1
|
Cao X, Huang L, Tang M, Liang Y, Liu X, Hou H, Liang S. Antibiotics daptomycin interacts with S protein of SARS-CoV-2 to promote cell invasion of Omicron (B1.1.529) pseudovirus. Virulence 2024; 15:2339703. [PMID: 38576396 PMCID: PMC11057663 DOI: 10.1080/21505594.2024.2339703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/03/2024] [Indexed: 04/06/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has posed enormous challenges to global public health. The use of antibiotics has greatly increased during the SARS-CoV-2 epidemic owing to the presence of bacterial co-infection and secondary bacterial infections. The antibiotics daptomycin (DAP) is widely used in the treatment of infectious diseases caused by gram-positive bacteria owing to its highly efficient antibacterial activity. It is pivotal to study the antibiotics usage options for patients of coronavirus infectious disease (COVID-19) with pneumonia those need admission to receive antibiotics treatment for bacterial co-infection in managing COVID-19 disease. Herein, we have revealed the interactions of DAP with the S protein of SARS-CoV-2 and the variant Omicron (B1.1.529) using the molecular docking approach and Omicron (B1.1.529) pseudovirus (PsV) mimic invasion. Molecular docking analysis shows that DAP has a certain degree of binding ability to the S protein of SARS-CoV-2 and several derived virus variants, and co-incubation of 1-100 μM DAP with cells promotes the entry of the PsV into human angiotensin-converting enzyme 2 (hACE2)-expressing HEK-293T cells (HEK-293T-hACE2), and this effect is related to the concentration of extracellular calcium ions (Ca2+). The PsV invasion rate in the HEK-293T-hACE2 cells concurrently with DAP incubation was 1.7 times of PsV infection alone. In general, our findings demonstrate that DAP promotes the infection of PsV into cells, which provides certain reference of antibiotics selection and usage optimization for clinicians to treat bacterial coinfection or secondary infection during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xu Cao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Min Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinpeng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huijin Hou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Hoste ACR, Smeralda W, Cugnet A, Brostaux Y, Deleu M, Garigliany M, Jacques P. The structure of lipopeptides impacts their antiviral activity and mode of action against SARS-CoV-2 in vitro. Appl Environ Microbiol 2024:e0103624. [PMID: 39445780 DOI: 10.1128/aem.01036-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Microbial lipopeptides are synthesized by nonribosomal peptide synthetases and are composed of a hydrophobic fatty acid chain and a hydrophilic peptide moiety. These structurally diverse amphiphilic molecules can interact with biological membranes and possess various biological activities, including antiviral properties. This study aimed to evaluate the cytotoxicity and antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) of 15 diverse lipopeptides to understand their structure-activity relationships. Non-ionic lipopeptides were generally more cytotoxic than charged ones, with cationic lipopeptides being less cytotoxic than anionic and non-ionic variants. At 100 µg/mL, six lipopeptides reduced SARS-CoV-2 RNA to undetectable levels in infected Vero E6 cells, while six others achieved a 2.5- to 4.1-log reduction, and three had no significant effect. Surfactin, white line-inducing principle (WLIP), fengycin, and caspofungin emerged as the most promising anti-SARS-CoV-2 agents. Detailed analysis revealed that these four lipopeptides affected various stages of the viral life cycle involving the viral envelope. Surfactin and WLIP significantly reduced viral RNA levels in replication assays, comparable to neutralizing serum. Surfactin uniquely inhibited viral budding, while fengycin impacted viral binding after pre-infection treatment of the cells. Caspofungin demonstrated a lower antiviral effect compared to the others. Key structural traits of lipopeptides influencing their cytotoxic and antiviral activities were identified. Lipopeptides with a high number of amino acids, especially charged (preferentially anionic) amino acids, showed potent anti-SARS-CoV-2 activity. This research paves the way for designing new lipopeptides with low cytotoxicity and high antiviral efficacy, potentially leading to effective treatments. IMPORTANCE This study advances our understanding of how lipopeptides, which are molecules mostly produced by bacteria, with both fat and protein components, can be used to fight viruses like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). By analyzing 15 different lipopeptides, researchers identified key structural features that make some of these molecules particularly effective at reducing viral levels while being less harmful to cells. Specifically, lipopeptides with certain charged amino acids were found to have the strongest antiviral effects. This research lays the groundwork for developing new antiviral treatments that are both potent against viruses and safe for human cells, offering hope for better therapeutic options in the future.
Collapse
Affiliation(s)
- Alexis C R Hoste
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
- Veterinary Pathology, FARAH Research Centre, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Willy Smeralda
- LBMI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Aurélien Cugnet
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Yves Brostaux
- Applied Statistics, Computer Science and Modelling laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Magali Deleu
- LBMI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Mutien Garigliany
- Veterinary Pathology, FARAH Research Centre, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Philippe Jacques
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
3
|
Righi E, Dalla Vecchia I, Auerbach N, Morra M, Górska A, Sciammarella C, Lambertenghi L, Gentilotti E, Mirandola M, Tacconelli E, Sartor A. Gut Microbiome Disruption Following SARS-CoV-2: A Review. Microorganisms 2024; 12:131. [PMID: 38257958 PMCID: PMC10820238 DOI: 10.3390/microorganisms12010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
COVID-19 has been associated with having a negative impact on patients' gut microbiome during both active disease and in the post-acute phase. In acute COVID-19, rapid alteration of the gut microbiome composition was observed, showing on one side a reduction in beneficial symbionts (e.g., Roseburia, Lachnospiraceae) and on the other side an increase in opportunistic pathogens such as Enterococcus and Proteobacteria. Alpha diversity tends to decrease, especially initially with symptom onset and hospital admission. Although clinical recovery appears to align with improved gut homeostasis, this process could take several weeks, even in mild infections. Moreover, patients with COVID-19 post-acute syndrome showed changes in gut microbiome composition, with specific signatures associated with decreased respiratory function up to 12 months following acute disease. Potential treatments, especially probiotic-based therapy, are under investigation. Open questions remain on the possibility to use gut microbiome data to predict disease progression and on potential confounders that may impair result interpretation (e.g., concomitant therapies in the acute phase; reinfection, vaccines, and occurrence of novel conditions or diseases in the post-acute syndrome). Understanding the relationships between gut microbiome dynamics and disease progression may contribute to better understanding post-COVID syndrome pathogenesis or inform personalized treatment that can affect specific targets or microbiome markers.
Collapse
Affiliation(s)
- Elda Righi
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Ilaria Dalla Vecchia
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Nina Auerbach
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Matteo Morra
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Anna Górska
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Concetta Sciammarella
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Lorenza Lambertenghi
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Elisa Gentilotti
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Massimo Mirandola
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Evelina Tacconelli
- IMID Laboratory, Department of Diagnostics and Public Health, Infectious Diseases Division, University of Verona, 37134 Verona, Italy (A.G.); (C.S.); (E.T.)
| | - Assunta Sartor
- Microbiology Unit, Udine University Hospital, 33100 Udine, Italy;
| |
Collapse
|
4
|
Mikhnovets IE, Holoubek J, Panina IS, Kotouček J, Gvozdev DA, Chumakov SP, Krasilnikov MS, Zhitlov MY, Gulyak EL, Chistov AA, Nikitin TD, Korshun VA, Efremov RG, Alferova VA, Růžek D, Eyer L, Ustinov AV. Alkyl Derivatives of Perylene Photosensitizing Antivirals: Towards Understanding the Influence of Lipophilicity. Int J Mol Sci 2023; 24:16483. [PMID: 38003673 PMCID: PMC10671050 DOI: 10.3390/ijms242216483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Amphipathic perylene derivatives are broad-spectrum antivirals against enveloped viruses that act as fusion inhibitors in a light-dependent manner. The compounds target the lipid bilayer of the viral envelope using the lipophilic perylene moiety and photogenerating singlet oxygen, thereby causing damage to unsaturated lipids. Previous studies show that variation of the polar part of the molecule is important for antiviral activity. Here, we report modification of the lipophilic part of the molecule, perylene, by the introduction of 4-, 8-, and 12-carbon alkyls into position 9(10) of the perylene residue. Using Friedel-Crafts acylation and Wolff-Kishner reduction, three 3-acetyl-9(10)-alkylperylenes were synthesized from perylene and used to prepare 9 nucleoside and 12 non-nucleoside amphipathic derivatives. These compounds were characterized as fluorophores and singlet oxygen generators, as well as tested as antivirals against herpes virus-1 (HSV-1) and vesicular stomatitis virus (VSV), both known for causing superficial skin/mucosa lesions and thus serving as suitable candidates for photodynamic therapy. The results suggest that derivatives with a short alkyl chain (butyl) have strong antiviral activity, whereas the introduction of longer alkyl substituents (n = 8 and 12) to the perylenyethynyl scaffold results in a dramatic reduction of antiviral activity. This phenomenon is likely attributable to the increased lipophilicity of the compounds and their ability to form insoluble aggregates. Moreover, molecular dynamic studies revealed that alkylated perylene derivatives are predominately located closer to the middle of the bilayer compared to non-alkylated derivatives. The predicted probability of superficial positioning correlated with antiviral activity, suggesting that singlet oxygen generation is achieved in the subsurface layer of the membrane, where the perylene group is more accessible to dissolved oxygen.
