1
|
Kumariya R, Sun J, Lusvarghi S, O'Dell S, Zhao G, Doria-Rose NA, Bewley CA. An engineered antibody-lectin conjugate targeting the HIV glycan shield protects humanized mice against HIV challenge. Mol Ther 2025:S1525-0016(25)00213-8. [PMID: 40156187 DOI: 10.1016/j.ymthe.2025.03.039] [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: 11/13/2024] [Revised: 02/19/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Enveloped viruses responsible for global health pandemics often display a glycan shield on their surface envelope glycoproteins. In HIV, the glycan shield is formed by clusters of high-mannose glycans and plays essential roles in viral fitness and immune evasion. A few mannose-binding lectins potently inactivate HIV but have not been fully exploited due to poor pharmacokinetics and short serum half-lives. To address this, we engineered an antibody-lectin conjugate comprising the anti-HIV lectin griffithsin (GRFT) to the Fc region of human IgG1, with the aim of extending its serum half-life and augmenting anti-HIV activity by inducing immune effector responses. Engineered mGRFT-Fc produced in bacteria exhibited picomolar anti-HIV activity and an extended serum half-life, and mGRFT-Fc produced in mammalian cells (mGRFT-Fcglyc) elicited immune effector responses. In HIV-infected CD34+-humanized mice, both GRFT and mGRFT-Fcglyc effectively suppressed viral loads for up to 8 weeks after a single dose. Significantly, mGRFT-Fcglyc prevented HIV infection by neutralizing HIV and provided sustained protection from break-through infections via Fc-mediated immune effector responses, exhibiting a dual mode of protection. This study demonstrates the successful engineering of a lectin-based biologic and provides early evidence that a glycan-targeting agent alone can confer protection from viral infection in vivo.
Collapse
Affiliation(s)
- Rashmi Kumariya
- Natural Products Chemistry Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jiadong Sun
- Natural Products Chemistry Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sabrina Lusvarghi
- Natural Products Chemistry Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sijy O'Dell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Gengxiang Zhao
- Natural Products Chemistry Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nicole A Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Carole A Bewley
- Natural Products Chemistry Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Xu H, Deng B, Wu E, Zhu Y, Qi Q, Feng Y, Lu Y. Enhanced Immunogenicity of Chicken H9N2 Influenza Inactivated Vaccine Through a Novel Dual-Targeting Fusion Protein Strategy. Vaccines (Basel) 2025; 13:294. [PMID: 40266216 PMCID: PMC11945385 DOI: 10.3390/vaccines13030294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Targeted delivery of antigens to dendritic cells (DCs) is an effective strategy for enhancing vaccine efficacy. METHODS In this study, dual-targeting fusion proteins (GRFT-VHH54 and GRFT-VHH74) were constructed by fusing Griffithsin (GRFT), an algae-derived lectin with enveloped virus-binding properties, to DC-specific binding nanobodies (VHH54 and VHH74). Vaccines were formulated by combining the inactivated H9N2 avian influenza virus with these fusion proteins, and the potential of the fusion proteins to enhance vaccine-induced immunity in chickens was systematically evaluated. For parallel comparison, control groups included H9N2 avian influenza vaccines containing the inactivated virus alone, the inactivated virus with the immune enhancer CVCVA5, and a commercial H9N2 avian influenza inactivated vaccine. RESULTS At 4 weeks post-immunization, chickens vaccinated with the inactivated H9N2 virus combined with the GRFT-VHH74 fusion protein (1/2 H9+GRFT-VHH74) exhibited significantly enhanced humoral, mucosal, and cellular immune responses compared to those vaccinated with the inactivated H9N2 virus alone or the commercial H9N2 vaccine (p < 0.05). Additionally, chickens in the 1/2 H9+GRFT-VHH74 group exhibited enhanced resistance to the heterologous H9N2 subtype avian influenza virus, achieving a 90% protection rate, which was higher than that of the other groups. CONCLUSIONS These results indicate that the GRFT-VHH74 fusion protein has significant potential for advancing the development of inactivated vaccines against the H9N2 subtype avian influenza. Furthermore, it provides valuable insights for enhancing the immunogenicity and efficacy of inactivated vaccines targeting other avian influenza subtypes.
Collapse
Affiliation(s)
- Hai Xu
- Institute of Taizhou Agricultural Science, Jiangsu Academy of Agricultural Science, Taizhou 225300, China;
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Bihua Deng
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Erzhong Wu
- Animal Husbandry and Veterinary Department, Taizhou Municipal Bureau of Agriculture and Rural Affairs, Taizhou 225300, China
| | - Yalu Zhu
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Qiurong Qi
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Yaming Feng
- Institute of Taizhou Agricultural Science, Jiangsu Academy of Agricultural Science, Taizhou 225300, China;
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| |
Collapse
|
3
|
Wang LL, Alfson K, Eaton B, Mattix ME, Goez-Gazi Y, Holbrook MR, Carrion R, Xiang SH. Algal Lectin Griffithsin Inhibits Ebola Virus Infection. Molecules 2025; 30:892. [PMID: 40005201 PMCID: PMC11858388 DOI: 10.3390/molecules30040892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Algal lectin Griffithsin (GRFT) is a well-known mannose-binding protein which has broad-spectrum antiviral activity against several important infectious viruses including HIV, HCV, and SARS-CoV-2. Therefore, GRFT has been brought great attention to antiviral therapeutic development. In this report, we have tested GRFT's activity against the lethal Ebola virus in vitro and in vivo. Our data have shown that the IC50 value is about 42 nM for inhibiting Zaire Ebola virus (EBOV) infection in vitro. The preliminary in vivo mice model using mouse-adapted EBOV has also shown a certain efficacy for delayed mortality compared to the control animals. A GRFT pull-down experiment using viral particles demonstrates that GRFT can bind to N-glycans of EBOV. Thus, it can be concluded that GRFT, through binding to viral glycans, may block Ebola virus infection and has potential for the treatment of Ebola virus disease (EVD).
Collapse
Affiliation(s)
- Leah Liu Wang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| | - Kendra Alfson
- Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA (Y.G.-G.)
| | - Brett Eaton
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Marc E. Mattix
- Nonclinical Pathology Services, LLC, 5920 Clubhouse Pointe Dr., Medina 44256, OH, USA
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA (Y.G.-G.)
| | - Michael R. Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA (Y.G.-G.)
| | - Shi-Hua Xiang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| |
Collapse
|
4
|
Parajuli B, Acharya K, Bach HC, Zhang S, Abrams CF, Chaiken I. Monovalent Lectin Microvirin Utilizes Hydropathic Recognition of HIV-1 Env for Inhibition of Virus Cell Infection. Viruses 2025; 17:82. [PMID: 39861871 PMCID: PMC11768445 DOI: 10.3390/v17010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Microvirin is a lectin molecule known to have monovalent interaction with glycoprotein gp120. A previously reported high-resolution structural analysis defines the mannobiose-binding cavity of Microvirin. Nonetheless, structure does not directly define the energetics of binding contributions of protein contact residues. To better understand the nature of the MVN-Env glycan interaction, we used mutagenesis to evaluate the residue contributions to the mannobiose binding site of MVN that are important for Env gp120 glycan binding. MVN binding site amino acid residues were individually replaced by alanine, and the resulting purified recombinant MVN variants were examined for gp120 interaction using competition Enzyme-Linked Immunosorbent Assay (ELISA), biosensor surface plasmon resonance, calorimetry, and virus neutralization assays. Our findings highlight the role of both uncharged polar and non-polar residues in forming a hydropathic recognition site for the monovalent glycan engagement of Microvirin, in marked contrast to the charged residues utilized in the two Cyanovirin-N (CVN) glycan-binding sites.
Collapse
Affiliation(s)
- Bibek Parajuli
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (K.A.); (H.C.B.); (S.Z.)
| | - Kriti Acharya
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (K.A.); (H.C.B.); (S.Z.)
| | - Harry Charles Bach
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (K.A.); (H.C.B.); (S.Z.)
| | - Shiyu Zhang
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (K.A.); (H.C.B.); (S.Z.)
| | - Cameron F. Abrams
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA;
| | - Irwin Chaiken
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (K.A.); (H.C.B.); (S.Z.)
| |
Collapse
|
5
|
Tang J, Li R, Jiang T, Lv J, Jiang Y, Zhou X, Chen H, Li M, Wu A, Yu B, Takala TM, Saris PEJ, Li S, Fang Z. Heterologous Expression of the Antiviral Lectin Griffithsin in Probiotic Saccharomyces boulardii and In Vitro Characterization of Its Properties. Microorganisms 2024; 12:2414. [PMID: 39770617 PMCID: PMC11678560 DOI: 10.3390/microorganisms12122414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
In this study, the probiotic yeast Saccharomyces boulardii was engineered to secrete the antiviral lectin griffithsin. Twelve genetic tools with the griffithsin gene were cloned into the vector pSF-TEF1-URA3 and introduced into S. boulardii. In the recombinant strains, a 16.9 kDa band was detected using SDS-PAGE and further recognized by griffithsin antibody with Western blotting. S. boulardii strains FM, FT, HC, and HE with a high yield of griffithsin were acquired for property characterization in vitro. The four recombinant strains displayed a similar growth pattern to that of the control strains, while their morphological characteristics had changed according to scanning electron microscopy. In simulated gastrointestinal digestive fluids, the survival rates of S. boulardii FM, FT, and HC were significantly decreased (86.32 ± 1.49% to 95.36 ± 1.94%) compared with those of the control strains, with survival rates between 95.88 ± 0.00% and 98.74 ± 1.97%. The hydrophobicity of S. boulardii FM, the strain with the highest griffithsin production, was significantly increased to 21.89 ± 1.07%, and it exhibited a reduced auto-aggregation rate (57.64 ± 2.61%). Finally, Vero cells infected with porcine epidemic diarrhea virus (PEDV) were used to evaluate the strains' antiviral activity, and the rate at which S. boulardii FM inhibited PEDV reached 131.36 ± 1.06%, which was significantly higher than that of the control group.
Collapse
Affiliation(s)
- Jie Tang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Ran Li
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Tingyu Jiang
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Jiachen Lv
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Yuwei Jiang
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Xingjian Zhou
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Hong Chen
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Meiliang Li
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Aimin Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Timo M. Takala
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, 00014 Helsinki, Finland
| | - Per E. J. Saris
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, 00014 Helsinki, Finland
| | - Shuhong Li
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-Construction by Ministry of Agriculture and Rural Affairs of China and Sichuan Province), College of Food Science, Sichuan Agricultural University, Ya’an 625014, China
| |
Collapse
|
6
|
Tsui CK, Twells N, Durieux J, Doan E, Woo J, Khosrojerdi N, Brooks J, Kulepa A, Webster B, Mahal LK, Dillin A. CRISPR screens and lectin microarrays identify high mannose N-glycan regulators. Nat Commun 2024; 15:9970. [PMID: 39557836 PMCID: PMC11574202 DOI: 10.1038/s41467-024-53225-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/02/2024] [Indexed: 11/20/2024] Open
Abstract
Glycans play critical roles in cellular signaling and function. Unlike proteins, glycan structures are not templated from genetic sequences but synthesized by the concerted activity of many genes, making them historically challenging to study. Here, we present a strategy that utilizes CRISPR screens and lectin microarrays to uncover and characterize regulators of glycosylation. We applied this approach to study the regulation of high mannose glycans - the starting structure of all asparagine(N)-linked-glycans. We used CRISPR screens to uncover the expanded network of genes controlling high mannose levels, followed by lectin microarrays to fully measure the complex effect of select regulators on glycosylation globally. Through this, we elucidated how two high mannose regulators - TM9SF3 and the CCC complex - control complex N-glycosylation via regulating Golgi morphology and function. Notably, this allows us to interrogate Golgi function in-depth and reveals that similar disruption to Golgi morphology can lead to drastically different glycosylation outcomes. Collectively, this work demonstrates a generalizable approach for systematically dissecting the regulatory network underlying glycosylation.
