51
|
Li X, Zuo S, Wang B, Zhang K, Wang Y. Antimicrobial Mechanisms and Clinical Application Prospects of Antimicrobial Peptides. Molecules 2022; 27:2675. [PMID: 35566025 PMCID: PMC9104849 DOI: 10.3390/molecules27092675] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial peptides are a type of small-molecule peptide that widely exist in nature and are components of the innate immunity of almost all living things. They play an important role in resisting foreign invading microorganisms. Antimicrobial peptides have a wide range of antibacterial activities against bacteria, fungi, viruses and other microorganisms. They are active against traditional antibiotic-resistant strains and do not easily induce the development of drug resistance. Therefore, they have become a hot spot of medical research and are expected to become a new substitute for fighting microbial infection and represent a new method for treating drug-resistant bacteria. This review briefly introduces the source and structural characteristics of antimicrobial peptides and describes those that have been used against common clinical microorganisms (bacteria, fungi, viruses, and especially coronaviruses), focusing on their antimicrobial mechanism of action and clinical application prospects.
Collapse
Affiliation(s)
- Xin Li
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun 130021, China; (X.L.); (B.W.)
| | - Siyao Zuo
- Department of Dermatology and Venereology, First Hospital of Jilin University, Changchun 130021, China;
| | - Bin Wang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun 130021, China; (X.L.); (B.W.)
| | - Kaiyu Zhang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun 130021, China; (X.L.); (B.W.)
| | - Yang Wang
- Department of Infectious Diseases, First Hospital of Jilin University, Changchun 130021, China; (X.L.); (B.W.)
| |
Collapse
|
52
|
Hu CT, Diaz K, Yang LC, Sharma A, Greenberg HB, Smith JG. VP4 Is a Determinant of Alpha-Defensin Modulation of Rotaviral Infection. J Virol 2022; 96:e0205321. [PMID: 35285683 PMCID: PMC9006894 DOI: 10.1128/jvi.02053-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Fecal-oral pathogens encounter constitutively expressed enteric alpha-defensins in the intestine during replication and transmission. Alpha-defensins can be potently antiviral and antibacterial; however, their primary sequences, the number of isoforms, and their activity against specific microorganisms often vary greatly between species, reflecting adaptation to species-specific pathogens. Therefore, alpha-defensins might influence not only microbial evolution and tissue tropism within a host but also species tropism and zoonotic potential. To investigate these concepts, we generated a panel of enteric and myeloid alpha-defensins from humans, rhesus macaques, and mice and tested their activity against group A rotaviruses, an important enteric viral pathogen of humans and animals. Rotaviral adaptation to the rhesus macaque correlated with resistance to rhesus enteric, but not myeloid, alpha-defensins and sensitivity to human alpha-defensins. While mouse rotaviral infection was increased in the presence of mouse enteric alpha-defensins, two prominent genotypes of human rotaviruses were differentially sensitive to human enteric alpha-defensins. Furthermore, the effects of cross-species alpha-defensins on human and mouse rotaviruses did not follow an obvious pattern. Thus, exposure to alpha-defensins may have shaped the evolution of some, but not all, rotaviruses. We then used a genetic approach to identify the viral attachment and penetration protein, VP4, as a determinant of alpha-defensin sensitivity. Our results provide a foundation for future studies of the VP4-dependent mechanism of defensin neutralization, highlight the species-specific activities of alpha-defensins, and focus future efforts on a broader range of rotaviruses that differ in VP4 to uncover the potential for enteric alpha-defensins to influence species tropism. IMPORTANCE Rotavirus is a leading cause of severe diarrhea in young children. Like other fecal-oral pathogens, rotaviruses encounter abundant, constitutively expressed defensins in the small intestine. These peptides are a vital part of the vertebrate innate immune system. By investigating the impact that defensins from multiple species have on the infectivity of different strains of rotavirus, we show that some rotaviral infections can be inhibited by defensins. We also found that some, but not all, rotaviruses may have evolved resistance to defensins in the intestine of their host species, and some even appropriate defensins to increase their infectivity. Because rotaviruses infect a broad range of animals and rotaviral infections are highly prevalent in children, identifying immune defenses against infection and how they vary across species and among viral genotypes is important for our understanding of the evolution, transmission, and zoonotic potential of these viruses as well as the improvement of vaccines.
