collapse
Affiliation(s)
- Ryley D McClelland
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, Katz Center for Health Research, University of Alberta, Edmonton, AB, Canada
| | - Tyce N Culp
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, Katz Center for Health Research, University of Alberta, Edmonton, AB, Canada
| | - David J Marchant
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, Katz Center for Health Research, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
33
|
Mese K, Bunz O, Volkwein W, Vemulapalli SPB, Zhang W, Schellhorn S, Heenemann K, Rueckner A, Sing A, Vahlenkamp TW, Severing AL, Gao J, Aydin M, Jung D, Bachmann HS, Zänker KS, Busch U, Baiker A, Griesinger C, Ehrhardt A. Enhanced Antiviral Function of Magnesium Chloride-Modified Heparin on a Broad Spectrum of Viruses. Int J Mol Sci 2021; 22:10075. [PMID: 34576237 PMCID: PMC8466540 DOI: 10.3390/ijms221810075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Previous studies reported on the broad-spectrum antiviral function of heparin. Here we investigated the antiviral function of magnesium-modified heparin and found that modified heparin displayed a significantly enhanced antiviral function against human adenovirus (HAdV) in immortalized and primary cells. Nuclear magnetic resonance analyses revealed a conformational change of heparin when complexed with magnesium. To broadly explore this discovery, we tested the antiviral function of modified heparin against herpes simplex virus type 1 (HSV-1) and found that the replication of HSV-1 was even further decreased compared to aciclovir. Moreover, we investigated the antiviral effect against the new severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and measured a 55-fold decreased viral load in the supernatant of infected cells associated with a 38-fold decrease in virus growth. The advantage of our modified heparin is an increased antiviral effect compared to regular heparin.
Collapse
Affiliation(s)
- Kemal Mese
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
| | - Oskar Bunz
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
- Department of Prosthodontics, School of Dentistry, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
| | - Wolfram Volkwein
- Bavarian Health and Food Safety Authority (LGL), 85764 Oberschleissheim, Germany; (W.V.); (A.S.); (U.B.)
| | - Sahithya P. B. Vemulapalli
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany;
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
| | - Sebastian Schellhorn
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
| | - Kristin Heenemann
- Center for Infectious Diseases, Institute of Virology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany; (K.H.); (A.R.); (T.W.V.)
| | - Antje Rueckner
- Center for Infectious Diseases, Institute of Virology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany; (K.H.); (A.R.); (T.W.V.)
| | - Andreas Sing
- Bavarian Health and Food Safety Authority (LGL), 85764 Oberschleissheim, Germany; (W.V.); (A.S.); (U.B.)
| | - Thomas W. Vahlenkamp
- Center for Infectious Diseases, Institute of Virology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany; (K.H.); (A.R.); (T.W.V.)
| | - Anna-Lena Severing
- Centre for Biomedical Education and Research (ZBAF), Institute for Translational Wound Research, Witten/Herdecke University, 58453 Witten, Germany;
| | - Jian Gao
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
| | - Malik Aydin
- Center for Child and Adolescent Medicine, Center for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
| | - Dominik Jung
- Centre for Biomedical Education and Research, Institute of Pharmacology and Toxicology, Witten/Herdecke University, 58453 Witten, Germany; (D.J.); (H.S.B.)
| | - Hagen S. Bachmann
- Centre for Biomedical Education and Research, Institute of Pharmacology and Toxicology, Witten/Herdecke University, 58453 Witten, Germany; (D.J.); (H.S.B.)
| | - Kurt S. Zänker
- Center for Biomedical Education and Research (ZBAF), Institute of Immunology, Witten/Herdecke University, 58453 Witten, Germany;
| | - Ulrich Busch
- Bavarian Health and Food Safety Authority (LGL), 85764 Oberschleissheim, Germany; (W.V.); (A.S.); (U.B.)
| | - Armin Baiker
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
- Bavarian Health and Food Safety Authority (LGL), 85764 Oberschleissheim, Germany; (W.V.); (A.S.); (U.B.)
| | - Christian Griesinger
- Centre for Biomedical Education and Research, Institute of Pharmacology and Toxicology, Witten/Herdecke University, 58453 Witten, Germany; (D.J.); (H.S.B.)
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany; (K.M.); (O.B.); (W.Z.); (S.S.); (J.G.); (A.B.)
| |
Collapse
|
34
|
Sarabipour S, Mac Gabhann F. Targeting neuropilins as a viable SARS-CoV-2 treatment. FEBS J 2021; 288:5122-5129. [PMID: 34185437 PMCID: PMC8420456 DOI: 10.1111/febs.16096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/01/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022]
Abstract
The SARS-CoV-2 pandemic has significantly impacted global health. Research on viral mechanisms, highly effective vaccines, and other therapies is in progress. Neuropilins have recently been identified as host cell receptors enabling viral fusion. Here, we provide context to neuropilin's tissue-specific role in infection and the potential impact of NRP-based therapeutics. We conclude that the central roles of neuropilins in vascular, neural, and other pathways may render it a less suitable target for treating SARS-CoV-2 than agents that target its binding partner, the viral spike protein.