Collapse
Affiliation(s)
- C Kimberly Tsui
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA.
| | - Nicholas Twells
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Jenni Durieux
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Emma Doan
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Jacqueline Woo
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Noosha Khosrojerdi
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Janiya Brooks
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Ayodeji Kulepa
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Brant Webster
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Andrew Dillin
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
7
|
Wiggins J, Karim SU, Liu B, Li X, Zhou Y, Bai F, Yu J, Xiang SH. Identification of a Novel Antiviral Lectin against SARS-CoV-2 Omicron Variant from Shiitake-Mushroom-Derived Vesicle-like Nanoparticles. Viruses 2024; 16:1546. [PMID: 39459880 PMCID: PMC11512411 DOI: 10.3390/v16101546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Lectins are a class of carbohydrate-binding proteins that may have antiviral activity by binding to the glycans on the virion surface to interfere with viral entry. We have identified a novel lectin (named Shictin) from Shiitake mushroom (Lentinula edodes)-derived vesicle-like nanoparticles (VLNs, or exosomes) that exhibits strong activity against the SARS-CoV-2 Omicron variant with an IC50 value of 87 nM. Shictin contains 298 amino acids and consists of two unique domains (N-terminal and C-terminal domain). The N-terminal domain is the carbohydrate-binding domain (CBD) that is homologous with CBDs of other lectins, suggesting that Shictin inhibits SARS-CoV-2 infection by binding to the glycans on the virion surface to prevent viral entry. This finding demonstrates that exosomes of vegetables are a valuable source for the identification of antiviral lectins. Therefore, it is believed that lectins from vegetable VLNs have potential as antiviral therapeutic agents.
Collapse
Affiliation(s)
- Joshua Wiggins
- Nebraska Center for Virology, University of Nebraska-Lincoln, Morrison Center 143, 4240 Fair Street, Lincoln, NE 68583, USA
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Shazeed-Ul Karim
- Department of Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Baolong Liu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Xingzhi Li
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - You Zhou
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Fengwei Bai
- Department of Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Shi-Hua Xiang
- Nebraska Center for Virology, University of Nebraska-Lincoln, Morrison Center 143, 4240 Fair Street, Lincoln, NE 68583, USA
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
8
|
Liu S, Yu Y, Guo K, Zhang Q, Jia Z, Alfredo MR, Ma P, Xie H, Bian X. Expression and antiviral application of exogenous lectin (griffithsin) in sweetpotatoes. FRONTIERS IN PLANT SCIENCE 2024; 15:1421244. [PMID: 39081525 PMCID: PMC11286482 DOI: 10.3389/fpls.2024.1421244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 08/02/2024]
Abstract
Griffithsin (GRFT) is a highly effective, broad-spectrum, safe, and stable viral inhibitor used to suppress a variety of viruses. However, little information is available on whether GRFT can prevent plant viral diseases. In this study, we constructed a GRFT overexpression vector containing the sweetpotato storage cell signal peptide and generated exogenous GRFT overexpression lines through genetic transformation. The transgenic plants showed notable resistance to sweetpotato virus disease in the virus nursery. To verify the antiplant virus function of GRFT, transient expression in tobacco leaves showed that GRFT inhibited the sweetpotato leaf curl virus (SPLCV). The replication of SPLCV was entirely inhibited when the concentration of GRFT reached a certain level. The results of pulldown and BIFC assays showed that GRFT did not interact with the six components of SPLCV. In addition, the mutated GRFTD/A without the binding ability of carbohydrate and anticoronavirus function, in which three aspartate residues at carbohydrate binding sites were all mutated to alanine, also inhibited SPLCV. Quantitative reverse-transcription PCR analyses showed that the tobacco antiviral-related genes HIN1, ICS1, WRKY40, and PR10 were overexpressed after GRFT/GRFTD/A injection. Furthermore, HIN1, ICS1, and PR10 were more highly expressed in the leaves injected with GRFTD/A. The results suggest that sweetpotato is able to express GRFT exogenously as a bioreactor. Moreover, exogenous GRFT expression inhibits plant viruses by promoting the expression of plant antiviral genes.
Collapse
Affiliation(s)
- Shuai Liu
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Yang Yu
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Ke Guo
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qian Zhang
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Zhaodong Jia
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Morales Rodriguez Alfredo
- Center for Tropical Crop Research, Research Institute of Tropical Roots and Tuber Crops (INIVIT), Santo Domingo, Cuba
| | - Peiyong Ma
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Hao Xie
- Xuzhou Institute of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Xuzhou, China
| | - Xiaofeng Bian
- Institute of Food Crops, Provincial Key Laboratory of Agrobiology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| |
Collapse
|
9
|
Devi OS, Singh SS, Kamei R, Sharma HJ, Devi MA, Brahmacharimayum N. Glycosylated SARs Cov 2 interaction with plant lectins. Glycoconj J 2024; 41:185-199. [PMID: 38748325 DOI: 10.1007/s10719-024-10154-x] [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: 02/27/2023] [Revised: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 08/17/2024]
Abstract
Lectins are non-immune carbohydrate-binding proteins/glycoproteins that are found everywhere in nature, from bacteria to human cells. They have also been a valuable biological tool for the purification and subsequent characterisation of glycoproteins due to their carbohydrate binding recognition capacity. Antinociceptive, antiulcer, anti-inflammatory activities and immune modulatory properties have been discovered in several plant lectins, with these qualities varying depending on the lectin carbohydrate-binding site. The Coronavirus of 2019 (COVID-19) is a respiratory disease that has swept the globe, killing millions and infecting millions more. Despite the availability of COVID-19 vaccinations and the vaccination of a huge portion of the world's population, viral infection rates continue to rise, causing major concern. Part of the reason for the vaccine's ineffectiveness has been attributed to repeated mutations in the virus's epitope determinant elements. The surface of the Coronavirus envelope is heavily glycosylated, with approximately sixty N-linked oligomannose, composite, and hybrid glycans covering the core of Man3GlcNAc2Asn. Some O-linked glycans have also been discovered. Many of these glyco-chains have also been subjected to multiple mutations, with only a few remaining conserved. As a result, numerous plant lectins with specificity for these viral envelope sugars have been discovered to interact preferentially with them and are being investigated as a potential future tool to combat coronaviruses such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by preventing viral attachment to the host. The review will discuss the possible applications of plant lectins as anti-coronaviruses including SARS-CoV-2, antinociceptive, anti-inflammation and its immune modulating effect.
Collapse
Affiliation(s)
| | | | - Rana Kamei
- Department of Biochemistry, Manipur University, Imphal, India
| | | | | | | |
Collapse
|
10
|
Tsui CK, Twells N, Doan E, Woo J, Khosrojerdi N, Brooks J, Kulepa A, Webster B, Mahal LK, Dillin A. CRISPR screens and lectin microarrays identify novel high mannose N-glycan regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.23.563662. [PMID: 37961200 PMCID: PMC10634773 DOI: 10.1101/2023.10.23.563662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Glycans play critical roles in cellular signaling and function. Unlike proteins, glycan structures are not templated from genes but the concerted activity of many genes, making them historically challenging to study. Here, we present a strategy that utilizes pooled CRISPR screens and lectin microarrays to uncover and characterize regulators of cell surface glycosylation. We applied this approach to study the regulation of high mannose glycans - the starting structure of all asparagine(N)-linked-glycans. We used CRISPR screens to uncover the expanded network of genes controlling high mannose surface levels, followed by lectin microarrays to fully measure the complex effect of select regulators on glycosylation globally. Through this, we elucidated how two novel high mannose regulators - TM9SF3 and the CCC complex - control complex N-glycosylation via regulating Golgi morphology and function. Notably, this method allowed us to interrogate Golgi function in-depth and reveal that similar disruption to Golgi morphology can lead to drastically different glycosylation outcomes. Collectively, this work demonstrates a generalizable approach for systematically dissecting the regulatory network underlying glycosylation.
Collapse
Affiliation(s)
- C Kimberly Tsui
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nicholas Twells
- Department of Chemistry, University of Alberta, Edmonton, Canada, T6G 2G2
| | - Emma Doan
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jacqueline Woo
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noosha Khosrojerdi
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Janiya Brooks
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ayodeji Kulepa
- Department of Chemistry, University of Alberta, Edmonton, Canada, T6G 2G2
| | - Brant Webster
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, Canada, T6G 2G2
| | - Andrew Dillin
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
11
|
Iyer K, Yan Z, Ross SR. Entry inhibitors as arenavirus antivirals. Front Microbiol 2024; 15:1382953. [PMID: 38650890 PMCID: PMC11033450 DOI: 10.3389/fmicb.2024.1382953] [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: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Arenaviruses belonging to the Arenaviridae family, genus mammarenavirus, are enveloped, single-stranded RNA viruses primarily found in rodent species, that cause severe hemorrhagic fever in humans. With high mortality rates and limited treatment options, the search for effective antivirals is imperative. Current treatments, notably ribavirin and other nucleoside inhibitors, are only partially effective and have significant side effects. The high lethality and lack of treatment, coupled with the absence of vaccines for all but Junín virus, has led to the classification of these viruses as Category A pathogens by the Centers for Disease Control (CDC). This review focuses on entry inhibitors as potential therapeutics against mammarenaviruses, which include both New World and Old World arenaviruses. Various entry inhibition strategies, including small molecule inhibitors and neutralizing antibodies, have been explored through high throughput screening, genome-wide studies, and drug repurposing. Notable progress has been made in identifying molecules that target receptor binding, internalization, or fusion steps. Despite promising preclinical results, the translation of entry inhibitors to approved human therapeutics has faced challenges. Many have only been tested in in vitro or animal models, and a number of candidates showed efficacy only against specific arenaviruses, limiting their broader applicability. The widespread existence of arenaviruses in various rodent species and their potential for their zoonotic transmission also underscores the need for rapid development and deployment of successful pan-arenavirus therapeutics. The diverse pool of candidate molecules in the pipeline provides hope for the eventual discovery of a broadly effective arenavirus antiviral.
Collapse
Affiliation(s)
| | | | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois, College of Medicine, Chicago, IL, United States
| |
Collapse
|
12
|
Zhang X, Zhang T, Zhao Y, Jiang L, Sui X. Structural, extraction and safety aspects of novel alternative proteins from different sources. Food Chem 2024; 436:137712. [PMID: 37852073 DOI: 10.1016/j.foodchem.2023.137712] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/25/2023] [Accepted: 10/08/2023] [Indexed: 10/20/2023]
Abstract
With rapid population growth and continued environmental degradation, it is no longer sustainable to rely on conventional proteins to meet human requirements. This has prompted the search for novel alternative protein sources of greater sustainability. Currently, proteins of non-conventional origin have been developed, with such alternative protein sources including plants, insects, algae, and even bacteria and fungi. Most of these protein sources have a high protein content, along with a balanced amino acid composition, and are regarded as healthy and nutritious sources of protein. While these novel alternative proteins have excellent nutritional, research on their structure are still at a preliminary stage, particularly so for insects, algae, bacteria, and fungi. Therefore, this review provides a comprehensive overview of promising novel alternative proteins developed in recent years with a focus on their nutrition, sustainability, classification, and structure. In addition, methods of extraction and potential safety factors for these proteins are summarized.
Collapse
Affiliation(s)
- Xin Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Tianyi Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yu Zhao
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lianzhou Jiang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xiaonan Sui
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
13
|
Marković V, Szczepańska A, Berlicki Ł. Antiviral Protein-Protein Interaction Inhibitors. J Med Chem 2024; 67:3205-3231. [PMID: 38394369 PMCID: PMC10945500 DOI: 10.1021/acs.jmedchem.3c01543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/04/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Continually repeating outbreaks of pathogenic viruses necessitate the construction of effective antiviral strategies. Therefore, the development of new specific antiviral drugs in a well-established and efficient manner is crucial. Taking into account the strong ability of viruses to change, therapies with diversified molecular targets must be sought. In addition to the widely explored viral enzyme inhibitor approach, inhibition of protein-protein interactions is a very valuable strategy. In this Perspective, protein-protein interaction inhibitors targeting HIV, SARS-CoV-2, HCV, Ebola, Dengue, and Chikungunya viruses are reviewed and discussed. Antibodies, peptides/peptidomimetics, and small molecules constitute three classes of compounds that have been explored, and each of them has some advantages and disadvantages for drug development.