Collapse
Affiliation(s)
- Igor E. Mikhnovets
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Jiří Holoubek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic (D.R.); (L.E.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Irina S. Panina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Jan Kotouček
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic;
| | - Daniil A. Gvozdev
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia;
| | - Stepan P. Chumakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Maxim S. Krasilnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Mikhail Y. Zhitlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia
| | - Evgeny L. Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Alexey A. Chistov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Timofei D. Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Roman G. Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| | - Daniel Růžek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic (D.R.); (L.E.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Luděk Eyer
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Hudcova 296/70, CZ-621 00 Brno, Czech Republic (D.R.); (L.E.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1160/31, CZ-370 05 České Budějovice, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (I.E.M.); (I.S.P.); (S.P.C.); (M.S.K.); (M.Y.Z.); (E.L.G.); (A.A.C.); (T.D.N.); (V.A.K.); (R.G.E.); (V.A.A.)
| |
Collapse
|
5
|
Ayusso GM, da Silva Sanches PR, Carvalho T, Santos IA, Martins DOS, Lima MLD, da Conceição PJP, Bittar C, Merits A, Cilli EM, Jardim ACG, Rahal P, Calmon MF. The Synthetic Peptide GA-Hecate and Its Analogs Inhibit Multiple Steps of the Chikungunya Virus Infection Cycle In Vitro. Pharmaceuticals (Basel) 2023; 16:1389. [PMID: 37895860 PMCID: PMC10610090 DOI: 10.3390/ph16101389] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/08/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Chikungunya virus (CHIKV) belongs to the Alphavirus genus and is responsible for significant outbreaks worldwide. Currently, there is no approved antiviral therapy against CHIKV. Bioactive peptides have great potential for new drug development. Here, we evaluated the antiviral activity of the synthetic peptide GA-Hecate and its analogs PSSct1905 and PSSct1910 against CHIKV infection. Initial screening showed that all three peptides inhibited the CHIKV replication cycle in baby hamster kidney fibroblast cells (BHK-21) and human hepatocarcinoma epithelial cells (Huh-7). GA-Hecate and its analog PSSct1905 were the most active, demonstrating suppression of viral infection by more than 91%. The analog PSSct1905 exhibited a protective effect in cells against CHIKV infection. We also observed that the analogs PSSct1905 and PSSct1910 affected CHIKV entry into both cell lines, inhibiting viral attachment and internalization. Finally, all tested compounds presented antiviral activity on the post-entry steps of CHIKV infection in all cells evaluated. In conclusion, this study highlights the potential of the peptide GA-Hecate and its analogs as novel anti-CHIKV compounds targeting different stages of the viral replication cycle, warranting the development of GA-Hecate-based compounds with broad antiviral activity.
Collapse
Affiliation(s)
- Gabriela Miranda Ayusso
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
| | | | - Tamara Carvalho
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
| | - Igor Andrade Santos
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Daniel Oliveira Silva Martins
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Maria Letícia Duarte Lima
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
| | - Pâmela Jóyce Previdelli da Conceição
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
| | - Cíntia Bittar
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Andres Merits
- Institute of Technology, University of Tartu, 50090 Tartu, Estonia;
| | - Eduardo Maffud Cilli
- Institute of Chemistry, São Paulo State University, Araraquara 14800-060, SP, Brazil;
| | - Ana Carolina Gomes Jardim
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Paula Rahal
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
| | - Marilia Freitas Calmon
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil; (G.M.A.); (T.C.); (D.O.S.M.); (M.L.D.L.); (P.J.P.d.C.); (C.B.); (A.C.G.J.); (P.R.)
| |
Collapse
|
6
|
Durairajan SSK, Singh AK, Saravanan UB, Namachivayam M, Radhakrishnan M, Huang JD, Dhodapkar R, Zhang H. Gastrointestinal Manifestations of SARS-CoV-2: Transmission, Pathogenesis, Immunomodulation, Microflora Dysbiosis, and Clinical Implications. Viruses 2023; 15:1231. [PMID: 37376531 DOI: 10.3390/v15061231] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023] Open
Abstract
The clinical manifestation of COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in the respiratory system of humans is widely recognized. There is increasing evidence suggesting that SARS-CoV-2 possesses the capability to invade the gastrointestinal (GI) system, leading to the manifestation of symptoms such as vomiting, diarrhea, abdominal pain, and GI lesions. These symptoms subsequently contribute to the development of gastroenteritis and inflammatory bowel disease (IBD). Nevertheless, the pathophysiological mechanisms linking these GI symptoms to SARS-CoV-2 infection remain unelucidated. During infection, SARS-CoV-2 binds to angiotensin-converting enzyme 2 and other host proteases in the GI tract during the infection, possibly causing GI symptoms by damaging the intestinal barrier and stimulating inflammatory factor production, respectively. The symptoms of COVID-19-induced GI infection and IBD include intestinal inflammation, mucosal hyperpermeability, bacterial overgrowth, dysbiosis, and changes in blood and fecal metabolomics. Deciphering the pathogenesis of COVID-19 and understanding its exacerbation may provide insights into disease prognosis and pave the way for the discovery of potential novel targets for disease prevention or treatment. Besides the usual transmission routes, SARS-CoV-2 can also be transmitted via the feces of an infected person. Hence, it is crucial to implement preventive and control measures in order to mitigate the fecal-to-oral transmission of SARS-CoV-2. Within this context, the identification and diagnosis of GI tract symptoms during these infections assume significance as they facilitate early detection of the disease and the development of targeted therapeutics. The present review discusses the receptors, pathogenesis, and transmission of SARS-CoV-2, with a particular focus on the induction of gut immune responses, the influence of gut microbes, and potential therapeutic targets against COVID-19-induced GI infection and IBD.
Collapse
Affiliation(s)
| | - Abhay Kumar Singh
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Udhaya Bharathy Saravanan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Mayurikaa Namachivayam
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Moorthi Radhakrishnan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Jian-Dong Huang
- Department of Biochemistry, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong 999077, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rahul Dhodapkar
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Government of India, Puducherry 605006, India
| | - Hongjie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| |
Collapse
|
7
|
Roshni J, Sivakumar M, Bahammam FA, Bhandi S, Patil S, Kamath M, Abusharha A, Ahmed SSSJ. New Ways to Protect the Host from SARS-CoV-2? Lung Microbiome Metabolites Inhibit STAT3 and Modulate the Immunological Network. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:237-244. [PMID: 37140561 DOI: 10.1089/omi.2023.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
COVID-19 caused by the SARS-CoV-2 infection is a systemic disease that affects multiple organs, biological pathways, and cell types. A systems biology approach would benefit the study of COVID-19 in the pandemic as well as the endemic state. Notably, patients with COVID-19 have dysbiosis of lung microbiota whose functional relevance to the host is largely unknown. We carried out a systems biology investigation of the impact of lung microbiome-derived metabolites on host immune system during COVID-19. RNAseq was performed to identify the host-specific pro- and anti-inflammatory differentially expressed genes (DEGs) in bronchial epithelium and alveolar cells during SARS-CoV-2 infection. The overlapping DEGs were harnessed to construct an immune network while their key transcriptional regulator was deciphered. We identified 68 overlapping genes from both cell types to construct the immune network, and Signal Transducer and Activator of Transcription 3 (STAT3) was found to regulate the majority of the network proteins. Furthermore, thymidine diphosphate produced from the lung microbiome had the highest affinity with STAT3 (-6.349 kcal/mol) than the known STAT3 inhibitors (n = 410), with an affinity ranging from -5.39 to 1.31 kcal/mol. In addition, the molecular dynamic studies showed distinguishable changes in the behavior of the STAT3 complex when compared with free STAT3. Overall, our results provide new observations on the importance of lung microbiome metabolites that regulate the host immune system in patients with COVID-19, and may open up new avenues for preventive medicine and therapeutics innovation.
Collapse
Affiliation(s)
- Jency Roshni
- Drug Discovery and Multi-omics Lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Mahema Sivakumar
- Drug Discovery and Multi-omics Lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Faris Ahmed Bahammam
- Fellow Rhinology and Facial Plastics, Imperial College London, London, United Kingdom
| | - Shilpa Bhandi
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, Utah, USA
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, Utah, USA
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Manjunath Kamath
- Centre for Advance Studies, Sathyabama Institute of Science and Technology, Tamil Nadu, Chennai, India
| | - Ali Abusharha
- Department of Optometry, Applied Medical Sciences College, King Saud University, Riyadh, Saudi Arabia
| | - Shiek S S J Ahmed
- Drug Discovery and Multi-omics Lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
8
|
Urata S, Takouda J, Watanabe Y, Sakaguchi M, Sakurai Y, Inahashi Y, Iwatsuki M, Yasuda J, Tanaka Y, Takeda K. Identification of surfactin as an anti-severe fever with thrombocytopenia syndrome virus multi-target compound extracted from the culture broth of marine microbes. FRONTIERS IN VIROLOGY 2023. [DOI: 10.3389/fviro.2022.1064265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne virus first identified in China in 2011 and later reported in other Asian countries. Significant efforts have been made to develop anti-SFTSV compounds; however, there are no approved vaccines or antivirals against SFTSV infections. Marine organisms provide nearly unlimited biological resources to produce therapeutic drugs for the treatment and control of disease. In this study, we aimed to identify anti-SFTSV chemical compounds from the culture broth extracts of marine microbes collected from the coasts of the Nagasaki Prefecture, Japan. Of the 80 extracts, two showed an anti-SFTSV effect. One of them, which exhibited low cell toxicity, was used for further characterization. Chemical analysis combined with the anti-SFTSV effect identified surfactin as one of the main components of the selected extract. Our study showed a proof-of-concept to identify novel antiviral compounds from marine microbes against the virus of interest. Further analysis showed that surfactin affected the integrity of the virion membrane and inhibited SFTSV infection-induced membrane fusion at low pH conditions. Furthermore, surfactin inhibits the post-entry step of viral replication in the cell, which is a novel mode of antiviral action of surfactin. These results indicate that surfactin can target multiple steps of SFTSV replication in cells.