Collapse
Affiliation(s)
- Sarvenaz Sarabipour
- Department of Biomedical EngineeringInstitute for Computational MedicineJohns Hopkins UniversityBaltimoreMDUSA
| | - Feilim Mac Gabhann
- Department of Biomedical EngineeringInstitute for Computational MedicineJohns Hopkins UniversityBaltimoreMDUSA
| |
Collapse
|
35
|
Hudák A, Jósvay K, Domonkos I, Letoha A, Szilák L, Letoha T. The Interplay of Apoes with Syndecans in Influencing Key Cellular Events of Amyloid Pathology. Int J Mol Sci 2021; 22:ijms22137070. [PMID: 34209175 PMCID: PMC8268055 DOI: 10.3390/ijms22137070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 01/06/2023] Open
Abstract
Apolipoprotein E (ApoE) isoforms exert intricate effects on cellular physiology beyond lipid transport and metabolism. ApoEs influence the onset of Alzheimer’s disease (AD) in an isoform-dependent manner: ApoE4 increases AD risk, while ApoE2 decreases it. Previously we demonstrated that syndecans, a transmembrane proteoglycan family with increased expression in AD, trigger the aggregation and modulate the cellular uptake of amyloid beta (Aβ). Utilizing our previously established syndecan-overexpressing cellular assays, we now explore how the interplay of ApoEs with syndecans contributes to key events, namely uptake and aggregation, in Aβ pathology. The interaction of ApoEs with syndecans indicates isoform-specific characteristics arising beyond the frequently studied ApoE–heparan sulfate interactions. Syndecans, and among them the neuronal syndecan-3, increased the cellular uptake of ApoEs, especially ApoE2 and ApoE3, while ApoEs exerted opposing effects on syndecan-3-mediated Aβ uptake and aggregation. ApoE2 increased the cellular internalization of monomeric Aβ, hence preventing its extracellular aggregation, while ApoE4 decreased it, thus helping the buildup of extracellular plaques. The contrary effects of ApoE2 and ApoE4 remained once Aβ aggregated: while ApoE2 reduced the uptake of Aβ aggregates, ApoE4 facilitated it. Fibrillation studies also revealed ApoE4′s tendency to form fibrillar aggregates. Our results uncover yet unknown details of ApoE cellular biology and deepen our molecular understanding of the ApoE-dependent mechanism of Aβ pathology.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
| | - Katalin Jósvay
- Institute of Biochemistry, Biological Research Centre, H-6726 Szeged, Hungary;
| | - Ildikó Domonkos
- Institute of Plant Biology, Biological Research Centre, H-6726 Szeged, Hungary;
| | - Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - László Szilák
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
| | - Tamás Letoha
- Pharmacoidea Ltd., H-6726 Szeged, Hungary; (A.H.); (L.S.)
- Correspondence: ; Tel.: +36-(30)-2577393
| |
Collapse
|
36
|
Heparan Sulfate Proteoglycans in Viral Infection and Treatment: A Special Focus on SARS-CoV-2. Int J Mol Sci 2021; 22:ijms22126574. [PMID: 34207476 PMCID: PMC8235362 DOI: 10.3390/ijms22126574] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/27/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) encompass a group of glycoproteins composed of unbranched negatively charged heparan sulfate (HS) chains covalently attached to a core protein. The complex HSPG biosynthetic machinery generates an extraordinary structural variety of HS chains that enable them to bind a plethora of ligands, including growth factors, morphogens, cytokines, chemokines, enzymes, matrix proteins, and bacterial and viral pathogens. These interactions translate into key regulatory activity of HSPGs on a wide range of cellular processes such as receptor activation and signaling, cytoskeleton assembly, extracellular matrix remodeling, endocytosis, cell-cell crosstalk, and others. Due to their ubiquitous expression within tissues and their large functional repertoire, HSPGs are involved in many physiopathological processes; thus, they have emerged as valuable targets for the therapy of many human diseases. Among their functions, HSPGs assist many viruses in invading host cells at various steps of their life cycle. Viruses utilize HSPGs for the attachment to the host cell, internalization, intracellular trafficking, egress, and spread. Recently, HSPG involvement in the pathogenesis of SARS-CoV-2 infection has been established. Here, we summarize the current knowledge on the molecular mechanisms underlying HSPG/SARS-CoV-2 interaction and downstream effects, and we provide an overview of the HSPG-based therapeutic strategies that could be used to combat such a fearsome virus.
Collapse
|