Collapse
Affiliation(s)
- Violeta Marković
- Wrocław
University of Science and Technology, Department
of Bioorganic Chemistry, Wyb. Wyspiańskiego 27, 50-370 Wrocław, Poland
- University
of Kragujevac, Faculty of Science,
Department of Chemistry, R. Domanovića 12, 34000 Kragujevac, Serbia
| | - Anna Szczepańska
- Wrocław
University of Science and Technology, Department
of Bioorganic Chemistry, Wyb. Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Łukasz Berlicki
- Wrocław
University of Science and Technology, Department
of Bioorganic Chemistry, Wyb. Wyspiańskiego 27, 50-370 Wrocław, Poland
| |
Collapse
|
14
|
Gupta A, Yadav K, Yadav A, Ahmad R, Srivastava A, Kumar D, Khan MA, Dwivedi UN. Mannose-specific plant and microbial lectins as antiviral agents: A review. Glycoconj J 2024; 41:1-33. [PMID: 38244136 DOI: 10.1007/s10719-023-10142-7] [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: 09/02/2023] [Revised: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024]
Abstract
Lectins are non-immunological carbohydrate-binding proteins classified on the basis of their structure, origin, and sugar specificity. The binding specificity of such proteins with the surface glycan moiety determines their activity and clinical applications. Thus, lectins hold great potential as diagnostic and drug discovery agents and as novel biopharmaceutical products. In recent years, significant advancements have been made in understanding plant and microbial lectins as therapeutic agents against various viral diseases. Among them, mannose-specific lectins have being proven as promising antiviral agents against a variety of viruses, such as HIV, Influenza, Herpes, Ebola, Hepatitis, Severe Acute Respiratory Syndrome Coronavirus-1 (SARS-CoV-1), Middle Eastern Respiratory Syndrome Coronavirus (MERS-CoV) and most recent Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The binding of mannose-binding lectins (MBLs) from plants and microbes to high-mannose containing N-glycans (which may be simple or complex) of glycoproteins found on the surface of viruses has been found to be highly specific and mainly responsible for their antiviral activity. MBLs target various steps in the viral life cycle, including viral attachment, entry and replication. The present review discusses the brief classification and structure of lectins along with antiviral activity of various mannose-specific lectins from plants and microbial sources and their diagnostic and therapeutic applications against viral diseases.
Collapse
Affiliation(s)
- Ankita Gupta
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Kusum Yadav
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India.
| | - Anurag Yadav
- Department of Microbiology, C.P. College of Agriculture, Sardarkrushinagar Dantiwada Agriculture University, District-Banaskantha, Gujarat, India
| | - Rumana Ahmad
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India.
| | - Aditi Srivastava
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Dileep Kumar
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India
- Department of Biotechnology, Khwaja Moinuddin Chishti Language University, Lucknow, Uttar Pradesh, India
| | - Mohammad Amir Khan
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India
| | - U N Dwivedi
- Department of Biochemistry, University of Lucknow, Lucknow, Uttar Pradesh, India
| |
Collapse
|
15
|
Bilal M, Bashir H, Ameen R, Sumrin A, Hussain M, Manzoor S. Anti HCV activity and expression inhibition of HCC markers by protein extract from Iberis gibraltarica. BRAZ J BIOL 2024; 84:e252676. [PMID: 35384980 DOI: 10.1590/1519-6984.252676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 01/02/2022] [Indexed: 11/22/2022] Open
Abstract
Abstract Hepatitis C virus infection (HCV) is the foremost reason of progressive hepatic fibrosis and cirrhosis, with an elevated risk of hepatocellular carcinoma (HCC) development. Medicinal plants have been used for human health benefits for several years, but their therapeutic potential needs to be explored. The main objective of this study was to figure out the in vitro antiviral and anticancer characteristics of total crude protein of Iberis gibraltarica against HCV and HCC. Total crude protein of Iberis gibraltarica was isolated and quantified. The level of cytotoxicity was measured against the HepG2 cell line and it shows no significant cytotoxicity at the concentration of 504µg/ml. The anti-HCV effect was determined by absolute quantification via real time RT-PCR method and viral titer was reduced up to 66% in a dose dependent manner against the total protein of Iberis gibraltarica. The anticancer potential of Iberis gibraltarica was also examined through mRNA expression studies of AFP and GPC3 genes against the total protein of Iberis gibraltarica-treated HepG2 cells. The results show up to 90% of the down-regulation expression of AFP and GPC3. The obtained results indicate the therapeutic potential of total protein of Iberis gibraltarica against HCV and hepatocellular carcinoma in vitro.
Collapse
Affiliation(s)
- M. Bilal
- University of the Punjab, Pakistan
| | | | - R. Ameen
- University of the Punjab, Pakistan
| | | | | | | |
Collapse
|
16
|
Mia ME, Howlader M, Akter F, Hossain MM. Preclinical and Clinical Investigations of Potential Drugs and Vaccines for COVID-19 Therapy: A Comprehensive Review With Recent Update. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2024; 17:2632010X241263054. [PMID: 39070952 PMCID: PMC11282570 DOI: 10.1177/2632010x241263054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
The COVID-19 pandemic-led worldwide healthcare crisis necessitates prompt societal, ecological, and medical efforts to stop or reduce the rising number of fatalities. Numerous mRNA based vaccines and vaccines for viral vectors have been licensed for use in emergencies which showed 90% to 95% efficacy in preventing SARS-CoV-2 infection. However, safety issues, vaccine reluctance, and skepticism remain major concerns for making mass vaccination a successful approach to treat COVID-19. Hence, alternative therapeutics is needed for eradicating the global burden of COVID-19 from developed and low-resource countries. Repurposing current medications and drug candidates could be a more viable option for treating SARS-CoV-2 as these therapies have previously passed a number of significant checkpoints for drug development and patient care. Besides vaccines, this review focused on the potential usage of alternative therapeutic agents including antiviral, antiparasitic, and antibacterial drugs, protease inhibitors, neuraminidase inhibitors, and monoclonal antibodies that are currently undergoing preclinical and clinical investigations to assess their effectiveness and safety in the treatment of COVID-19. Among the repurposed drugs, remdesivir is considered as the most promising agent, while favipiravir, molnupiravir, paxlovid, and lopinavir/ritonavir exhibited improved therapeutic effects in terms of elimination of viruses. However, the outcomes of treatment with oseltamivir, umifenovir, disulfiram, teicoplanin, and ivermectin were not significant. It is noteworthy that combining multiple drugs as therapy showcases impressive effectiveness in managing individuals with COVID-19. Tocilizumab is presently employed for the treatment of patients who exhibit COVID-19-related pneumonia. Numerous antiviral drugs such as galidesivir, griffithsin, and thapsigargin are under clinical trials which could be promising for treating COVID-19 individuals with severe symptoms. Supportive treatment for patients of COVID-19 may involve the use of corticosteroids, convalescent plasma, stem cells, pooled antibodies, vitamins, and natural substances. This study provides an updated progress in SARS-CoV-2 medications and a crucial guide for inventing novel interventions against COVID-19.
Collapse
Affiliation(s)
- Md. Easin Mia
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Mithu Howlader
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Farzana Akter
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md. Murad Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
17
|
Arekal RN, J S, Kumar A, B S G, M S D. Structural and functional analysis of a novel galactose-binding lectin derived from Chlorella sorokiniana MW769776. J Biomol Struct Dyn 2023; 42:12970-12980. [PMID: 37904546 DOI: 10.1080/07391102.2023.2274519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023]
Abstract
A freshwater green microalgal strain was isolated and the lectin was identified in it by a strong hemagglutination activity (HA) assay. Characterization of the algal strain was found to be Chlorella sorokiniana (MW769776). A single step affinity chromatographic technique was developed to purify Chlorella sorokiniana lectin (CSL) using guar gum as the affinity matrix. The precipitate showed a single active peak with a titer value of 1024 HU, with a concentration of 1111 U, and a purification fold of 9. The purified protein exhibited a single band in SDS-PAGE with a molecular weight of 16 kDa. Analysis by liquid chromatography-electrospray ionization-quadrupole-time of flight mass spectrometry (LC-ESI-Q-TOF-MS) of tryptic-digested purified lectin showed that it was a monomeric protein. A multiple sequence alignment analysis revealed that the peptide sequences of CSL exhibited similarity with the H-type lectin domain of Micractinium conductrix. The structure of CSL was studied by FTIR and homology modeling methods, indicating the presence of α-helix as well as β-sheet in its secondary structure. Whereas the 3D structure exhibited the similarity with the core protein of light-harvesting reaction center complex of photosystem I. The significance of this study suggests that the characteristics of CSL are consistent with its identification as a hemagglutinin, a type of novel lectin, which suggests its candidature for various biological purposes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Roopashri N Arekal
- Department of Microbiology, Biotechnology and Food Technology, Bangalore University, Bengaluru, India
| | - Savitha J
- Department of Microbiology, Biotechnology and Food Technology, Bangalore University, Bengaluru, India
| | - Ashwini Kumar
- Department of Microbiology, Genei Lab Pvt Ltd, Bengaluru, India
| | - Gunashree B S
- Department of Microbiology, Mangalore University, Kodagu, India
| | - Divyashree M S
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
18
|
Afzal S, Ali L, Batool A, Afzal M, Kanwal N, Hassan M, Safdar M, Ahmad A, Yang J. Hantavirus: an overview and advancements in therapeutic approaches for infection. Front Microbiol 2023; 14:1233433. [PMID: 37901807 PMCID: PMC10601933 DOI: 10.3389/fmicb.2023.1233433] [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: 06/02/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Hantaviruses are a significant and emerging global public health threat, impacting more than 200,000 individuals worldwide each year. The single-stranded RNA viruses belong to the Hantaviridae family and are responsible for causing two acute febrile diseases in humans: Hantavirus pulmonary syndrome (HPS) and hemorrhagic fever with renal syndrome (HFRS). Currently, there are no licensed treatments or vaccines available globally for HTNV infection. Various candidate drugs have shown efficacy in increasing survival rates during the early stages of HTNV infection. Some of these drugs include lactoferrin, ribavirin, ETAR, favipiravir and vandetanib. Immunotherapy utilizing neutralizing antibodies (NAbs) generated from Hantavirus convalescent patients show efficacy against HTNV. Monoclonal antibodies such as MIB22 and JL16 have demonstrated effectiveness in protecting against HTNV infection. The development of vaccines and antivirals, used independently and/or in combination, is critical for elucidating hantaviral infections and the impact on public health. RNA interference (RNAi) arised as an emerging antiviral therapy, is a highly specific degrades RNA, with post-transcriptional mechanism using eukaryotic cells platform. That has demonstrated efficacy against a wide range of viruses, both in vitro and in vivo. Recent antiviral methods involve using small interfering RNA (siRNA) and other, immune-based therapies to target specific gene segments (S, M, or L) of the Hantavirus. This therapeutic approach enhances viral RNA clearance through the RNA interference process in Vero E6 cells or human lung microvascular endothelial cells. However, the use of siRNAs faces challenges due to their low biological stability and limited in vivo targeting ability. Despite their successful inhibition of Hantavirus replication in host cells, their antiviral efficacy may be hindered. In the current review, we focus on advances in therapeutic strategies, as antiviral medications, immune-based therapies and vaccine candidates aimed at enhancing the body's ability to control the progression of Hantavirus infections, with the potential to reduce the risk of severe disease.
Collapse
Affiliation(s)
- Samia Afzal
- CEMB, University of the Punjab, Lahore, Pakistan
| | - Liaqat Ali
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Anum Batool
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Momina Afzal
- CEMB, University of the Punjab, Lahore, Pakistan
| | - Nida Kanwal
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | | | | | - Atif Ahmad
- CEMB, University of the Punjab, Lahore, Pakistan
| | - Jing Yang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, Hubei, China
| |
Collapse
|
19
|
Borhani SG, Levine MZ, Krumpe LH, Wilson J, Henrich CJ, O'Keefe BR, Lo DC, Sittampalam GS, Godfrey AG, Lunsford RD, Mangalampalli V, Tao D, LeClair CA, Thole AP, Frey D, Swartz J, Rao G. An approach to rapid distributed manufacturing of broad spectrum anti-viral griffithsin using cell-free systems to mitigate pandemics. N Biotechnol 2023; 76:13-22. [PMID: 37054948 PMCID: PMC10330340 DOI: 10.1016/j.nbt.2023.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
This study describes the cell-free biomanufacturing of a broad-spectrum antiviral protein, griffithsin (GRFT) such that it can be produced in microgram quantities with consistent purity and potency in less than 24 h. We demonstrate GRFT production using two independent cell-free systems, one plant and one microbial. Griffithsin purity and quality were verified using standard regulatory metrics. Efficacy was demonstrated in vitro against SARS-CoV-2 and HIV-1 and was nearly identical to that of GRFT expressed in vivo. The proposed production process is efficient and can be readily scaled up and deployed wherever a viral pathogen might emerge. The current emergence of viral variants of SARS-CoV-2 has resulted in frequent updating of existing vaccines and loss of efficacy for front-line monoclonal antibody therapies. Proteins such as GRFT with its efficacious and broad virus neutralizing capability provide a compelling pandemic mitigation strategy to promptly suppress viral emergence at the source of an outbreak.