Collapse
|
9
|
Production and characterization of lentivirus vector-based SARS-CoV-2 pseudoviruses with dual reporters: Evaluation of anti-SARS-CoV-2 viral effect of Korean Red Ginseng. J Ginseng Res 2023; 47:123-132. [PMID: 35855181 PMCID: PMC9283196 DOI: 10.1016/j.jgr.2022.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 01/09/2023] Open
Abstract
Background Pseudotyped virus systems that incorporate viral proteins have been widely employed for the rapid determination of the effectiveness and neutralizing activity of drug and vaccine candidates in biosafety level 2 facilities. We report an efficient method for producing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudovirus with dual luciferase and fluorescent protein reporters. Moreover, using the established method, we also aimed to investigate whether Korean Red Ginseng (KRG), a valuable Korean herbal medicine, can attenuate infectivity of the pseudotyped virus. Methods A pseudovirus of SARS-CoV-2 (SARS-2pv) was constructed and efficiently produced using lentivirus vector systems available in the public domain by the introduction of critical mutations in the cytoplasmic tail of the spike protein. KRG extract was dose-dependently treated to Calu-3 cells during SARS2-pv treatment to evaluate the protective activity against SARS-CoV-2. Results The use of Calu-3 cells or the expression of angiotensin-converting enzyme 2 (ACE2) in HEK293T cells enabled SARS-2pv infection of host cells. Coexpression of transmembrane protease serine subtype 2 (TMPRSS2), which is the activator of spike protein, with ACE2 dramatically elevated luciferase activity, confirming the importance of the TMPRSS2-mediated pathway during SARS-CoV-2 entry. Our pseudovirus assay also revealed that KRG elicited resistance to SARS-CoV-2 infection in lung cells, suggesting its beneficial health effect. Conclusion The method demonstrated the production of SARS-2pv for the analysis of vaccine or drug candidates. When KRG was assessed by the method, it protected host cells from coronavirus infection. Further studies will be followed for demonstrating this potential benefit.
Collapse
|
10
|
Xiang H, Liu QP. Alterations of the gut microbiota in coronavirus disease 2019 and its therapeutic potential. World J Gastroenterol 2022; 28:6689-6701. [PMID: 36620345 PMCID: PMC9813939 DOI: 10.3748/wjg.v28.i47.6689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/07/2022] [Accepted: 11/23/2022] [Indexed: 12/19/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious threat to global health. SARS-CoV-2 infects host cells primarily by binding to angiotensin-converting enzyme 2, which is coexpressed in alveolar type 2 cells and gut epithelial cells. It is known that COVID-19 often presents with gastrointestinal symptoms and gut dysbiosis, mainly characterized by an increase in opportunistic pathogens and a decrease in beneficial commensal bacteria. In recent years, multiple studies have comprehensively explored gut microbiota alterations in COVID-19 and highlighted the clinical correlation between dysbiosis and COVID-19. SARS-CoV-2 causes gastrointestinal infections and dysbiosis mainly through fecal-oral transmission and the circulatory and immune pathways. Studies have shown that the gut microbiota and its metabolites can regulate the immune response and modulate antiviral effects. In addition, the gut microbiota is closely related to gastrointestinal symptoms, such as diarrhea, a common gastrointestinal symptom among COVID-19. Therefore, the contribution of the gut microbiota in COVID-19 should not be overlooked. Strategies targeting the gut microbiota via probiotics, prebiotics and fecal microbiota transplantation should be considered to treat this patient population in the future. However, the specific alterations and mechanisms as well as the contributions of gut microbiota in COVID-19 should be urgently further explored.
Collapse
Affiliation(s)
- Hui Xiang
- Department of Infectious Disease, Chongqing University Three Gorges Hospital, Chongqing 404100, China
| | - Qi-Ping Liu
- Department of Pulmonary and Critical Care Medicine, Chongqing University Three Gorges Hospital, Chongqing 404100, China
| |
Collapse
|
11
|
Fermented Foods of Korea and Their Functionalities. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8110645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Fermented foods are loved and enjoyed worldwide and are part of a tradition in several regions of the world. Koreans have traditionally had a healthy diet since people in this region have followed a fermented-foods diet for at least 5000 years. Fermented-product footprints are evolving beyond boundaries and taking the lead in the world of food. Fermented foods, such as jang (fermented soybean products), kimchi (fermented vegetables), jeotgal (fermented fish), and vinegar (liquor with grain and fruit fermentation), are prominent fermented foods in the Korean culture. These four major fermented foods have been passed down through the generations and define Korean cuisine. However, scientific advancements in the fermentation process have increased productivity rates and facilitated global exports. Recently, Korean kimchi and jang have garnered significant attention due to their nutritional and health-beneficial properties. The health benefits of various Korean fermented foods have been consistently supported by both preclinical and clinical research. Korean fermented foods effectively reduce the risk of cardiovascular and chronic metabolic diseases, such as immune regulation, memory improvement, obesity, diabetes, and high blood pressure. Additionally, kimchi is known to prevent and improve multiple metabolic diseases, including irritable bowel syndrome (IBS), and improve beneficial intestinal bacteria. These functional health benefits may reflect the synergistic effect between raw materials and various physiologically active substances produced during fermentation. Thus, fermented foods all over the world not only enrich our dining table with taste, aroma, and nutrition, but also the microorganisms involved in fermentation and metabolites of various fermentations have a profound effect on human health. This article describes the production and physiological functions of Korean fermented foods, which are anticipated to play a significant role in the wellness of the world’s population in the coming decades.
Collapse
|
12
|
Added Value of Biophysics to Study Lipid-Driven Biological Processes: The Case of Surfactins, a Class of Natural Amphiphile Molecules. Int J Mol Sci 2022; 23:ijms232213831. [PMID: 36430318 PMCID: PMC9693386 DOI: 10.3390/ijms232213831] [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: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
The role of membrane lipids is increasingly claimed to explain biological activities of natural amphiphile molecules. To decipher this role, biophysical studies with biomimetic membrane models are often helpful to obtain insights at the molecular and atomic levels. In this review, the added value of biophysics to study lipid-driven biological processes is illustrated using the case of surfactins, a class of natural lipopeptides produced by Bacillus sp. showing a broad range of biological activities. The mechanism of interaction of surfactins with biomimetic models showed to be dependent on the surfactins-to-lipid ratio with action as membrane disturber without membrane lysis at low and intermediate ratios and a membrane permeabilizing effect at higher ratios. These two mechanisms are relevant to explain surfactins' biological activities occurring without membrane lysis, such as their antiviral and plant immunity-eliciting activities, and the one involving cell lysis, such as their antibacterial and hemolytic activities. In both biological and biophysical studies, influence of surfactin structure and membrane lipids on the mechanisms was observed with a similar trend. Hence, biomimetic models represent interesting tools to elucidate the biological mechanisms targeting membrane lipids and can contribute to the development of new molecules for pharmaceutical or agronomic applications.
Collapse
|
13
|
Malik J, Ahmed S, Yaseen Z, Alanazi M, Alharby TN, Alshammari HA, Anwar S. Association of SARS-CoV-2 and Polypharmacy with Gut-Lung Axis: From Pathogenesis to Treatment. ACS OMEGA 2022; 7:33651-33665. [PMID: 36164411 PMCID: PMC9491241 DOI: 10.1021/acsomega.2c02524] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/29/2022] [Indexed: 06/12/2023]
Abstract
SARS-CoV-2 is a novel infectious contagion leading to COVID-19 disease. The virus has affected the lives of millions of people across the globe with a high mortality rate. It predominantly affects the lung (respiratory system), but it also affects other organs, including the cardiovascular, psychological, and gastrointestinal (GIT) systems. Moreover, elderly and comorbid patients with compromised organ functioning and pre-existing polypharmacy have worsened COVID-19-associated complications. Microbiota (MB) of the lung plays an important role in developing COVID-19. The extent of damage mainly depends on the predominance of opportunistic pathogens and, inversely, with the predominance of advantageous commensals. Changes in the gut MB are associated with a bidirectional shift in the interaction among the gut with a number of vital human organs, which leads to severe disease symptoms. This review focuses on dysbiosis in the gut-lung axis, COVID-19-induced worsening of comorbidities, and the influence of polypharmacy on MB.
Collapse
Affiliation(s)
- Jonaid
Ahmad Malik
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Guwahati, Assam 781101, India
- Department
of Biomedical Engineering, Indian Institute
of Technology Rupnagar 140001, India
| | - Sakeel Ahmed
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat 382355, India
| | - Zahid Yaseen
- Department
of Pharmaceutical Biotechnology, Delhi Pharmaceutical
Sciences and Research University, New Delhi, Delhi 110017, India
| | - Muteb Alanazi
- Department
of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
| | - Tareq Nafea Alharby
- Department
of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
| | | | - Sirajudheen Anwar
- Department
of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
| |
Collapse
|
14
|
Abstract
Despite great efforts have been made worldwide, the coronavirus disease 19 (COVID-19) still has not a definitive cure, although the availability of different vaccines are slowing down the transmission and severity. It has been shown that surfactin, a cyclic lipopeptide produced by Bacillus subtilis, is a molecule able to counteract both SARS-CoV-1, MERS-CoV and HCoV-229E coronaviruses. In this study the potential antiviral activity of surfactin against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was tested in vitro in a cellular model of infection. Our results show that 2 h treatment with surfactin is able to reduce SARS-CoV-2 infectivity on Vero E6 cells both at 24 h and after 7 days from viral inoculation, probably impairing the viral membrane integrity. Moreover, surfactin, at the concentrations used in our experimental settings, is not cytotoxic. We suggest surfactin as a new potential molecule against SARS-CoV-2, to be employed at least as a disinfectant.