Collapse
Affiliation(s)
- Shayan G Borhani
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Max Z Levine
- Department of Chemical Engineering and Department of Bioengineering, Stanford University, Stanford, CA 94305-5025, USA
| | - Lauren H Krumpe
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jennifer Wilson
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Curtis J Henrich
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Barry R O'Keefe
- Molecular Targets Program, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA; Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD 21702, USA
| | - Donald C Lo
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - G Sitta Sittampalam
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Alexander G Godfrey
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - R Dwayne Lunsford
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Venkata Mangalampalli
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Aaron P Thole
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Douglas Frey
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - James Swartz
- Department of Chemical Engineering and Department of Bioengineering, Stanford University, Stanford, CA 94305-5025, USA
| | - Govind Rao
- Center for Advanced Sensor Technology, Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA; Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
20
|
Raman K, Rajagopal K, Swaminathan G, Jupudi S, Dhama K, Barua R, Emran TB, Osman H, Khandaker MU. A Critical Review on the Potency of Phytoconstituents in the Management of COVID-19. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2023; 17:1320-1340. [DOI: 10.22207/jpam.17.3.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Natural products and their derivatives have traditionally been used as a source of therapeutic agents. Their beneficial properties are due to large varieties in their chemical structures and biochemical actions. The discovery of natural products such as phytoconstituents have crucial role in the development of less toxic and more effective drugs. Phytoconstituents have shown to be beneficial in treating viral diseases such as the previous chikungunya virus, hepatitis C virus, SARS, and MERS viral diseases. Flavonoids, alkaloids, terpenoids, and other group of compounds combat against COVID-19 in several ways like by protease inhibition, spike protein inhibition, Nrf2 inhibition. The accumulation of NRF2 inhibits the development of the SARS-CoV-2 virus and stimulates anti-inflammatory action. The present review highlights the therapeutic importance of compounds isolated from medicinal plants and/or herbs, such as crude extracts of Curcumin I-III, Leptodactylone, Ginsenoside-Rb1, Lycorine, Reserpine, Saikosaponin B2, Cepharanthine, Withanoside V, Gingerol, Piperanine, chromans, flavonoids, Amentoflavone etc. against SARS-CoV-2. Natural products are typically safe, stable, and dependable source for finding drugs to control the current pandemic. Antiviral secondary metabolites many medicinal plants have given ingredients that were isolated. The selected plants based phytoconstituents may potentially be used against viruses’ development on anti-SARS-CoV-2 to offer a reference point in this field.
Collapse
|
21
|
Guan W, Zhang N, Bains A, Martinez A, LiWang PJ. Sustained Delivery of the Antiviral Protein Griffithsin and Its Adhesion to a Biological Surface by a Silk Fibroin Scaffold. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5547. [PMID: 37629837 PMCID: PMC10456748 DOI: 10.3390/ma16165547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/02/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023]
Abstract
The protein Griffithsin (Grft) is a lectin that tightly binds to high-mannose glycosylation sites on viral surfaces. This property allows Grft to potently inhibit many viruses, including HIV-1. The major route of HIV infection is through sexual activity, so an important tool for reducing the risk of infection would be a film that could be inserted vaginally or rectally to inhibit transmission of the virus. We have previously shown that silk fibroin can encapsulate, stabilize, and release various antiviral proteins, including Grft. However, for broad utility as a prevention method, it would be useful for an insertable film to adhere to the mucosal surface so that it remains for several days or weeks to provide longer-term protection from infection. We show here that silk fibroin can be formulated with adhesive properties using the nontoxic polymer hydroxypropyl methylcellulose (HPMC) and glycerol, and that the resulting silk scaffold can both adhere to biological surfaces and release Grft over the course of at least one week. This work advances the possible use of silk fibroin as an anti-viral insertable device to prevent infection by sexually transmitted viruses, including HIV-1.
Collapse
Affiliation(s)
- Wenyan Guan
- Materials and Biomaterials Science and Engineering, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Ning Zhang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China;
| | - Arjan Bains
- Chemistry and Biochemistry, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Airam Martinez
- Department of Bioengineering, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Patricia J. LiWang
- Molecular Cell Biology, Health Sciences Research Institute, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| |
Collapse
|
22
|
Nangarlia A, Hassen FF, Canziani G, Bandi P, Talukder C, Zhang F, Krauth D, Gary EN, Weiner DB, Bieniasz P, Navas-Martin S, O'Keefe BR, Ang CG, Chaiken I. Irreversible Inactivation of SARS-CoV-2 by Lectin Engagement with Two Glycan Clusters on the Spike Protein. Biochemistry 2023; 62:2115-2127. [PMID: 37341186 PMCID: PMC10663058 DOI: 10.1021/acs.biochem.3c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Host cell infection by SARS-CoV-2, similar to that by HIV-1, is driven by a conformationally metastable and highly glycosylated surface entry protein complex, and infection by these viruses has been shown to be inhibited by the mannose-specific lectins cyanovirin-N (CV-N) and griffithsin (GRFT). We discovered in this study that CV-N not only inhibits SARS-CoV-2 infection but also leads to irreversibly inactivated pseudovirus particles. The irreversibility effect was revealed by the observation that pseudoviruses first treated with CV-N and then washed to remove all soluble lectin did not recover infectivity. The infection inhibition of SARS-CoV-2 pseudovirus mutants with single-site glycan mutations in spike suggested that two glycan clusters in S1 are important for both CV-N and GRFT inhibition: one cluster associated with the RBD (receptor binding domain) and the second with the S1/S2 cleavage site. We observed lectin antiviral effects with several SARS-CoV-2 pseudovirus variants, including the recently emerged omicron, as well as a fully infectious coronavirus, therein reflecting the breadth of lectin antiviral function and the potential for pan-coronavirus inactivation. Mechanistically, observations made in this work indicate that multivalent lectin interaction with S1 glycans is likely a driver of the lectin infection inhibition and irreversible inactivation effect and suggest the possibility that lectin inactivation is caused by an irreversible conformational effect on spike. Overall, lectins' irreversible inactivation of SARS-CoV-2, taken with their breadth of function, reflects the therapeutic potential of multivalent lectins targeting the vulnerable metastable spike before host cell encounter.
Collapse
Affiliation(s)
- Aakansha Nangarlia
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19102, United States
| | - Farah Fazloon Hassen
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Gabriela Canziani
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Praneeta Bandi
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Choya Talukder
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Fengwen Zhang
- Laboratory of Retrovirology, The Rockefeller University, New York, New York 10065, United States
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10065, United States
| | - Douglas Krauth
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Ebony N Gary
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - David B Weiner
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Paul Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, New York 10065, United States
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10065, United States
| | - Sonia Navas-Martin
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
- Department of Microbiology and Immunology, Center for Molecular Virology & Translational Neuroscience, Institute for Molecular Medicine & Infectious Disease, Philadelphia, Pennsylvania 19102, United States
| | - Barry R O'Keefe
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland 21702, United States
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Charles G Ang
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Irwin Chaiken
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| |
Collapse
|
23
|
Guo L, Lin S, Chen Z, Cao Y, He B, Lu G. Targetable elements in SARS-CoV-2 S2 subunit for the design of pan-coronavirus fusion inhibitors and vaccines. Signal Transduct Target Ther 2023; 8:197. [PMID: 37164987 PMCID: PMC10170451 DOI: 10.1038/s41392-023-01472-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/04/2023] [Accepted: 04/23/2023] [Indexed: 05/12/2023] Open
Abstract
The ongoing global pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused devastating impacts on the public health and the global economy. Rapid viral antigenic evolution has led to the continual generation of new variants. Of special note is the recently expanding Omicron subvariants that are capable of immune evasion from most of the existing neutralizing antibodies (nAbs). This has posed new challenges for the prevention and treatment of COVID-19. Therefore, exploring broad-spectrum antiviral agents to combat the emerging variants is imperative. In sharp contrast to the massive accumulation of mutations within the SARS-CoV-2 receptor-binding domain (RBD), the S2 fusion subunit has remained highly conserved among variants. Hence, S2-based therapeutics may provide effective cross-protection against new SARS-CoV-2 variants. Here, we summarize the most recently developed broad-spectrum fusion inhibitors (e.g., nAbs, peptides, proteins, and small-molecule compounds) and candidate vaccines targeting the conserved elements in SARS-CoV-2 S2 subunit. The main focus includes all the targetable S2 elements, namely, the fusion peptide, stem helix, and heptad repeats 1 and 2 (HR1-HR2) bundle. Moreover, we provide a detailed summary of the characteristics and action-mechanisms for each class of cross-reactive fusion inhibitors, which should guide and promote future design of S2-based inhibitors and vaccines against new coronaviruses.
Collapse
Affiliation(s)
- Liyan Guo
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Sheng Lin
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zimin Chen
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yu Cao
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Disaster Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Bin He
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Guangwen Lu
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
24
|
Franzén Boger M, Benhach N, Hasselrot T, Brand RM, Rohan LC, Wang L, McGowan I, Edick S, Ho K, Meyn L, Matoba N, Palmer KE, Broliden K, Tjernlund A. A topical rectal douche product containing Q-Griffithsin does not disrupt the epithelial border or alter CD4 + cell distribution in the human rectal mucosa. Sci Rep 2023; 13:7547. [PMID: 37161022 PMCID: PMC10169179 DOI: 10.1038/s41598-023-34107-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/24/2023] [Indexed: 05/11/2023] Open
Abstract
To reduce HIV transmission, locally applied pre-exposure prophylaxis (PrEP) products for anorectal use will be important complements to oral and injectable PrEP products already available. It is critical to preserve an intact rectal epithelium and avoid an influx of mucosal HIV target cells with such product use. In this phase 1 clinical trial, we evaluated application of a topical rectal douche product containing Q-Griffithsin (Q-GRFT). Colorectal tissue samples were obtained via sigmoidoscopy at baseline, 1 and 24 h after single-dose exposure in 15 healthy volunteers. In situ staining for epithelial junction markers and CD4+ cells were assessed as an exploratory endpoint. A high-throughput, digitalized in situ imaging analysis workflow was developed to visualize and quantify these HIV susceptibility markers. We observed no significant differences in epithelial distribution of E-cadherin, desmocollin-2, occludin, claudin-1, or zonula occludens-1 when comparing the three timepoints or Q-GRFT versus placebo. There were also no differences in %CD4+ cells within the epithelium or lamina propria in any of these comparisons. In conclusion, the rectal epithelium and CD4+ cell distribution remained unchanged following topical application of Q-GRFT. In situ visualization of HIV susceptibility markers at mucosal sites could be useful to complement standard product safety assessments.
Collapse
Affiliation(s)
- Mathias Franzén Boger
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 64, Solna, Sweden.
| | - Nora Benhach
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 64, Solna, Sweden
| | - Tyra Hasselrot
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 64, Solna, Sweden
| | - Rhonda M Brand
- Magee Womens Research Institute, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa C Rohan
- Magee Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Lin Wang
- Magee Womens Research Institute, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Ian McGowan
- Magee Womens Research Institute, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Orion Biotechnology, Ottawa, Canada
| | - Stacey Edick
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ken Ho
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Leslie Meyn
- Magee Womens Research Institute, Pittsburgh, PA, USA
| | - Nobuyuki Matoba
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY, USA
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Kenneth E Palmer
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY, USA
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Kristina Broliden
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 64, Solna, Sweden
| | - Annelie Tjernlund
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 64, Solna, Sweden
| |
Collapse
|
25
|
Borhani SG, Levine MZ, Krumpe LH, Wilson J, Henrich CJ, O’Keefe BR, Lo D, Sittampalam GS, Godfrey AG, Lunsford RD, Mangalampalli V, Tao D, LeClair CA, Thole A, Frey D, Swartz J, Rao G. An approach to rapid distributed manufacturing of broad spectrum anti-viral griffithsin using cell-free systems to mitigate pandemics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.19.521044. [PMID: 36597541 PMCID: PMC9810220 DOI: 10.1101/2022.12.19.521044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study describes the cell-free biomanufacturing of a broad-spectrum antiviral protein, griffithsin (GRFT) such that it can be produced with consistent purity and potency in less than 24 hours. We demonstrate GRFT production using two independent cell-free systems, one plant and one microbial. Griffithsin purity and quality were verified using standard regulatory metrics. Efficacy was demonstrated in vitro against SARS-CoV-2 and HIV-1 and was nearly identical to that of GRFT expressed in vivo . The proposed production process is efficient and can be readily scaled up and deployed anywhere in the world where a viral pathogen might emerge. The current emergence of viral variants has resulted in frequent updating of existing vaccines and loss of efficacy for front-line monoclonal antibody therapies. Proteins such as GRFT with its efficacious and broad virus neutralizing capability provide a compelling pandemic mitigation strategy to promptly suppress viral emergence at the source of an outbreak.
Collapse
|
26
|
Zhao Y, Zhao N, Cai Y, Zhang H, Li J, Liu J, Ye C, Wang Y, Dang Y, Li W, Liu H, Zhang L, Li Y, Zhang L, Cheng L, Dong Y, Xu Z, Lei Y, Lu L, Wang Y, Ye W, Zhang F. An algal lectin griffithsin inhibits Hantaan virus infection in vitro and in vivo. Front Cell Infect Microbiol 2022; 12:881083. [PMID: 36579342 PMCID: PMC9791197 DOI: 10.3389/fcimb.2022.881083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022] Open
Abstract
Hantaan virus (HTNV) is the etiological pathogen of hemorrhagic fever with renal syndrome in East Asia. There are currently no effective therapeutics approved for HTNV and other hantavirus infections. We found that griffithsin (GRFT), an algae-derived lectin with broad-spectrum antiviral activity against various enveloped viruses, can inhibit the growth and spread of HTNV. In vitro experiments using recombinant vesicular stomatitis virus (rVSV) with HTNV glycoproteins as a model revealed that the GRFT inhibited the entry of rVSV-HTNV-G into host cells. In addition, we demonstrated that GRFT prevented authentic HTNV infection in vitro by binding to the viral N-glycans. In vivo experiments showed that GRFT partially protected the suckling mice from death induced by intracranial exposure to HTNV. These results demonstrated that GRFT can be a promising agent for inhibiting HTNV infection.
Collapse
Affiliation(s)
- Yajing Zhao
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China,Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Ningbo Zhao
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China,Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Yanxing Cai
- Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou, China,Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and BSL-3 Facility, Fudan University, Shanghai, China
| | - Hui Zhang
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Jia Li
- Department of Neurology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Jiaqi Liu
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, Airforce Medical University, Xi’an, Shaanxi, China
| | - Yuan Wang
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Yamei Dang
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Wanying Li
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China,Department of Pathogenic Biology, School of Preclinical Medicine, Yan’an University, Yan’an, Shaanxi, China
| | - He Liu
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Lianqing Zhang
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yuexiang Li
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Liang Zhang
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Linfeng Cheng
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Yangchao Dong
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Zhikai Xu
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Yingfeng Lei
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and BSL-3 Facility, Fudan University, Shanghai, China
| | - Yingjuan Wang
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China,*Correspondence: Fanglin Zhang, ; Wei Ye, ; Yingjuan Wang,
| | - Wei Ye
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China,*Correspondence: Fanglin Zhang, ; Wei Ye, ; Yingjuan Wang,
| | - Fanglin Zhang
- Department of Microbiology, School of Preclinical Medicine, Airforce Medical University, Xi’an, Shaanxi, China,*Correspondence: Fanglin Zhang, ; Wei Ye, ; Yingjuan Wang,
| |
Collapse
|
27
|
Özkul G, Kehribar EŞ, Ahan RE, Köksaldı İÇ, Özkul A, Dinç B, Aydoğan S, Şeker UÖŞ. A Genetically Engineered Biofilm Material for SARS-CoV-2 Capturing and Isolation. ADVANCED MATERIALS INTERFACES 2022; 9:2201126. [PMID: 36248312 PMCID: PMC9538133 DOI: 10.1002/admi.202201126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/10/2022] [Indexed: 06/16/2023]
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is continuously infecting people all around the world since its outbreak in 2019. Studies for numerous infection detection strategies are continuing. The sensitivity of detection methods is crucial to separate people with mild infections from people who are asymptomatic. In this sense, a strategy that would help to capture and isolate the SARS-CoV-2 virus prior to tests can be effective and beneficial. To this extent, genetically engineered biomaterials grounding from the biofilm protein of Escherichia coli are beneficial due to their robustness and adaptability to various application areas. Through functionalizing the E. coli biofilm protein, diverse properties can be attained such as enzyme display, nanoparticle production, and medical implant structures. Here, E. coli species are employed to express major curli protein CsgA and Griffithsin (GRFT) as fusion proteins, through a complex formation using SpyTag and SpyCatcher domains. In this study, a complex system with a CsgA scaffold harboring the affinity of GRFT against Spike protein to capture and isolate SARS-CoV-2 virus is successfully developed. It is shown that the hybrid recombinant protein can dramatically increase the sensitivity of currently available lateral flow assays for Sars-CoV-2 diagnostics.
Collapse
Affiliation(s)
- Gökçe Özkul
- UNAM ‐ Institute of Materials Science and NanotechnologyBilkent UniversityAnkara06800Turkey
| | - Ebru Şahin Kehribar
- UNAM ‐ Institute of Materials Science and NanotechnologyBilkent UniversityAnkara06800Turkey
| | - Recep Erdem Ahan
- UNAM ‐ Institute of Materials Science and NanotechnologyBilkent UniversityAnkara06800Turkey
| | - İlkay Çisil Köksaldı
- UNAM ‐ Institute of Materials Science and NanotechnologyBilkent UniversityAnkara06800Turkey
| | - Aykut Özkul
- Department of VirologyFaculty of Veterinary MedicineAnkara UniversityDışkapıAnkara06110Turkey
| | - Bedia Dinç
- Medical Microbiology Laboratory and Department of Clinical Microbiology and Infectious DiseasesAnkara Bilkent City HospitalHealth Sciences UniversityAnkara06800Turkey
| | - Sibel Aydoğan
- Medical Microbiology Laboratory and Department of Clinical Microbiology and Infectious DiseasesAnkara Bilkent City HospitalHealth Sciences UniversityAnkara06800Turkey
| | | |
Collapse
|
28
|
Bao Q, Yang T, Yang M, Mao C. Detection, prevention and treatment of COVID-19 and opportunities for nanobiotechnology. VIEW 2022; 3:20200181. [PMID: 35600668 PMCID: PMC9111118 DOI: 10.1002/viw.20200181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/12/2022] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
Since the outbreak of COVID-19, the number of confirmed cases and deaths has increased globally at a dramatic speed. In view of the serious health threat to humans, this review discusses the state-of-the-art studies about fighting this disease. It summarizes the current strategies and recent advances in detecting, preventing, and treating COVID-19 and interprets the underlying mechanisms in detail. Detection of COVID-19 can be successfully achieved by multiple techniques such as polymerase chain reaction, computed tomography imaging, and nano-biosensing. Inactivated virus vaccine, nucleic acid vaccine, and different nanoparticles have been employed to effectively prevent COVID-19. A variety of agents such as antiviral agents, neutralizing antibodies, and nanotherapeutics have been developed to treat COVID-19 with exciting efficacy. Although nanobiotechnology has shown great potential in the diagnosis, prevention, and treatment of COVID-19, efforts should be made to explore new biocompatible nano-biomaterials to advance this field to clinical applications. Hence, nanobiotechnology paves a new way to detect, prevent, and treat COVID-19 effectively.
Collapse
Affiliation(s)
- Qing Bao
- School of Materials Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Tao Yang
- School of Materials Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Mingying Yang
- College of Animal ScienceInstitute of Applied Bioresource ResearchZhejiang UniversityHangzhouZhejiangChina
| | - Chuanbin Mao
- School of Materials Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
- Department of Chemistry and BiochemistryUniversity of OklahomaNormanOklahomaUSA
| |
Collapse
|
29
|
Miller NL, Raman R, Clark T, Sasisekharan R. Complexity of Viral Epitope Surfaces as Evasive Targets for Vaccines and Therapeutic Antibodies. Front Immunol 2022; 13:904609. [PMID: 35784339 PMCID: PMC9247215 DOI: 10.3389/fimmu.2022.904609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
The dynamic interplay between virus and host plays out across many interacting surfaces as virus and host evolve continually in response to one another. In particular, epitope-paratope interactions (EPIs) between viral antigen and host antibodies drive much of this evolutionary race. In this review, we describe a series of recent studies examining aspects of epitope complexity that go beyond two interacting protein surfaces as EPIs are typically understood. To structure our discussion, we present a framework for understanding epitope complexity as a spectrum along a series of axes, focusing primarily on 1) epitope biochemical complexity (e.g., epitopes involving N-glycans) and 2) antigen conformational/dynamic complexity (e.g., epitopes with differential properties depending on antigen state or fold-axis). We highlight additional epitope complexity factors including epitope tertiary/quaternary structure, which contribute to epistatic relationships between epitope residues within- or adjacent-to a given epitope, as well as epitope overlap resulting from polyclonal antibody responses, which is relevant when assessing antigenic pressure against a given epitope. Finally, we discuss how these different forms of epitope complexity can limit EPI analyses and therapeutic antibody development, as well as recent efforts to overcome these limitations.
Collapse
Affiliation(s)
- Nathaniel L. Miller
- Harvard Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Rahul Raman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Thomas Clark
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ram Sasisekharan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
30
|
Konozy E, Osman M, Dirar A. Plant lectins as potent Anti-coronaviruses, Anti-inflammatory, antinociceptive and antiulcer agents. Saudi J Biol Sci 2022; 29:103301. [PMID: 35475119 PMCID: PMC9026953 DOI: 10.1016/j.sjbs.2022.103301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/12/2022] [Accepted: 04/17/2022] [Indexed: 12/14/2022] Open
Abstract
Lectins are defined as carbohydrate-binding proteins/glycoproteins of none immune origin, they are ubiquitous in nature, exist from bacteria to human cells. And due to their carbohydrate-binding recognition capacity, they have been a useful biological tool for the purification of glycoproteins and their subsequent characterization. Some plant lectins have also been revealed to own antinociceptive, antiulcer, and anti-inflammatory properties, where these features, in many instances, depending on the lectin carbohydrate-binding site. Coronavirus disease of 2019 (COVID-19) is a respiratory disease that struck the entire world leaving millions of people dead and more infected. Although COVID-19 vaccines have been made available, and quite a large number of world populations have already been immunized, the viral infection rates remained in acceleration, which continues to provoke major concern about the vaccines' efficacy. The belief in the ineffectiveness of the vaccine has been attributed in part to the recurrent mutations that occur in the epitope determinant fragments of the virus. Coronavirus envelope surface is extensively glycosylated being covered by more than sixty N-linked oligomannose, composite, and hybrid glycans with a core of Man3GlcNAc2Asn. In addition some O-linked glycans are also detected. Of these glyco-chains, many have also been exposed to several mutations, and a few remained conserved. Therefore, numerous plant lectins with a specificity directed towards these viral envelope sugars have been found to interact preferentially with them and are suggested to be scrutinized as a possible future tool to combat coronaviruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) through blocking the viral attachment to the host cells. In this review, we will discuss the possible applications of plant lectins as anti-coronaviruses including SARS-CoV-2, antinociceptive, anti-inflammatory, and antiulcer agents with the proposed mechanism of their actions.