Collapse
|
15
|
Moresco V, Charatzidou A, Oliver DM, Weidmann M, Matallana-Surget S, Quilliam RS. Binding, recovery, and infectiousness of enveloped and non-enveloped viruses associated with plastic pollution in surface water. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 308:119594. [PMID: 35680062 DOI: 10.1016/j.envpol.2022.119594] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/16/2022] [Accepted: 06/05/2022] [Indexed: 05/25/2023]
Abstract
Microplastics in wastewater and surface water rapidly become colonised by microbial biofilm. Such 'plastisphere' communities are hypothesised to persist longer and be disseminated further in the environment and may act as a vector for human pathogens, particularly as microplastics entering wastewater treatment plants are exposed to high concentrations of pathogenic bacteria. However, the potential for human viral pathogens to become associated with the plastisphere has never before been quantified. Here, we have used rotavirus (RV) SA11 (a non-enveloped enteric virus) and the enveloped bacteriophage Phi6 as model viruses to quantify binding and recovery from biofilm-colonised microplastic pellets in three different water treatments (filtered and non-filtered surface water, and surface water with added nutrients). Viruses associated with biofilm-colonised pellets were more stable compared to those remaining in the water. While infectious particles and genome copies of RV remained stable over the 48 h sampling period, Phi6 stability was highly impacted, with a reduction ranging from 2.18 to 3.94 log10. Virus particles were protected against inactivation factors when associated with the biofilm on microplastic surfaces, and when there was a high concentration of particulate matter in the liquid phase. Although our results suggest that the presence of an envelope may limit virus interaction with the plastisphere, the ability to recover both enveloped and non-enveloped infectious viruses from colonised microplastic pellets highlights an additional potential public health risk of surface waters becoming contaminated with microplastics, and subsequent human exposure to microplastics in the environment.
Collapse
Affiliation(s)
- Vanessa Moresco
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Anna Charatzidou
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - David M Oliver
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Manfred Weidmann
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, Senftenberg, D-01968, Germany
| | - Sabine Matallana-Surget
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Richard S Quilliam
- Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK.
| |
Collapse
|
16
|
Gudiña EJ, Teixeira JA. Bacillus licheniformis: The unexplored alternative for the anaerobic production of lipopeptide biosurfactants? Biotechnol Adv 2022; 60:108013. [PMID: 35752271 DOI: 10.1016/j.biotechadv.2022.108013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/27/2022] [Accepted: 06/19/2022] [Indexed: 11/02/2022]
Abstract
Microbial biosurfactants have attracted the attention of researchers and companies for the last decades, as they are considered promising candidates to replace chemical surfactants in numerous applications. Although in the last years, considerable advances were performed regarding strain engineering and the use of low-cost substrates in order to reduce their production costs, one of the main bottlenecks is their production at industrial scale. Conventional aerobic biosurfactant production processes result in excessive foaming, due to the use of high agitation and aeration rates necessary to increase dissolved oxygen concentration to allow microbial growth and biosurfactant production. Different approaches have been studied to overcome this problem, although with limited success. A not widely explored alternative is the development of foam-free processes through the anaerobic growth of biosurfactant-producing microorganisms. Surfactin, produced by Bacillus subtilis, is the most widely studied lipopeptide biosurfactant, and the most powerful biosurfactant known so far. Bacillus licheniformis strains produce lichenysin, a lipopeptide biosurfactant which structure is similar to surfactin. However, despite its extraordinary surface-active properties and potential applications, lichenysin has been scarcely studied. According to previous studies, B. licheniformis is better adapted to anaerobic growth than B. subtilis, and could be a good alternative for the anaerobic production of lipopeptide biosurfactants. In this review, the potential and limitations of surfactin and lichenysin production under anaerobic conditions will be analyzed, and the possibility of implementing foam-free processes for lichenysin production, in order to expand the market and applications of biosurfactants in different fields, will be discussed.
Collapse
Affiliation(s)
- Eduardo J Gudiña
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal.
| | - José A Teixeira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
17
|
Perturbation of alphavirus and flavivirus infectivity by components of the bacterial cell wall. J Virol 2022; 96:e0006022. [PMID: 35107376 DOI: 10.1128/jvi.00060-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The impact of the host microbiota on arbovirus infections is currently not well understood. Arboviruses are viruses transmitted through the bites of infected arthropods, predominantly mosquitoes or ticks. The first site of arbovirus inoculation is the biting site in the host skin, which is colonized by a complex microbial community that could possibly influence arbovirus infection. We demonstrated that pre-incubation of arboviruses with certain components of the bacterial cell wall, including lipopolysaccharides (LPS) of some Gram-negative bacteria and lipoteichoic acids or peptidoglycan of certain Gram-positive bacteria, significantly reduced arbovirus infectivity in vitro. This inhibitory effect was observed for arboviruses of different virus families, including chikungunya virus of the Alphavirus genus and Zika virus of the Flavivirus genus, showing that this is a broad phenomenon. A modest inhibitory effect was observed following incubation with a panel of heat-inactivated bacteria, including bacteria residing on the skin. No viral inhibition was observed after pre-incubation of cells with LPS. Furthermore, a virucidal effect of LPS on viral particles was noticed by electron microscopy. Therefore, the main inhibitory mechanism seems to be due to a direct effect on the virus particles. Together, these results suggest that bacteria are able to decrease the infectivity of alphaviruses and flaviviruses. Importance During the past decades the world has experienced a vast increase in epidemics of alphavirus and flavivirus infections. These viruses can cause severe diseases such as hemorrhagic fever, encephalitis and arthritis. Several alpha- and flaviviruses, such as chikungunya virus, Zika virus and dengue virus, are significant global health threats because of their high disease burden, their widespread (re-)emergence and the lack of (good) anti-arboviral strategies. Despite the clear health burden, alphavirus and flavivirus infection and disease are not fully understood. A knowledge gap in the interplay between the host and the arbovirus is the potential interaction with host skin bacteria. Therefore, we studied the effect of (skin) bacteria and bacterial cell wall components on alphavirus and flavivirus infectivity in cell culture. Our results show that certain bacterial cell wall components markedly reduced viral infectivity by directly interacting with the virus particle.
Collapse
|
18
|
Zhang F, Wan Y, Zuo T, Yeoh YK, Liu Q, Zhang L, Zhan H, Lu W, Xu W, Lui GC, Li AY, Cheung CP, Wong CK, Chan PK, Chan FK, Ng SC. Prolonged Impairment of Short-Chain Fatty Acid and L-Isoleucine Biosynthesis in Gut Microbiome in Patients With COVID-19. Gastroenterology 2022; 162:548-561.e4. [PMID: 34687739 PMCID: PMC8529231 DOI: 10.1053/j.gastro.2021.10.013] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/16/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with altered gut microbiota composition. Phylogenetic groups of gut bacteria involved in the metabolism of short chain fatty acids (SCFAs) were depleted in SARS-CoV-2-infected patients. We aimed to characterize a functional profile of the gut microbiome in patients with COVID-19 before and after disease resolution. METHODS We performed shotgun metagenomic sequencing on fecal samples from 66 antibiotics-naïve patients with COVID-19 and 70 non-COVID-19 controls. Serial fecal samples were collected (at up to 6 times points) during hospitalization and beyond 1 month after discharge. We assessed gut microbial pathways in association with disease severity and blood inflammatory markers. We also determined changes of microbial functions in fecal samples before and after disease resolution and validated these functions using targeted analysis of fecal metabolites. RESULTS Compared with non-COVID-19 controls, patients with COVID-19 with severe/critical illness showed significant alterations in gut microbiome functionality (P < .001), characterized by impaired capacity of gut microbiome for SCFA and L-isoleucine biosynthesis and enhanced capacity for urea production. Impaired SCFA and L-isoleucine biosynthesis in gut microbiome persisted beyond 30 days after recovery in patients with COVID-19. Targeted analysis of fecal metabolites showed significantly lower fecal concentrations of SCFAs and L-isoleucine in patients with COVID-19 before and after disease resolution. Lack of SCFA and L-isoleucine biosynthesis significantly correlated with disease severity and increased plasma concentrations of CXCL-10, NT- proB-type natriuretic peptide, and C-reactive protein (all P < .05). CONCLUSIONS Gut microbiome of patients with COVID-19 displayed impaired capacity for SCFA and L-isoleucine biosynthesis that persisted even after disease resolution. These 2 microbial functions correlated with host immune response underscoring the importance of gut microbial functions in SARS-CoV-2 infection pathogenesis and outcome.
Collapse
Affiliation(s)
- Fen Zhang
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yating Wan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Tao Zuo
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yun Kit Yeoh
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Qin Liu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Lin Zhang
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hui Zhan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenqi Lu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenye Xu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Grace C.Y. Lui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Amy Y.L. Li
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chun Pan Cheung
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Paul K.S. Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Francis K.L. Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Microbiota I-Center (MagIC), Shatin, Hong Kong, China
| | - Siew C. Ng
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Microbiota I-Center (MagIC), Shatin, Hong Kong, China,Correspondence Address correspondence to: Siew C. Ng, PhD, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, 9/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Shatin, Hong Kong
| |
Collapse
|
19
|
Mironenko A, Holubka O, Radchenko L, Zakharchuk I, Teteriuk N. VIRULICIDAL EFFECT OF THE PROBIOTIC DRUG "SVITECO-MULTI" ON POLIOVIRUS TYPE 1 AND INFLUENZA VIRUS. BULLETIN OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV. SERIES: BIOLOGY 2022. [DOI: 10.17721/1728.2748.2022.91.24-27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The experimental work shows the antiviral activity of the probiotic drug "Sviteco-Multi" which contains bacteria of the genus Bacillusin model system in cell culturesMDCKandHEp-2, against influenza A (H1N1)pdm2009 virus and vaccine poliovirus type 1, which allows to recommend it for use, in particular, as an alternative to traditional antiviral disinfectants.