Collapse
Affiliation(s)
- Emadeldin Konozy
- Department of Biotechnology, Africa City of Technology, Khartoum, Sudan
| | - Makarim Osman
- Department of Zoology, University of Khartoum, Khartoum, Sudan
| | - Amina Dirar
- Medicinal, Aromatic Plants and Traditional Medicine Research Institute (MAPTRI), National Center for Research, Mek Nimr Street, Khartoum, Sudan
| |
Collapse
|
31
|
Nabi-Afjadi M, Heydari M, Zalpoor H, Arman I, Sadoughi A, Sahami P, Aghazadeh S. Lectins and lectibodies: potential promising antiviral agents. Cell Mol Biol Lett 2022; 27:37. [PMID: 35562647 PMCID: PMC9100318 DOI: 10.1186/s11658-022-00338-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/21/2022] [Indexed: 12/30/2022] Open
Abstract
In nature, lectins are widely dispersed proteins that selectively recognize and bind to carbohydrates and glycoconjugates via reversible bonds at specific binding sites. Many viral diseases have been treated with lectins due to their wide range of structures, specificity for carbohydrates, and ability to bind carbohydrates. Through hemagglutination assays, these proteins can be detected interacting with various carbohydrates on the surface of cells and viral envelopes. This review discusses the most robust lectins and their rationally engineered versions, such as lectibodies, as antiviral proteins. Fusion of lectin and antibody’s crystallizable fragment (Fc) of immunoglobulin G (IgG) produces a molecule called a “lectibody” that can act as a carbohydrate-targeting antibody. Lectibodies can not only bind to the surface glycoproteins via their lectins and neutralize and clear viruses or infected cells by viruses but also perform Fc-mediated antibody effector functions. These functions include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), and antibody-dependent cell-mediated phagocytosis (ADCP). In addition to entering host cells, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein S1 binds to angiotensin-converting enzyme 2 (ACE2) and downregulates it and type I interferons in a way that may lead to lung disease. The SARS-CoV-2 spike protein S1 and human immunodeficiency virus (HIV) envelope are heavily glycosylated, which could make them a major target for developing vaccines, diagnostic tests, and therapeutic drugs. Lectibodies can lead to neutralization and clearance of viruses and cells infected by viruses by binding to glycans located on the envelope surface (e.g., the heavily glycosylated SARS-CoV-2 spike protein).
Collapse
Affiliation(s)
- Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Morteza Heydari
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 13145-1384, Iran
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,American Association of Kidney Patients, Tampa, FL, USA
| | - Ibrahim Arman
- Department of Molecular Biology and Genetics, Faculty of Sciences and Arts, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Arezoo Sadoughi
- Department of Immunology, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Parisa Sahami
- Medical Biology Research Center, Health Technologies Institute, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Safiyeh Aghazadeh
- Division of Biochemistry, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, 5756151818, Iran.
| |
Collapse
|
32
|
Decker JS, Menacho-Melgar R, Lynch MD. Integrated autolysis, DNA hydrolysis and precipitation enables an improved bioprocess for Q-Griffithsin, a broad-spectrum antiviral and clinical-stage anti-COVID-19 candidate. Biochem Eng J 2022; 181:108403. [PMID: 35308834 PMCID: PMC8917701 DOI: 10.1016/j.bej.2022.108403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 12/03/2022]
Abstract
Across the biomanufacturing industry, innovations are needed to improve efficiency and flexibility, especially in the face of challenges such as the COVID-19 pandemic. Here we report an improved bioprocess for Q-Griffithsin, a broad-spectrum antiviral currently in clinical trials for COVID-19. Q-Griffithsin is produced at high titer in E. coli and purified to anticipated clinical grade without conventional chromatography or the need for any fixed downstream equipment. The process is thus both low-cost and highly flexible, facilitating low sales prices and agile modifications of production capacity, two key features for pandemic response. The simplicity of this process is enabled by a novel unit operation that integrates cellular autolysis, autohydrolysis of nucleic acids, and contaminant precipitation, giving essentially complete removal of host cell DNA as well as reducing host cell proteins and endotoxin by 3.6 and 2.4 log10 units, respectively. This unit operation can be performed rapidly and in the fermentation vessel, such that Q-GRFT is obtained with 100% yield and > 99.9% purity immediately after fermentation and requires only a flow-through membrane chromatography step for further contaminant removal. Using this operation or variations of it may enable improved bioprocesses for a range of other high-value proteins in E. coli.
Collapse
Affiliation(s)
- John S Decker
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Michael D Lynch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
33
|
Chandel S, Joon A, Ghosh S. Enteroaggregative Escherichia coli induces altered glycosylation in membrane proteins of cultured human intestinal epithelial cells. Biochimie 2022; 199:68-80. [DOI: 10.1016/j.biochi.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/11/2022] [Accepted: 04/05/2022] [Indexed: 11/02/2022]
|
34
|
Algal Metabolites Can Be an Immune Booster against COVID-19 Pandemic. Antioxidants (Basel) 2022; 11:antiox11030452. [PMID: 35326102 PMCID: PMC8944855 DOI: 10.3390/antiox11030452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/05/2023] Open
Abstract
The world has faced the challenges of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) for the last two years, first diagnosed at the end of 2019 in Wuhan and widely distributed worldwide. As a result, the WHO has proclaimed the illness brought on by this virus to be a global pandemic. To combat COVID-19, researcher communities continuously develop and implement rapid diagnoses, safe and effective vaccinations and other alternative therapeutic procedures. However, synthetic drug-related side effects and high costs have piqued scientists’ interest in natural product-based therapies and medicines. In this regard, antiviral substances derived from natural resources and some medicines have seen a boom in popularity. For instance, algae are a rich source of compounds such as lectins and sulfated polysaccharides, which have potent antiviral and immunity-boosting properties. Moreover, Algae-derived compounds or metabolites can be used as antibodies and vaccine raw materials against COVID-19. Furthermore, some algal species can boost immunity, reduce viral activity in humans and be recommended for usage as a COVID-19 preventative measure. However, this field of study is still in its early stages of development. Therefore, this review addresses critical characteristics of algal metabolites, their antioxidant potential and therapeutic potential in COVID-19.
Collapse
|
35
|
Zeng W, Ren J, Li Z, Jiang C, Sun Q, Li C, Li W, Li W, He Q. Levistolide A Inhibits PEDV Replication via Inducing ROS Generation. Viruses 2022; 14:v14020258. [PMID: 35215851 PMCID: PMC8878026 DOI: 10.3390/v14020258] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) variant strains adversely affect the production of pigs globally. Vaccines derived from PEDV traditional strains impart less protection against the variant strains. Moreover, sequence diversity among different PEDV variant strains is also complicated. This necessitates developing alternative antiviral strategies for defending against PEDV. This study explored a natural product, Levistolide A (LA), to possess antiviral activity against PEDV. LA was found to suppress PEDV replication in a dose-dependent manner. And the inhibitory effect of LA against PEDV was maintained in the course of time. In terms of viral RNA and protein production, LA also showed a strong inhibitory effect. In addition, LA was indicated to inhibit PEDV from attaching to the cellular membrane or penetrating the cells. Further study revealed that LA can induce the generation of reactive oxygen species (ROS), and the corresponding inhibitor, NAC, was found to antagonize the effect of LA on inhibiting PEDV replication. This illustrated that the LA-induced ROS generation played an important role in its anti-PEDV activity. LA was also identified to stimulate ER stress, which is an important consequence of ROS production and was proven to be able to inhibit PEDV replication. To conclude, this study revealed that LA can inhibit PEDV replication via inducing ROS generation.
Collapse
Affiliation(s)
- Wei Zeng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Jingping Ren
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhonghua Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China;
| | - Changsheng Jiang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Qi Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Chang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Wan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (C.J.); (Q.S.); (C.L.); (W.L.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
36
|
Seaweed-Derived Proteins and Peptides: Promising Marine Bioactives. Antioxidants (Basel) 2022; 11:antiox11010176. [PMID: 35052680 PMCID: PMC8773382 DOI: 10.3390/antiox11010176] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/29/2022] Open
Abstract
Seaweeds are a typical food of East-Asian cuisine, to which are alleged several beneficial health effects have been attributed. Their availability and their nutritional and chemical composition have favored the increase in its consumption worldwide, as well as a focus of research due to their bioactive properties. In this regard, seaweed proteins are nutritionally valuable and comprise several specific enzymes, glycoproteins, cell wall-attached proteins, red algae phycobiliproteins, lectins, peptides, or mycosporine-like amino acids. This great extent of molecules has been reported to exert significant antioxidant, antimicrobial, anti-inflammatory, antihypertensive, antidiabetic, or antitumoral properties. Hence, knowledge on algae proteins and derived compounds have gained special interest for the potential nutraceutical, cosmetic or pharmaceutical industries based on these bioactivities. Although several molecular mechanisms of action on how these proteins and peptides exert biological activities have been described, many gaps in knowledge still need to be filled. Updating the current knowledge related to seaweed proteins and peptides is of interest to further asses their potential health benefits. This review addresses the characteristics of seaweed protein and protein-derived molecules, their natural occurrence, their studied bioactive properties, and their described potential mechanisms of action.
Collapse
|
37
|
Reis JG, Cadamuro RD, Cabral AC, Thaís da Silva I, Rodríguez-Lázaro D, Fongaro G. Broad Spectrum Algae Compounds Against Viruses. Front Microbiol 2022; 12:809296. [PMID: 35095816 PMCID: PMC8795700 DOI: 10.3389/fmicb.2021.809296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/24/2021] [Indexed: 12/24/2022] Open
Abstract
The pharmaceutical industry is currently trying to develop new bioactive compounds to inactivate both enveloped and non-enveloped viruses for therapeutic purposes. Consequently, microalgal and macroalgal bioactive compounds are being explored by pharmaceutical, as well as biotechnology and food industries. In this review, we show how compounds produced by algae include important candidates for viral control applications. We discuss their mechanisms of action and activity against enveloped and non-enveloped viruses, including those causing infections by enteric, parenteral, and respiratory routes. Indeed, algal products have potential in human and animal medicine.
Collapse
Affiliation(s)
- Jacqueline Graff Reis
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Rafael Dorighello Cadamuro
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Ariadne Cristiane Cabral
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Dentistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Izabella Thaís da Silva
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - David Rodríguez-Lázaro
- Microbiology Division, Faculty of Sciences, University of Burgos, Burgos, Spain
- Research Centre for Emerging Pathogens and Global Health, University of Burgos, Burgos, Spain
| | - Gislaine Fongaro
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis, Brazil
- *Correspondence: Gislaine Fongaro,
| |
Collapse
|
38
|
Decker JS, Menacho-Melgar R, Lynch MD. Integrated Autolysis, DNA Hydrolysis and Precipitation Enables an Improved Bioprocess for Q-Griffithsin, a Broad-Spectrum Antiviral and Clinical-Stage anti-COVID-19 Candidate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.12.30.474602. [PMID: 35018377 PMCID: PMC8750652 DOI: 10.1101/2021.12.30.474602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
UNLABELLED Across the biomanufacturing industry, innovations are needed to improve efficiency and flexibility, especially in the face of challenges such as the COVID-19 pandemic. Here we report an improved bioprocess for Q-Griffithsin, a broad-spectrum antiviral currently in clinical trials for COVID-19. Q-Griffithsin is produced at high titer in E. coli and purified to anticipated clinical grade without conventional chromatography or the need for any fixed downstream equipment. The process is thus both low-cost and highly flexible, facilitating low sales prices and agile modifications of production capacity, two key features for pandemic response. The simplicity of this process is enabled by a novel unit operation that integrates cellular autolysis, autohydrolysis of nucleic acids, and contaminant precipitation, giving essentially complete removal of host cell DNA as well as reducing host cell proteins and endotoxin by 3.6 and 2.4 log 10 units, respectively. This unit operation can be performed rapidly and in the fermentation vessel, such that Q-GRFT is obtained with 100% yield and >99.9% purity immediately after fermentation and requires only a flow-through membrane chromatography step for further contaminant removal. Using this operation or variations of it may enable improved bioprocesses for a range of other high-value proteins in E. coli . HIGHLIGHTS Integrating autolysis, DNA hydrolysis and precipitation enables process simplificationAutolysis reduces endotoxin release and burden to purificationQ-Griffithsin recovered from fermentation vessel at >99.9% purity and 100% yieldQ-Griffithsin purified to anticipated clinical grade without conventional chromatographyThe resulting bioprocess is 100% disposables-compatible, scalable, and low-cost.
Collapse
|
39
|
Algal and Cyanobacterial Lectins and Their Antimicrobial Properties. Mar Drugs 2021; 19:md19120687. [PMID: 34940686 PMCID: PMC8707200 DOI: 10.3390/md19120687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
Lectins are proteins with a remarkably high affinity and specificity for carbohydrates. Many organisms naturally produce them, including animals, plants, fungi, protists, bacteria, archaea, and viruses. The present report focuses on lectins produced by marine or freshwater organisms, in particular algae and cyanobacteria. We explore their structure, function, classification, and antimicrobial properties. Furthermore, we look at the expression of lectins in heterologous systems and the current research on the preclinical and clinical evaluation of these fascinating molecules. The further development of these molecules might positively impact human health, particularly the prevention or treatment of diseases caused by pathogens such as human immunodeficiency virus, influenza, and severe acute respiratory coronaviruses, among others.
Collapse
|
40
|
Islam F, Bibi S, Meem AFK, Islam MM, Rahaman MS, Bepary S, Rahman MM, Rahman MM, Elzaki A, Kajoak S, Osman H, ElSamani M, Khandaker MU, Idris AM, Emran TB. Natural Bioactive Molecules: An Alternative Approach to the Treatment and Control of COVID-19. Int J Mol Sci 2021; 22:12638. [PMID: 34884440 PMCID: PMC8658031 DOI: 10.3390/ijms222312638] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 02/07/2023] Open
Abstract
Several coronaviruses (CoVs) have been associated with serious health hazards in recent decades, resulting in the deaths of thousands around the globe. The recent coronavirus pandemic has emphasized the importance of discovering novel and effective antiviral medicines as quickly as possible to prevent more loss of human lives. Positive-sense RNA viruses with group spikes protruding from their surfaces and an abnormally large RNA genome enclose CoVs. CoVs have already been related to a range of respiratory infectious diseases possibly fatal to humans, such as MERS, SARS, and the current COVID-19 outbreak. As a result, effective prevention, treatment, and medications against human coronavirus (HCoV) is urgently needed. In recent years, many natural substances have been discovered with a variety of biological significance, including antiviral properties. Throughout this work, we reviewed a wide range of natural substances that interrupt the life cycles for MERS and SARS, as well as their potential application in the treatment of COVID-19.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Shabana Bibi
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming 650091, China;
- International Joint Research Center for Sustainable Utilization of Cordyceps Bioresources in China and Southeast Asia, Yunnan University, Kunming 650091, China
| | - Atkia Farzana Khan Meem
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Md. Mohaimenul Islam
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Md. Saidur Rahaman
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Sristy Bepary
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Md. Mizanur Rahman
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Md. Mominur Rahman
- Department of Pharmacy, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (A.F.K.M.); (M.M.I.); (M.S.R.); (S.B.); (M.M.R.); (M.M.R.)
| | - Amin Elzaki
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.E.); (S.K.); (H.O.); (M.E.)
| | - Samih Kajoak
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.E.); (S.K.); (H.O.); (M.E.)
| | - Hamid Osman
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.E.); (S.K.); (H.O.); (M.E.)
| | - Mohamed ElSamani
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia; (A.E.); (S.K.); (H.O.); (M.E.)
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Selangor, Malaysia;
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha 62529, Saudi Arabia;
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62529, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|
41
|
Cazorla-Luna R, Ruiz-Caro R, Veiga MD, Malcolm RK, Lamprou DA. Recent advances in electrospun nanofiber vaginal formulations for women's sexual and reproductive health. Int J Pharm 2021; 607:121040. [PMID: 34450222 DOI: 10.1016/j.ijpharm.2021.121040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/17/2021] [Accepted: 08/21/2021] [Indexed: 12/22/2022]
Abstract
Electrospinning is an innovative technique that allows production of nanofibers and microfibers by applying a high voltage to polymer solutions of melts. The properties of these fibers - which include high surface area, high drug loading capacity, and ability to be manufactured from mucoadhesive polymers - may be particularly useful in a myriad of drug delivery and tissue engineering applications. The last decade has witnessed a surge of interest in the application of electrospinning technology for the fabrication of vaginal drug delivery systems for the treatment and prevention of diseases associated with women's sexual and reproductive health, including sexually transmitted infections (e.g. infection with human immunodeficiency virus and herpes simplex virus) vaginitis, preterm birth, contraception, multipurpose prevention technology strategies, cervicovaginal cancer, and general maintenance of vaginal health. Due to their excellent mechanical properties, electrospun scaffolds are also being investigated as next-generation materials in the surgical treatment of pelvic organ prolapse. In this article, we review the latest advances in the field.
Collapse
Affiliation(s)
- Raúl Cazorla-Luna
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Departamento de Farmacia Galénica y Tecnología Alimentaria, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Roberto Ruiz-Caro
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María-Dolores Veiga
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - R Karl Malcolm
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | - Dimitrios A Lamprou
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
42
|
Choudhary P, G VS, Khade M, Savant S, Musale A, G RKK, Chelliah MS, Dasgupta S. Empowering blue economy: From underrated ecosystem to sustainable industry. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2021; 291:112697. [PMID: 33934021 DOI: 10.1016/j.jenvman.2021.112697] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 06/12/2023]
Abstract
With increasing demand for resources to achieve global food-water-energy nexus and rapid decline in land-based sources, oceans represent both solution and boost to sustainable environment and economy. In addition to fundamental part of earth's ecosystem for uncatalogued diversity of life, oceans are undervalued economy powerhouse with gross marine product value. With sustainable management of existing assets including shipping, transportation, manufacturing, fisheries, tourism and exploration of new business like marine biotechnology and renewable energy, the ocean or blue economy has potential to fulfill sustainable development goals (SDG). In spite of recognition of blue economy as a new economic frontier, investments by existing industries and emergence of new ones are limited and less known, hence require more in depth attention and scientific understanding. In the present study, authors present a systematic comparative assessment of blue economy sectors with distinct challenges and strategies to be further explored and implemented for industrial deployment. The conceptualization of integrated routes of bio(economy) by the current study can act as gateway for key stakeholders, i.e. governance, bluepreneurs (scientists and industries) to prioritize technologies for sustainable applications of marine resources.
Collapse
Affiliation(s)
- Poonam Choudhary
- RIL Biofuel R&D Site, Reliance Industries Limited, Motikhavadi, Jamnagar, India.
| | - Venkata Subhash G
- Reliance Technology Group, Reliance Industries Limited, Reliance Corporate Park, Ghansoli, Thane- Belapur Road, Navi Mumbai, 400701, India.
| | - Monika Khade
- Reliance Technology Group, Reliance Industries Limited, Reliance Corporate Park, Ghansoli, Thane- Belapur Road, Navi Mumbai, 400701, India.
| | - Sandip Savant
- RIL Biofuel R&D Site, Reliance Industries Limited, Motikhavadi, Jamnagar, India.
| | - Amar Musale
- Reliance Technology Group, Reliance Industries Limited, Reliance Corporate Park, Ghansoli, Thane- Belapur Road, Navi Mumbai, 400701, India.
| | - Raja Krishna Kumar G
- Reliance Technology Group, Reliance Industries Limited, Reliance Corporate Park, Ghansoli, Thane- Belapur Road, Navi Mumbai, 400701, India.
| | | | - Santanu Dasgupta
- Reliance Technology Group, Reliance Industries Limited, Reliance Corporate Park, Ghansoli, Thane- Belapur Road, Navi Mumbai, 400701, India.
| |
Collapse
|
43
|
Nile SH, Nile A, Jalde S, Kai G. Recent advances in potential drug therapies combating COVID-19 and related coronaviruses-A perspective. Food Chem Toxicol 2021; 154:112333. [PMID: 34118347 PMCID: PMC8189744 DOI: 10.1016/j.fct.2021.112333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Coronaviruses (CoVs) are a large family of viruses responsible for the severe pathophysiological effects on human health. The most severe outbreak includes Severe Acute Respiratory Syndrome (SARS-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Coronavirus disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2). The COVID-19 poses major challenges to clinical management because no specific FDA-approved therapy yet to be available. Thus, the existing therapies are being used for the treatment of COVID-19, which are under clinical trials and compassionate use, based on in vitro and in silico studies. In this review, we summarize the potential therapies utilizing small molecules, bioactive compounds, nucleoside and nucleotide analogs, peptides, antibodies, natural products, and synthetic compounds targeting the complex molecular signaling network involved in COVID-19. In this review>230 natural and chemically synthesized drug therapies are described with their recent advances in research and development being done in terms of their chemical, structural and functional properties. This review focuses on possible targets for viral cells, viral proteins, viral replication, and different molecular pathways for the discovery of novel viral- and host-based therapeutic targets against SARS-CoV-2.
Collapse
Affiliation(s)
- Shivraj Hariram Nile
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Arti Nile
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, South Korea
| | - Shivkumar Jalde
- Department of Medicinal Chemistry, Jungwon University, Goesan, 28420, South Korea
| | - Guoyin Kai
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
44
|
Gupta A, Gupta GS. Status of mannose-binding lectin (MBL) and complement system in COVID-19 patients and therapeutic applications of antiviral plant MBLs. Mol Cell Biochem 2021; 476:2917-2942. [PMID: 33745077 PMCID: PMC7981598 DOI: 10.1007/s11010-021-04107-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by a virus called "Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)." In the majority of patients, infection with COVID-19 may be asymptomatic or may cause only mild symptoms. However, in some patients, there can also be immunological problems, such as macrophage activation syndrome (CSS) that results in cytokine storm syndrome (CSS) and acute respiratory distress syndrome (ARDS). Comprehension of host-microbe communications is the critical aspect in the advancement of new therapeutics against infectious illnesses. Endogenous animal lectins, a class of proteins, may perceive non-self glycans found on microorganisms. Serum mannose-binding lectin (sMBL), as a part of the innate immune framework, recognizes a wide range of microbial microorganisms and activates complement cascade via an antibody-independent pathway. Although the molecular basis for the intensity of SARS-CoV-2 infection is not generally understood, scientific literature indicates that COVID-19 is correlated with unregulated activation of the complement in terms of disease severity. Disseminated intravascular coagulation (DIC), inflammation, and immune paralysis contribute to unregulated complement activation. Pre-existing genetic defects in MBL and their association with complement play a major role in immune response dysregulation caused by SARS-CoV-2. In order to generate anti-complement-based therapies in Covid-19, an understanding of sMBL in immune response to SARS-CoV-2 and complement is therefore essential. This review highlights the role of endogenous sMBL and complement activation during SARS-CoV-2 infection and their therapeutic management by various agents, mainly plant lectins, since antiviral mannose-binding plant lectins (pMBLs) offer potential applications in the prevention and control of viral infections.
Collapse
Affiliation(s)
- Anita Gupta
- Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, India
| | - G S Gupta
- Department of Biophysics, Sector 25, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
45
|
Alsaidi S, Cornejal N, Mahoney O, Melo C, Verma N, Bonnaire T, Chang T, O’Keefe BR, Sailer J, Zydowsky TM, Teleshova N, Romero JAF. Griffithsin and Carrageenan Combination Results in Antiviral Synergy against SARS-CoV-1 and 2 in a Pseudoviral Model. Mar Drugs 2021; 19:md19080418. [PMID: 34436255 PMCID: PMC8400000 DOI: 10.3390/md19080418] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/15/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Over 182 million confirmed cases of COVID-19 and more than 4 million deaths have been reported to date around the world. It is essential to identify broad-spectrum antiviral agents that may prevent or treat infections by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) but also by other coronaviruses that may jump the species barrier in the future. We evaluated the antiviral selectivity of griffithsin and sulfated and non-sulfated polysaccharides against SARS-CoV-1 and SARS-CoV-2 using a cytotoxicity assay and a cell-based pseudoviral model. The half-maximal cytotoxic concentration (CC50) and half-maximal effective concentration (EC50) were determined for each compound, using a dose-response-inhibition analysis on GraphPad Prism v9.0.2 software (San Diego, CA, USA). The therapeutic index (TI = CC50/EC50) was calculated for each compound. The potential synergistic, additive, or antagonistic effect of different compound combinations was determined by CalcuSyn v1 software (Biosoft, Cambridge, UK), which estimated the combination index (CI) values. Iota and lambda carrageenan showed the most potent antiviral activity (EC50 between 3.2 and 7.5 µg/mL). Carrageenan and griffithsin combinations exhibited synergistic activity (EC50 between 0.2 and 3.8 µg/mL; combination index <1), including against recent SARS-CoV-2 mutations. The griffithsin and carrageenan combination is a promising candidate to prevent or treat infections by SARS-CoV-1 and SARS-CoV-2.
Collapse
Affiliation(s)
- Sahar Alsaidi
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Department of Anthropology, Lehman College, The City University of New York, New York, NY 10468, USA
| | - Nadjet Cornejal
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Center for Achievement in Science Education, Department of Biology and Chemistry, School of Natural and Behavioral Sciences, Brooklyn College, The City University of New York, New York, NY 11210, USA
| | - Oneil Mahoney
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
| | - Claudia Melo
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Center for Achievement in Science Education, Department of Biology and Chemistry, School of Natural and Behavioral Sciences, Brooklyn College, The City University of New York, New York, NY 11210, USA
| | - Neeharika Verma
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Thierry Bonnaire
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Theresa Chang
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA;
| | - Barry R. O’Keefe
- Natural Products Branch, Molecular Targets Program, Developmental Therapeutics Program, Center for Cancer Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD 21702, USA;
| | - James Sailer
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Thomas M. Zydowsky
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - Natalia Teleshova
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
| | - José A. Fernández Romero
- Population Council, New York, NY 10065, USA; (S.A.); (N.C.); (O.M.); (C.M.); (N.V.); (T.B.); (J.S.); (T.M.Z.); (N.T.)