Collapse
|
20
|
Muruato A, Vu MN, Johnson BA, Davis-Gardner ME, Vanderheiden A, Lokugamage K, Schindewolf C, Crocquet-Valdes PA, Langsjoen RM, Plante JA, Plante KS, Weaver SC, Debbink K, Routh AL, Walker D, Suthar MS, Shi PY, Xie X, Menachery VD. Mouse-adapted SARS-CoV-2 protects animals from lethal SARS-CoV challenge. PLoS Biol 2021; 19:e3001284. [PMID: 34735434 PMCID: PMC8594810 DOI: 10.1371/journal.pbio.3001284] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/16/2021] [Accepted: 10/17/2021] [Indexed: 01/16/2023] Open
Abstract
The emergence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has resulted in a pandemic causing significant damage to public health and the economy. Efforts to understand the mechanisms of Coronavirus Disease 2019 (COVID-19) have been hampered by the lack of robust mouse models. To overcome this barrier, we used a reverse genetic system to generate a mouse-adapted strain of SARS-CoV-2. Incorporating key mutations found in SARS-CoV-2 variants, this model recapitulates critical elements of human infection including viral replication in the lung, immune cell infiltration, and significant in vivo disease. Importantly, mouse adaptation of SARS-CoV-2 does not impair replication in human airway cells and maintains antigenicity similar to human SARS-CoV-2 strains. Coupled with the incorporation of mutations found in variants of concern, CMA3p20 offers several advantages over other mouse-adapted SARS-CoV-2 strains. Using this model, we demonstrate that SARS-CoV-2-infected mice are protected from lethal challenge with the original Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV), suggesting immunity from heterologous Coronavirus (CoV) strains. Together, the results highlight the use of this mouse model for further study of SARS-CoV-2 infection and disease.
Collapse
Affiliation(s)
- Antonio Muruato
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michelle N. Vu
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bryan A. Johnson
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Meredith E. Davis-Gardner
- Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Abigail Vanderheiden
- Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kumari Lokugamage
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Craig Schindewolf
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | | | - Rose M. Langsjoen
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jessica A. Plante
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center of Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kenneth S. Plante
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center of Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Scott C. Weaver
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center of Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kari Debbink
- Department of Natural Science, Bowie State University, Bowie, Maryland, United States of America
| | - Andrew L. Routh
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David Walker
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Mehul S. Suthar
- Department of Pediatrics, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Pei-Yong Shi
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Xuping Xie
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Vineet D. Menachery
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center of Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
21
|
Influence of inorganic and organic counter-cations on the surface properties and self-assembly of cyclic lipopeptide surfactin. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.126973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Lakdawala SS, Menachery VD. Catch Me if You Can: Superspreading of COVID-19. Trends Microbiol 2021; 29:919-929. [PMID: 34059436 PMCID: PMC8112283 DOI: 10.1016/j.tim.2021.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/03/2023]
Abstract
While significant insights have been gained concerning COVID-19, superspreading of coronaviruses remains a mystery. The vast majority of cases have been linked to a relatively small portion of infected individuals. Yet, the genetic sequence of the virus, severity of disease, and underlying host parameters, such as age, sex, and health conditions, are not clearly driving the superspreading phenomenon. In this commentary we discuss what is known and what is not known about coronavirus superspreader transmission and explore whether characteristics of the virion, the donor, or the environment contribute to this phenomenon.
Collapse
Affiliation(s)
- Seema S Lakdawala
- Department of Microbiology and Molecular Genetics, Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vineet D Menachery
- Department of Microbiology and Immunology, Institute for Human Infection and Immunity, World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| |
Collapse
|
23
|
Cherepanova EA, Galyautdinov IV, Burkhanova GF, Maksimov IV. Isolation and Identification of Lipopeptides of Bacillus subtilis 26D. APPL BIOCHEM MICRO+ 2021. [DOI: 10.1134/s0003683821050033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Oba M, Rongduo W, Saito A, Okabayashi T, Yokota T, Yasuoka J, Sato Y, Nishifuji K, Wake H, Nibu Y, Mizutani T. Natto extract, a Japanese fermented soybean food, directly inhibits viral infections including SARS-CoV-2 in vitro. Biochem Biophys Res Commun 2021; 570:21-25. [PMID: 34271432 PMCID: PMC8276596 DOI: 10.1016/j.bbrc.2021.07.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Natto, a traditional Japanese fermented soybean food, is well known to be nutritious and beneficial for health. In this study, we examined whether natto impairs infection by viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as well as bovine herpesvirus 1 (BHV-1). Interestingly, our results show that both SARS-CoV-2 and BHV-1 treated with a natto extract were fully inhibited infection to the cells. We also found that the glycoprotein D of BHV-1 was shown to be degraded by Western blot analysis and that a recombinant SARS-CoV-2 receptor-binding domain (RBD) was proteolytically degraded when incubated with the natto extract. In addition, RBD protein carrying a point mutation (UK variant N501Y) was also degraded by the natto extract. When the natto extract was heated at 100 °C for 10 min, the ability of both SARS-CoV-2 and BHV-1 to infect to the cells was restored. Consistent with the results of the heat inactivation, a serine protease inhibitor inhibited anti-BHV-1 activity caused by the natto extract. Thus, our findings provide the first evidence that the natto extract contains a protease(s) that inhibits viral infection through the proteolysis of the viral proteins.
Collapse
Affiliation(s)
- Mami Oba
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan
| | - Wen Rongduo
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan; Graduate School of Agriculture Cooperative Division of Veterinary Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| | - Tamaki Okabayashi
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| | - Tomoko Yokota
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan
| | - Junko Yasuoka
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan
| | - Yoko Sato
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan
| | - Koji Nishifuji
- Laboratory of Veterinary Internal Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hitoshi Wake
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan; National Institute of Technology (KOSEN), Tokyo, Japan
| | - Yutaka Nibu
- The University Research Administration Center (URAC), Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tetsuya Mizutani
- Center for Infectious Diseases of Epidemiology and Prevention Research (CEPiR), Tokyo, Japan; Graduate School of Agriculture Cooperative Division of Veterinary Science, Tokyo University of Agriculture and Technology, Tokyo, Japan.
| |
Collapse
|
25
|
Tan X, Shi L, Banerjee P, Liu X, Guo HF, Yu J, Bota-Rabassedas N, Rodriguez BL, Gibbons DL, Russell WK, Creighton CJ, Kurie JM. A protumorigenic secretory pathway activated by p53 deficiency in lung adenocarcinoma. J Clin Invest 2021; 131:137186. [PMID: 32931483 DOI: 10.1172/jci137186] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Therapeutic strategies designed to target TP53-deficient cancer cells remain elusive. Here, we showed that TP53 loss initiated a pharmacologically actionable secretory process that drove lung adenocarcinoma (LUAD) progression. Molecular, biochemical, and cell biological studies showed that TP53 loss increased the expression of Golgi reassembly and stacking protein 55 kDa (G55), a Golgi stacking protein that maintains Golgi organelle integrity and is part of a GOLGIN45 (G45)-myosin IIA-containing protein complex that activates secretory vesicle biogenesis in the Golgi. TP53 loss activated G55-dependent secretion by relieving G55 and myosin IIA from miR-34a-dependent silencing. G55-dependent secreted proteins enhanced the proliferative and invasive activities of TP53-deficient LUAD cells and promoted angiogenesis and CD8+ T cell exhaustion in the tumor microenvironment. A small molecule that blocks G55-G45 interactions impaired secretion and reduced TP53-deficient LUAD growth and metastasis. These results identified a targetable secretory vulnerability in TP53-deficient LUAD cells.
Collapse
Affiliation(s)
- Xiaochao Tan
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lei Shi
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Priyam Banerjee
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xin Liu
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hou-Fu Guo
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiang Yu
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Neus Bota-Rabassedas
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - B Leticia Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Chad J Creighton
- Department of Medicine, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA.,Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan M Kurie
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
26
|
Rehman S, Ravinayagam V, Nahvi I, Aldossary H, Al-Shammari M, Amiri MSA, Kishore U, Al-Suhaimi EA. Immunity, Sex Hormones, and Environmental Factors as Determinants of COVID-19 Disparity in Women. Front Immunol 2021; 12:680845. [PMID: 34484179 PMCID: PMC8416472 DOI: 10.3389/fimmu.2021.680845] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/29/2021] [Indexed: 01/16/2023] Open
Abstract
The current coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome virus 2 (SARS-CoV-2), has resulted in a major global pandemic, causing extreme morbidity and mortality. Few studies appear to suggest a significant impact of gender in morbidity and mortality, where men are reported at a higher risk than women. The infectivity, transmissibility, and varying degree of disease manifestation (mild, modest, and severe) in population studies reinforce the importance of a number of genetic and epigenetic factors, in the context of immune response and gender. The present review dwells on several contributing factors such as a stronger innate immune response, estrogen, angiotensin-converting enzyme 2 gene, and microbiota, which impart greater resistance to the SARS-CoV-2 infection and disease progression in women. In addition, the underlying importance of associated microbiota and certain environmental factors in gender-based disparity pertaining to the mortality and morbidity due to COVID-19 in women has also been addressed.
Collapse
Affiliation(s)
- Suriya Rehman
- Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Vijaya Ravinayagam
- Deanship of Scientific Research and Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Insha Nahvi
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al Hofuf, Saudi Arabia
| | - Hanan Aldossary
- Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Maha Al-Shammari
- Department of Public Health, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mai Saad Al Amiri
- Department of Obstetrics and Gynecology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Ebtesam A. Al-Suhaimi
- Biology Department, College of Science and Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
27
|
Ngo VL, Gewirtz AT. Microbiota as a potentially-modifiable factor influencing COVID-19. Curr Opin Virol 2021; 49:21-26. [PMID: 34000641 PMCID: PMC8059947 DOI: 10.1016/j.coviro.2021.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022]
Abstract
Impacts of respiratory tract viruses have long been appreciated to highly heterogeneous both between and within various populations. The SARS-CoV-2 pandemic, which is the first time that a pathogen's spread across the globe has been extensively monitored by direct detection of the pathogen itself rather just than the morbidity left in its wake, indicates such heterogeneity is not limited to outcomes of infections but whether infection of a particular host occurs at all. This suggests an important role for yet to be discovered environmental (i.e. non-genetic) factors that influence whether an exposure to the virus initiates a productive infection and, moreover, the severity of disease that results. This article discusses the emerging hypothesis that the composition of a host's commensal microbial communities, that is, its 'microbiome', may be one such determinant that influences outcomes following encounters with respiratory viral pathogens in general and SARS-CoV-2 in particular. Specifically, we will review the rationales and evidence that supports this hypothesis and, moreover, speculate as to possible approaches to manipulate microbiota to ameliorate disease induced by respiratory viral pathogens.
Collapse
Affiliation(s)
- Vu L Ngo
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
28
|
Zhang X, Tong Y, Wang J, Lyu X, Yang R. Screening of a Bacillus subtilis strain producing both nattokinase and milk-clotting enzyme and its application in fermented milk with thrombolytic activity. J Dairy Sci 2021; 104:9437-9449. [PMID: 34218912 DOI: 10.3168/jds.2020-19756] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/20/2021] [Indexed: 11/19/2022]
Abstract
Bacillus subtilis is a generally recognized as safe probiotic, which is used as a starter for natto fermentation. Natto is a functional food with antithrombus function due to nattokinase. Compared with natto, fermented milk is a more popular fermented food, which is commonly fermented by Lactobacillus bulgaricus and Streptococcus. However, there is no report on B. subtilis-fermented milk. In this study, to produce a functional fermented milk with antithrombus function, a B. subtilis strain (B. subtilis JNFE0126) that produced both nattokinase and milk-clotting enzyme was isolated from traditionally fermented natto and used as the starter for the functional fermented milk. In liquid fermentation culture, the peak values of thrombolytic activity and milk-clotting activity were 3,511 U/mL at 96 h and 874.5 Soxhlet unit/mL at 60 h, respectively. The optimal pH and temperature were pH 7.0 at 40°C for nattokinase and pH 6.5 and 55°C for milk-clotting enzyme, respectively. The thrombolytic activity in the fermented milk reached 215.1 U/mL after 8 h of fermentation. Sensory evaluation showed that the acceptance of the milk fermented by B. subtilis JNFE0126 was similar to the traditional milk fermented by L. bulgaricus and S. thermophilus. More importantly, oral intake of the fermented milk by the thrombosis-model mice prevented the development of thrombosis. Our results suggest that B. subtilis JNFE0126-fermented milk has potential as a novel, functional food in the prevention of thrombosis-related cardiovascular diseases.
Collapse
Affiliation(s)
- Xuan Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Yanjun Tong
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jing Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Xiaomei Lyu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Ruijin Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China.
| |
Collapse
|
29
|
Landay A, Bartley JM, Banerjee D, Hargis G, Haynes L, Keshavarzian A, Kuo CL, Kwon OS, Li S, Li S, Oh J, Ozbolat IT, Ucar D, Xu M, Yao X, Unutmaz D, Kuchel GA. Network Topology of Biological Aging and Geroscience-Guided Approaches to COVID-19. FRONTIERS IN AGING 2021; 2:695218. [PMID: 35128530 PMCID: PMC8813169 DOI: 10.3389/fragi.2021.695218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/22/2021] [Indexed: 01/08/2023]
Abstract
Aging has emerged as the greatest and most prevalent risk factor for the development of severe COVID-19 infection and death following exposure to the SARS-CoV-2 virus. The presence of multiple co-existing chronic diseases and conditions of aging further enhances this risk. Biological aging not only enhances the risk of chronic diseases, but the presence of such conditions further accelerates varied biological processes or "hallmarks" implicated in aging. Given growing evidence that it is possible to slow the rate of many biological aging processes using pharmacological compounds has led to the proposal that such geroscience-guided interventions may help enhance immune resilience and improve outcomes in the face of SARS-CoV-2 infection. Our review of the literature indicates that most, if not all, hallmarks of aging may contribute to the enhanced COVID-19 vulnerability seen in frail older adults. Moreover, varied biological mechanisms implicated in aging do not function in isolation from each other, and exhibit intricate effects on each other. With all of these considerations in mind, we highlight limitations of current strategies mostly focused on individual single mechanisms, and we propose an approach which is far more multidisciplinary and systems-based emphasizing network topology of biological aging and geroscience-guided approaches to COVID-19.
Collapse
Affiliation(s)
- Alan Landay
- Department of Medicine, Rush School of Medicine, Chicago, IL, United States
| | - Jenna M. Bartley
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Dishary Banerjee
- Engineering Science and Mechanics Department, The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, United States
| | - Geneva Hargis
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Laura Haynes
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Ali Keshavarzian
- Division of Digestive Diseases, Departments of Medicine, Pharmacology, Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL, United States
| | - Chia-Ling Kuo
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Connecticut Convergence Institute for Translation in Regenerative Engineering, Storrs, CT, United States
| | - Oh Sung Kwon
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
| | - Sheng Li
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Shuzhao Li
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Julia Oh
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Ibrahim Tarik Ozbolat
- Engineering Science and Mechanics Department, The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, United States
- Biomedical Engineering Department, Neurosurgery Department, Materials Research Institute, Penn State University, University Park, PA, United States
| | - Duygu Ucar
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Ming Xu
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Xudong Yao
- Department of Chemistry, University of Connecticut, Storrs, CT, United States
| | - Derya Unutmaz
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - George A. Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
30
|
Polymicrobial Interactions Operative during Pathogen Transmission. mBio 2021; 12:mBio.01027-21. [PMID: 34006664 PMCID: PMC8262881 DOI: 10.1128/mbio.01027-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Pathogen transmission is a key point not only for infection control and public health interventions but also for understanding the selective pressures in pathogen evolution. The “success” of a pathogen lies not in its ability to cause signs and symptoms of illness but in its ability to be shed from the initial hosts, survive between hosts, and then establish infection in a new host. Recent insights have shown the importance of the interaction between the pathogen and both the commensal microbiome and coinfecting pathogens on shedding, environmental survival, and acquisition of infection. Pathogens have evolved in the context of cooperation and competition with other microbes, and the roles of these cooperations and competitions in transmission can inform novel preventative and therapeutic strategies.
Collapse
|
31
|
Szlufman C, Shemesh M. Role of Probiotic Bacilli in Developing Synbiotic Food: Challenges and Opportunities. Front Microbiol 2021; 12:638830. [PMID: 33912147 PMCID: PMC8072055 DOI: 10.3389/fmicb.2021.638830] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/19/2021] [Indexed: 12/15/2022] Open
Abstract
The human body is inhabited by a vast diversity of probiotic microorganisms that could positively affect human physiology. Besides, prebiotic food substances may induce symbiotic relationship among probiotic species through the successful establishment of commensal microbiota, whose connections with the host are multifaceted and multidirectional. As deliberated throughout this review, prebiotic and synbiotic foods contain the capability to stimulate numerous health characteristics in host organisms through various means. Predominantly, the normal microbiota fosters the digestion of food and may boost the innate and adaptive immune system’s functionalities. Therefore, live probiotic bacteria, for instance, probiotic Bacilli obtained together with prebiotic food, can help stimulate healthiness in humans. Thus, we discuss how certain dietary fibers may preserve the probiotic efficacy by serving as the scaffold for probiotic Bacilli to colonize them through forming symbiotic interactions. The fibers can essentially promote protection by encapsulating probiotic Bacilli against various environmental and physical stresses that might kill the free-living bacterial cells. Besides, these fibers would serve as prebiotic substances that would eventually be utilized for the proliferation of probiotic cells. It is believed that applying this conceptual idea will provide a novel platform toward developing probiotic and synbiotic foods, as discussed in this review.