- Department of Science, Borough of Manhattan Community College, The City University of New York, New York, NY 10007, USA
- Correspondence:
| |
Collapse
|
46
|
Cai Y, Xu W, Tang J, Cao N, Lan Q, Lu L, Jiang S. A bivalent protein targeting glycans and HR1 domain in spike protein potently inhibited infection of SARS-CoV-2 and other human coronaviruses. Cell Biosci 2021; 11:128. [PMID: 34238357 PMCID: PMC8264481 DOI: 10.1186/s13578-021-00638-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background Our previous studies have shown that combining the antiviral lectin GRFT and the pan-CoV fusion inhibitory peptide EK1 results in highly potent inhibitory activity against SARS-CoV-2 infection. In this study, we aimed to design and construct a bivalent protein consisting of GRFT and EK1 components and evaluate its inhibitory activity and mechanism of action against infection by SARS-CoV-2 and its mutants, as well as other human coronaviruses (HCoVs). Methods The bivalent proteins were expressed in E. coli and purified with Ni-NTA column. HIV backbone-based pseudovirus (PsV) infection and HCoV S-mediated cell–cell fusion assays were performed to test their inhibitory activity. ELISA and Native-PAGE were conducted to illustrate the mechanism of action of these bivalent proteins. Five-day-old newborn mice were intranasally administrated with a selected bivalent protein before or after HCoV-OC43 challenge, and its protective effect was monitored for 14 days. Results Among the three bivalent proteins purified, GL25E exhibited the most potent inhibitory activity against infection of SARS-CoV-2 PsVs expressing wild-type and mutated S protein. GL25E was significantly more effective than GRFT and EK1 alone in inhibiting HCoV S-mediated cell–cell fusion, as well as infection by SARS-CoV-2 and other HCoVs, including SARS-CoV, MERS-CoV, HCoV-229E, HCoV-NL63 and HCoV-OC43. GL25E could inhibit authentic SASR-CoV-2, HCoV-OC43 and HCoV-229E infection in vitro and prevent newborn mice from authentic HCoV-OC43 infection in vivo. GL25E could bind to glycans in the S1 subunit and HR1 in the S2 subunit in S protein, showing a mechanism of action similar to that of GRFT and EK1 alone. Conclusions Since GL25E showed highly potent and broad-spectrum inhibitory activity against infection of SARS-CoV-2 and its mutants, as well as other HCoVs, it is a promising candidate for further development as a broad-spectrum anti-HCoV therapeutic and prophylactic to treat and prevent COVID-19 and other emerging HCoV diseases.
Collapse
Affiliation(s)
- Yanxing Cai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Jiayi Tang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Najing Cao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Qiaoshuai Lan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences and Biosafety Level 3 Laboratory, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
47
|
Musarra-Pizzo M, Pennisi R, Ben-Amor I, Mandalari G, Sciortino MT. Antiviral Activity Exerted by Natural Products against Human Viruses. Viruses 2021; 13:v13050828. [PMID: 34064347 PMCID: PMC8147851 DOI: 10.3390/v13050828] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022] Open
Abstract
Viral infections are responsible for several chronic and acute diseases in both humans and animals. Despite the incredible progress in human medicine, several viral diseases, such as acquired immunodeficiency syndrome, respiratory syndromes, and hepatitis, are still associated with high morbidity and mortality rates in humans. Natural products from plants or other organisms are a rich source of structurally novel chemical compounds including antivirals. Indeed, in traditional medicine, many pathological conditions have been treated using plant-derived medicines. Thus, the identification of novel alternative antiviral agents is of critical importance. In this review, we summarize novel phytochemicals with antiviral activity against human viruses and their potential application in treating or preventing viral disease.
Collapse
Affiliation(s)
- Maria Musarra-Pizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
| | - Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Shenzhen International Institute for Biomedical Research, 1301 Guanguang Rd. 3F Building 1-B, Silver Star Hi-Tech Park Longhua District, Shenzhen 518116, China
| | - Ichrak Ben-Amor
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Unit of Biotechnology and Pathologies, Higher Institute of Biotechnology of Sfax, University of Sfax, Sfax 3029, Tunisia
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Correspondence: (G.M.); (M.T.S.); Tel.: +39-090-6767-5217 (G.M. & M.T.S.)
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale SS. Annunziata, 98168 Messina, Italy; (M.M.-P.); (R.P.); (I.B.-A.)
- Correspondence: (G.M.); (M.T.S.); Tel.: +39-090-6767-5217 (G.M. & M.T.S.)
| |
Collapse
|
48
|
Chakravarti R, Singh R, Ghosh A, Dey D, Sharma P, Velayutham R, Roy S, Ghosh D. A review on potential of natural products in the management of COVID-19. RSC Adv 2021; 11:16711-16735. [PMID: 35479175 PMCID: PMC9031656 DOI: 10.1039/d1ra00644d] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
At the end of 2019, a life threatening viral infection (COVID-19) caused by a novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) was reported. This virus has spread worldwide in a short duration and forced the world to face unprecedented life and economic loss. To date, there are no known specific drugs to combat this virus and the process for new drug development is lengthy. Most promising candidates, which emerged as potential leads, were abandoned in the later phases of clinical trials. Repurposing of already approved drugs for other therapeutic applications can be done only after extensive testing for safety and efficacy. With no definite therapeutics in the horizon, natural products are in extensive use arbitrarily as anti-viral agents and immune boosters. For ages it has been known that most natural products possess potent anti-viral activity and it is no different for SARS-CoV-2. It has been shown that natural products display inhibitory effects on MERS-CoV and SARS-CoV infections. In silico studies have shown that various natural products have strong binding affinity for and inhibitory action on the non-structural proteins of the virus, namely PLPRO, MPRO, and RdRp, and structural proteins such as spike (S) protein. Since the virus utilizes the transmembrane ACE2 receptor of the host cell, it also proves to be a valid target for drug development. In this review promising targets for drug development against SARS-CoV-2 and anti-viral activities of some of the known natural products are discussed. In this review promising targets for drug development against SARS-CoV-2 and anti-viral activities of some of the known natural products (including plant secondary metabolites) are discussed.![]()
Collapse
Affiliation(s)
- Rudra Chakravarti
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata India
| | - Rajveer Singh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata India
| | - Arijit Ghosh
- Department of Chemistry, University of Calcutta Kolkata India
| | - Dhritiman Dey
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata India
| | - Priyanka Sharma
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata India
| | - Ravichandiran Velayutham
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata India
| | - Syamal Roy
- CSIR-Indian Institute of Chemical Biology Jadavpur Kolkata India
| | - Dipanjan Ghosh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata India
| |
Collapse
|
49
|
Crakes KR, Herrera C, Morgan JL, Olstad K, Hessell AJ, Ziprin P, LiWang PJ, Dandekar S. Efficacy of silk fibroin biomaterial vehicle for in vivo mucosal delivery of Griffithsin and protection against HIV and SHIV infection ex vivo. J Int AIDS Soc 2021; 23:e25628. [PMID: 33073530 PMCID: PMC7569169 DOI: 10.1002/jia2.25628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Introduction The majority of new HIV infections occur through mucosal transmission. The availability of readily applicable and accessible platforms for anti‐retroviral (ARV) delivery is critical for the prevention of HIV acquisition through sexual transmission in both women and men. There is a compelling need for developing new topical delivery systems that have advantages over the pills, gels and rings, which currently fail to guarantee protection against mucosal viral transmission in vulnerable populations due to lack of user compliance. The silk fibroin (SF) platform offers another option that may be better suited to individual circumstances and preferences to increase efficacy through user compliance. The objective of this study was to test safety and efficacy of SF for anti‐HIV drug delivery to mucosal sites and for viral prevention. Methods We formulated a potent HIV inhibitor Griffithsin (Grft) in a mucoadhesive silk fibroin (SF) drug delivery platform and tested the application in a non‐human primate model in vivo and a pre‐clinical human cervical and colorectal tissue explant model. Both vaginal and rectal compartments were assessed in rhesus macaques (Mucaca mulatta) that received SF (n = 4), no SF (n = 7) and SF‐Grft (n = 11). In this study, we evaluated the composition of local microbiota, inflammatory cytokine production, histopathological changes in the vaginal and rectal compartments and mucosal protection after ex vivo SHIV challenge. Results Effective Grft release and retention in mucosal tissues from the SF‐Grft platform resulted in protection against HIV in human cervical and colorectal tissue as well as against SHIV challenge in both rhesus macaque vaginal and rectal tissues. Mucoadhesion of SF‐Grft inserts did not cause any inflammatory responses or changes in local microbiota. Conclusions We demonstrated that in vivo delivery of SF‐Grft in rhesus macaques fully protects against SHIV challenge ex vivo after two hours of application and is safe to use in both the vaginal and rectal compartments. Our study provides support for the development of silk fibroin as a highly promising, user‐friendly HIV prevention modality to address the global disparity in HIV infection.
Collapse
Affiliation(s)
- Katti R Crakes
- Department of Medical Microbiology & Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Carolina Herrera
- Department of Medicine, St. Mary's Campus Imperial College, London, United Kingdom
| | - Jessica L Morgan
- Department of Molecular Cell Biology, University of California Merced, Merced, CA, USA
| | - Katie Olstad
- California National Primate Research Center, University of California Davis, Davis, CA, USA
| | - Ann J Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, USA
| | - Paul Ziprin
- Department of Surgery and Cancer, St. Mary's Campus Imperial College, London, United Kingdom
| | - Patricia J LiWang
- Department of Molecular Cell Biology, University of California Merced, Merced, CA, USA
| | - Satya Dandekar
- Department of Medical Microbiology & Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
50
|
Sivaji N, Harish N, Singh S, Singh A, Vijayan M, Surolia A. Mevo lectin specificity towards high-mannose structures with terminal αMan(1,2)αMan residues and its implication to inhibition of the entry of Mycobacterium tuberculosis into macrophages. Glycobiology 2021; 31:1046-1059. [PMID: 33822039 DOI: 10.1093/glycob/cwab022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/20/2022] Open
Abstract
Mannose-binding lectins can specifically recognize and bind complex glycan structures on pathogens and have potential as anti-viral and anti-bacterial agents. We previously reported the structure of a lectin from an archaeal species, Mevo lectin, which has specificity towards terminal α1,2 linked manno-oligosaccharides. Mycobacterium tuberculosis (M. tuberculosis) expresses mannosylated structures including, lipoarabinomannan (ManLAM) on its surface and exploits C-type lectins to gain entry into the host cells. ManLAM structure has mannose capping with terminal αMan(1,2)αMan residues and is important for recognition by innate immune cells. Here, we aim to address the specificity of Mevo lectin towards high-mannose type glycans with terminal αMan(1,2)αMan residues and its effect on M. tuberculosis internalization by macrophages. ITC studies demonstrated that Mevo lectin shows preferential binding towards manno-oligosaccharides with terminal αMan(1,2)αMan structures, and showed a strong affinity for ManLAM, whereas it binds weakly to Mycobacterium smegmatis (M. smegmatis) lipoarabinomannan (MsmLAM), which displays relatively fewer and shorter mannosyl caps. Crystal structure of Mevo lectin complexed with a Man7D1 revealed the multivalent cross-linking interaction, which explains avidity-based high affinity for these ligands when compared to previously studied manno-oligosaccharides lacking the specific termini. Functional studies suggest that M. tuberculosis internalization by the macrophage was impaired by binding of Mevo lectin to ManLAM present on the surface of M. tuberculosis. Selectivity shown by Mevo lectin towards glycans with terminal αMan(1,2)αMan structures, and its ability to compromise the internalization of M. tuberculosis in vitro, underscore the potential utility of Mevo lectin as a research tool to study host-pathogen interactions.
Collapse
Affiliation(s)
- Nukathoti Sivaji
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Nikitha Harish
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Samsher Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Amit Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Mamannamana Vijayan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|