Collapse
Affiliation(s)
- Carolina Szlufman
- Department of Food Science, Institute of Postharvest Technology and Food Sciences, Agricultural Research Organization, The Volcani Center, Rishon LeZion, Israel
| | - Moshe Shemesh
- Department of Food Science, Institute of Postharvest Technology and Food Sciences, Agricultural Research Organization, The Volcani Center, Rishon LeZion, Israel
| |
Collapse
|
32
|
Hou Q, Kolodkin-Gal I. Harvesting the complex pathways of antibiotic production and resistance of soil bacilli for optimizing plant microbiome. FEMS Microbiol Ecol 2021; 96:5872479. [PMID: 32672816 DOI: 10.1093/femsec/fiaa142] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/13/2020] [Indexed: 01/04/2023] Open
Abstract
A sustainable future increasing depends on our capacity to utilize beneficial plant microbiomes to meet our growing needs. Plant microbiome symbiosis is a hallmark of the beneficial interactions between bacteria and their host. Specifically, colonization of plant roots by biocontrol agents and plant growth-promoting bacteria can play an important role in maintaining the optimal rhizosphere environment, supporting plant growth and promoting its fitness. Rhizosphere communities confer immunity against a wide range of foliar diseases by secreting antibiotics and activating plant defences. At the same time, the rhizosphere is a highly competitive niche, with multiple microbial species competing for space and resources, engaged in an arms race involving the production of a vast array of antibiotics and utilization of a variety of antibiotic resistance mechanisms. Therefore, elucidating the mechanisms that govern antibiotic production and resistance in the rhizosphere is of great significance for designing beneficial communities with enhanced biocontrol properties. In this review, we used Bacillus subtilis and B. amyloliquefaciens as models to investigate the genetics of antibiosis and the potential for its translation of into improved plant microbiome performance.
Collapse
Affiliation(s)
- Qihui Hou
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ilana Kolodkin-Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
33
|
Théatre A, Cano-Prieto C, Bartolini M, Laurin Y, Deleu M, Niehren J, Fida T, Gerbinet S, Alanjary M, Medema MH, Léonard A, Lins L, Arabolaza A, Gramajo H, Gross H, Jacques P. The Surfactin-Like Lipopeptides From Bacillus spp.: Natural Biodiversity and Synthetic Biology for a Broader Application Range. Front Bioeng Biotechnol 2021; 9:623701. [PMID: 33738277 PMCID: PMC7960918 DOI: 10.3389/fbioe.2021.623701] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/02/2021] [Indexed: 11/21/2022] Open
Abstract
Surfactin is a lipoheptapeptide produced by several Bacillus species and identified for the first time in 1969. At first, the biosynthesis of this remarkable biosurfactant was described in this review. The peptide moiety of the surfactin is synthesized using huge multienzymatic proteins called NonRibosomal Peptide Synthetases. This mechanism is responsible for the peptide biodiversity of the members of the surfactin family. In addition, on the fatty acid side, fifteen different isoforms (from C12 to C17) can be incorporated so increasing the number of the surfactin-like biomolecules. The review also highlights the last development in metabolic modeling and engineering and in synthetic biology to direct surfactin biosynthesis but also to generate novel derivatives. This large set of different biomolecules leads to a broad spectrum of physico-chemical properties and biological activities. The last parts of the review summarized the numerous studies related to the production processes optimization as well as the approaches developed to increase the surfactin productivity of Bacillus cells taking into account the different steps of its biosynthesis from gene transcription to surfactin degradation in the culture medium.
Collapse
Affiliation(s)
- Ariane Théatre
- Microbial Processes and Interactions, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Avenue de la Faculté, Gembloux, Belgium
| | - Carolina Cano-Prieto
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Marco Bartolini
- Laboratory of Physiology and Genetics of Actinomycetes, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Facultad de Ciencias, Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Yoann Laurin
- Laboratoire de Biophysique Moléculaire aux Interfaces, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, Université de Liège, Gembloux, Belgium.,Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens, France
| | - Magali Deleu
- Laboratoire de Biophysique Moléculaire aux Interfaces, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, Université de Liège, Gembloux, Belgium
| | - Joachim Niehren
- Inria Lille, and BioComputing Team of CRISTAL Lab (CNRS UMR 9189), Lille, France
| | - Tarik Fida
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Saïcha Gerbinet
- Chemical Engineering, Products, Environment, and Processes, University of Liège, Liège, Belgium
| | - Mohammad Alanjary
- Bioinformatics Group, Wageningen University, Wageningen, Netherlands
| | - Marnix H Medema
- Bioinformatics Group, Wageningen University, Wageningen, Netherlands
| | - Angélique Léonard
- Chemical Engineering, Products, Environment, and Processes, University of Liège, Liège, Belgium
| | - Laurence Lins
- Laboratoire de Biophysique Moléculaire aux Interfaces, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, Université de Liège, Gembloux, Belgium
| | - Ana Arabolaza
- Laboratory of Physiology and Genetics of Actinomycetes, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Facultad de Ciencias, Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Hugo Gramajo
- Laboratory of Physiology and Genetics of Actinomycetes, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Facultad de Ciencias, Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Harald Gross
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Philippe Jacques
- Microbial Processes and Interactions, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Avenue de la Faculté, Gembloux, Belgium
| |
Collapse
|
34
|
Zhang X, Tong Y, Lyu X, Wang J, Wang Y, Yang R. Prevention and Alleviation of Dextran Sulfate Sodium Salt-Induced Inflammatory Bowel Disease in Mice With Bacillus subtilis-Fermented Milk via Inhibition of the Inflammatory Responses and Regulation of the Intestinal Flora. Front Microbiol 2021; 11:622354. [PMID: 33519783 PMCID: PMC7845695 DOI: 10.3389/fmicb.2020.622354] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of inflammatory bowel disease (IBD) might be related to the local inflammatory damage and the dysbacteriosis of intestinal flora. Probiotics can regulate the intestinal flora and ameliorate IBD. The probiotic Bacillus subtilis strain B. subtilis JNFE0126 was used as the starter of fermented milk. However, the therapeutic effects of B. subtilis-fermented milk on IBD remain to be explored. In this research, the therapeutic effect of B. subtilis-fermented milk on dextran sulfate sodium salt (DSS)-induced IBD mouse model was evaluated. Besides, the expression of pro-inflammatory/anti-inflammatory cytokines, the proliferation of the intestinal stem cells, and the reconstruction of the mucosa barrier were investigated. Finally, alteration of the gut microbiota was investigated by taxonomic analysis. As shown by the results, the disease activity index (DAI) of IBD was significantly decreased through oral administration of B. subtilis (JNFE0126)-fermented milk, and intestinal mucosa injury was attenuated. Moreover, B. subtilis could reduce the inflammatory response of the intestinal mucosa, induce proliferation of the intestinal stem cell, and promote reconstruction of the mucosal barrier. Furthermore, B. subtilis could rebalance the intestinal flora, increasing the abundance of Bacillus, Alistipes, and Lactobacillus while decreasing the abundance of Escherichia and Bacteroides. In conclusion, oral administration of the B. subtilis-fermented milk could alleviate DSS-induced IBD via inhibition of inflammatory response, promotion of the mucosal barrier reconstruction, and regulation of the intestinal flora.
Collapse
Affiliation(s)
- Xuan Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yanjun Tong
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaomei Lyu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jing Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yuxue Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ruijin Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
35
|
Nishimoto A, Wohlgemuth N, Rosch J, Schultz-Cherry S, Cortez V, Rowe HM. Transkingdom Interactions Important for the Pathogenesis of Human Viruses. J Infect Dis 2020; 223:S201-S208. [PMID: 33330907 DOI: 10.1093/infdis/jiaa735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The bacterial, fungal, and helminthic species that comprise the microbiome of the mammalian host have profound effects on health and disease. Pathogenic viruses must contend with the microbiome during infection and likely have evolved to exploit or evade the microbiome. Both direct interactions between the virions and the microbiota and immunomodulation and tissue remodeling caused by the microbiome alter viral pathogenesis in either host- or virus-beneficial ways. Recent insights from in vitro and murine models of viral pathogenesis have highlighted synergistic and antagonistic, direct and indirect interactions between the microbiome and pathogenic viruses. This review will focus on the transkingdom interactions between human gastrointestinal and respiratory viruses and the constituent microbiome of those tissues.
Collapse
Affiliation(s)
- Andrew Nishimoto
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Nicholas Wohlgemuth
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Valerie Cortez
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hannah M Rowe
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
36
|
Donati Zeppa S, Agostini D, Piccoli G, Stocchi V, Sestili P. Gut Microbiota Status in COVID-19: An Unrecognized Player? Front Cell Infect Microbiol 2020; 10:576551. [PMID: 33324572 PMCID: PMC7725702 DOI: 10.3389/fcimb.2020.576551] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/30/2020] [Indexed: 01/07/2023] Open
Abstract
Infection with the SARS-CoV-2 virus causes cardiopulmonary and vascular complications, ranging in severity. Understanding the pathogenic mechanisms of the novel SARS-CoV2 infection and progression can provide potential novel targets for its prevention and/or treatment. Virus microbiota reciprocal interactions have been studied in a variety of viral infections. For example, the integrity of Coronavirus particles can be disrupted by surfactin, a bacterial surface molecule that targets other viruses, including that of influenza A. In this light, intestinal microbiota likely influences COVID-19 virulence, while from its side SARS-CoV-2 may affect the intestinal microbiome promoting dysbiosis and other deleterious consequences. Hence, the microbiota pre-existing health status and its alterations in the course of SARS-CoV-2 infection, are likely to play an important, still underscored role in determining individual susceptibility and resilience to COVID-19. Indeed, the vast majority of COVID-19 worst clinical conditions and fatalities develop in subjects with specific risk factors such as aging and the presence of one or more comorbidities, which are intriguingly characterized also by unhealthy microbiome status. Moreover, these comorbidities require complex pharmacological regimens known as "polypharmacy" that may further affect microbiota integrity and worsen the resilience to viral infections. This complex situation may represent a further and underestimated risk with regard to COVID-19 clinical burden for the elderly and comorbid people. Here, we discuss the possible biological, physiopathological, and clinical implications of gut microbiota in COVID-19 and the strategies to improve/maintain its healthy status as a simple and adjunctive strategy to reduce COVID-19 virulence and socio-sanitary burden.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | | | | | | |
Collapse
|
37
|
Is a healthy microbiome responsible for lower mortality in COVID-19? Biologia (Bratisl) 2020; 76:819-829. [PMID: 33078028 PMCID: PMC7557238 DOI: 10.2478/s11756-020-00614-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/30/2020] [Indexed: 12/17/2022]
Abstract
The novel severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) is the cause of an ongoing pandemic with significant case fatality ratio (CFR) worldwide. Although SARS-CoV-2 primarily causes respiratory infection by binding to ACE2 receptors present on alveolar epithelial cells, studies have been published linking the disease to the small intestine enterocytes and its microbiome. Dysbiosis of microbiome, mainly intestinal and lung, can affect the course of the disease. Environmental factors, such as reduced intake of commensal bacteria from the environment or their products in the diet, play an important role in microbiome formation, which can significantly affect the immune response. In elderly, obese or chronically ill people, the microbiota is often damaged. Therefore, we speculate that a good microbiome may be one of the factors responsible for lower CFR from the coronavirus disease 2019 (COVID-19). An approach using tailored nutrition and supplements known to improve the intestinal microbiota and its immune function might help minimize the impact of the disease at least on people at higher risk from coronavirus.
Collapse
|
38
|
Huang SW, Tai CH, Hsu YM, Cheng D, Hung SJ, Chai KM, Wang YF, Wang JR. Assessing the application of a pseudovirus system for emerging SARS-CoV-2 and re-emerging avian influenza virus H5 subtypes in vaccine development. Biomed J 2020; 43:375-387. [PMID: 32611537 PMCID: PMC7274974 DOI: 10.1016/j.bj.2020.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/18/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Highly pathogenic emerging and re-emerging viruses continuously threaten lives worldwide. In order to provide prophylactic prevention from the emerging and re-emerging viruses, vaccine is suggested as the most efficient way to prevent individuals from the threat of viral infection. Nonetheless, the highly pathogenic viruses need to be handled in a high level of biosafety containment, which hinders vaccine development. To shorten the timeframe of vaccine development, the pseudovirus system has been widely applied to examine vaccine efficacy or immunogenicity in the emerging and re-emerging viruses. Methods We developed pseudovirus systems for emerging SARS coronavirus 2 (SARS-CoV-2) and re-emerging avian influenza virus H5 subtypes which can be handled in the biosafety level 2 facility. Through the generated pseudovirus of SARS-CoV-2 and avian influenza virus H5 subtypes, we successfully established a neutralization assay to quantify the neutralizing activity of antisera against the viruses. Results The result of re-emerging avian influenza virus H5Nx pseudoviruses provided valuable information for antigenic evolution and immunogenicity analysis in vaccine candidate selection. Together, our study assessed the potency of pseudovirus systems in vaccine efficacy, antigenic analysis, and immunogenicity in the vaccine development of emerging and re-emerging viruses. Conclusion Instead of handling live highly pathogenic viruses in a high biosafety level facility, using pseudovirus systems would speed up the process of vaccine development to provide community protection against emerging and re-emerging viral diseases with high pathogenicity.
Collapse
Affiliation(s)
- Sheng-Wen Huang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | - Ching-Hui Tai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Yin-Mei Hsu
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Dayna Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Su-Jhen Hung
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Ya-Fang Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Jen-Ren Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan; Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
39
|
Honarmand Ebrahimi K. SARS-CoV-2 spike glycoprotein-binding proteins expressed by upper respiratory tract bacteria may prevent severe viral infection. FEBS Lett 2020; 594:1651-1660. [PMID: 32449939 PMCID: PMC7280584 DOI: 10.1002/1873-3468.13845] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 12/26/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a major global challenge. The virus infects host cells using its spike glycoprotein (S-protein) and has significantly higher infectivity and mortality rates among the aged population. Here, based on bioinformatic analysis, I provide evidence that some members of the upper respiratory tract (URT) commensal bacteria express viral S-protein -binding proteins. Based on this analysis and available data showing a decline in the population of these bacteria in the elderly, I propose that some URT commensal bacteria hamper SARS-CoV-2 infectivity and that a decline in the population of these bacteria contributes to the severity of infection. Further studies should provide a better understanding of the interaction of URT bacteria and SARS-CoV-2, which may lead to new therapeutic approaches.
Collapse
|
40
|
Novel Ionophores Active against La Crosse Virus Identified through Rapid Antiviral Screening. Antimicrob Agents Chemother 2020; 64:AAC.00086-20. [PMID: 32284379 DOI: 10.1128/aac.00086-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022] Open
Abstract
Bunyaviruses are significant human pathogens, causing diseases ranging from hemorrhagic fevers to encephalitis. Among these viruses, La Crosse virus (LACV), a member of the California serogroup, circulates in the eastern and midwestern United States. While LACV infection is often asymptomatic, dozens of cases of encephalitis are reported yearly. Unfortunately, no antivirals have been approved to treat LACV infection. Here, we developed a method to rapidly test potential antivirals against LACV infection. From this screen, we identified several potential antiviral molecules, including known antivirals. Additionally, we identified many novel antivirals that exhibited antiviral activity without affecting cellular viability. Valinomycin, a potassium ionophore, was among our top targets. We found that valinomycin exhibited potent anti-LACV activity in multiple cell types in a dose-dependent manner. Valinomycin did not affect particle stability or infectivity, suggesting that it may preclude virus replication by altering cellular potassium ions, a known determinant of LACV entry. We extended these results to other ionophores and found that the antiviral activity of valinomycin extended to other viral families, including bunyaviruses (Rift Valley fever virus, Keystone virus), enteroviruses (coxsackievirus, rhinovirus), flavirivuses (Zika virus), and coronaviruses (human coronavirus 229E [HCoV-229E] and Middle East respiratory syndrome CoV [MERS-CoV]). In all viral infections, we observed significant reductions in virus titer in valinomycin-treated cells. In sum, we demonstrate the importance of potassium ions to virus infection, suggesting a potential therapeutic target to disrupt virus replication.
Collapse
|
41
|
Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat Med 2020. [PMID: 32284615 DOI: 10.1038/s41591‐020‐0820‐9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA. .,Scripps Research Translational Institute, La Jolla, CA, USA.
| | - Andrew Rambaut
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK
| | - W Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health of Columbia University, New York, NY, USA
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, The University of Sydney, Sydney, Australia
| | - Robert F Garry
- Tulane University, School of Medicine, Department of Microbiology and Immunology, New Orleans, LA, USA.,Zalgen Labs, Germantown, MD, USA
| |
Collapse
|
42
|
Affiliation(s)
- Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
- Scripps Research Translational Institute, La Jolla, CA, USA.
| | - Andrew Rambaut
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK
| | - W Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health of Columbia University, New York, NY, USA
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, The University of Sydney, Sydney, Australia
| | - Robert F Garry
- Tulane University, School of Medicine, Department of Microbiology and Immunology, New Orleans, LA, USA
- Zalgen Labs, Germantown, MD, USA
| |
Collapse
|
43
|
Affiliation(s)
- Ursula Neu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Bernardo A. Mainou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
44
|
Peptidoglycan-Associated Cyclic Lipopeptide Disrupts Viral Infectivity. J Virol 2019; 93:JVI.01282-19. [PMID: 31462558 PMCID: PMC6819921 DOI: 10.1128/jvi.01282-19] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 08/17/2019] [Indexed: 12/23/2022] Open
Abstract
In this article, we consider a role for bacteria in shaping coronavirus infection. Taking cues from studies of enteric viruses, we initially investigated how bacterial surface components might improve CoV infection. Instead, we found that peptidoglycan-associated surfactin is a potent viricidal compound that disrupts virion integrity with broad activity against enveloped viruses. Our results indicate that interactions with commensal bacterial may improve or disrupt viral infections, highlighting the importance of understanding these microbial interactions and their implications for viral pathogenesis and treatment. Enteric viruses exploit bacterial components, including lipopolysaccharides (LPS) and peptidoglycan (PG), to facilitate infection in humans. Because of their origin in the bat enteric system, we wondered if severe acute respiratory syndrome coronavirus (SARS-CoV) or Middle East respiratory syndrome CoV (MERS-CoV) also use bacterial components to modulate infectivity. To test this question, we incubated CoVs with LPS and PG and evaluated infectivity, finding no change following LPS treatment. However, PG from Bacillus subtilis reduced infection >10,000-fold, while PG from other bacterial species failed to recapitulate this. Treatment with an alcohol solvent transferred inhibitory activity to the wash, and mass spectrometry revealed surfactin, a cyclic lipopeptide antibiotic, as the inhibitory compound. This antibiotic had robust dose- and temperature-dependent inhibition of CoV infectivity. Mechanistic studies indicated that surfactin disrupts CoV virion integrity, and surfactin treatment of the virus inoculum ablated infection in vivo. Finally, similar cyclic lipopeptides had no effect on CoV infectivity, and the inhibitory effect of surfactin extended broadly to enveloped viruses, including influenza, Ebola, Zika, Nipah, chikungunya, Una, Mayaro, Dugbe, and Crimean-Congo hemorrhagic fever viruses. Overall, our results indicate that peptidoglycan-associated surfactin has broad viricidal activity and suggest that bacteria by-products may negatively modulate virus infection. IMPORTANCE In this article, we consider a role for bacteria in shaping coronavirus infection. Taking cues from studies of enteric viruses, we initially investigated how bacterial surface components might improve CoV infection. Instead, we found that peptidoglycan-associated surfactin is a potent viricidal compound that disrupts virion integrity with broad activity against enveloped viruses. Our results indicate that interactions with commensal bacterial may improve or disrupt viral infections, highlighting the importance of understanding these microbial interactions and their implications for viral pathogenesis and treatment.
Collapse
|