Collapse
Affiliation(s)
- Ciara T. Hu
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Karina Diaz
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Linda C. Yang
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Anjali Sharma
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Harry B. Greenberg
- Department of Medicine and Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, California, USA
| | - Jason G. Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
53
|
Xu C, Wang A, Honnen W, Pinter A, Weston WK, Harness JA, Narayanan A, Chang TL. Brilacidin, a Non-Peptide Defensin-Mimetic Molecule, Inhibits SARS-CoV-2 Infection by Blocking Viral Entry. EC MICROBIOLOGY 2022; 18:1-12. [PMID: 35695877 PMCID: PMC9186380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Brilacidin (PMX-30063), a non-peptide defensin-mimetic small molecule, inhibits SARS-CoV-2 viral infection but the anti-viral mechanism is not defined. Here we determined its effect on the specific step of the viral life cycle. Brilacidin blocked SARS-CoV-2 infection but had no effect after viral entry. Brilacidin inhibited pseudotyped SARS-CoV-2 viruses expressing spike proteins from the P.1 Brazil strain and the B.1.1.7 UK strain. Brilacidin affected viral attachment in hACE2-dependent and independent manners depending on the concentrations. The inhibitory effect on viral entry was not mediated through blocking the binding of either the spike receptor-binding domain or the spike S1 protein to hACE2 proteins. Taken together, brilacidin inhibits SARS-CoV-2 infection by blocking viral entry and is active against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Chuan Xu
- Public Health Research Institute, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Annie Wang
- Public Health Research Institute, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - William Honnen
- Public Health Research Institute, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Abraham Pinter
- Public Health Research Institute, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Warren K Weston
- Innovation Pharmaceuticals, Inc., Wakefield, Massachusetts, USA
| | - Jane A Harness
- Innovation Pharmaceuticals, Inc., Wakefield, Massachusetts, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, USA
| | - Theresa L Chang
- Public Health Research Institute, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
54
|
Inhibition of SARS-CoV-2 Infection by Human Defensin HNP1 and Retrocyclin RC-101. J Mol Biol 2022; 434:167225. [PMID: 34487793 PMCID: PMC8413479 DOI: 10.1016/j.jmb.2021.167225] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 is an enveloped virus responsible for the COVID-19 pandemic. The emergence of new potentially more transmissible and vaccine-resistant variants of SARS-CoV-2 is an ever-present threat. Thus, it remains essential to better understand innate immune mechanisms that can inhibit the virus. One component of the innate immune system with broad antipathogen, including antiviral, activity is a group of cationic immune peptides termed defensins. The ability of defensins to neutralize enveloped and non-enveloped viruses and to inactivate numerous bacterial toxins correlate with their ability to promote the unfolding of proteins with high conformational plasticity. We found that human neutrophil α-defensin HNP1 binds to SARS-CoV-2 Spike protein with submicromolar affinity that is more than 20 fold stronger than its binding to serum albumin. As such, HNP1, as well as a θ-defensin retrocyclin RC-101, both interfere with Spike-mediated membrane fusion, Spike-pseudotyped lentivirus infection, and authentic SARS-CoV-2 infection in cell culture. These effects correlate with the abilities of the defensins to destabilize and precipitate Spike protein and inhibit the interaction of Spike with the ACE2 receptor. Serum reduces the anti-SARS-CoV-2 activity of HNP1, though at high concentrations, HNP1 was able to inactivate the virus even in the presence of serum. Overall, our results suggest that defensins can negatively affect the native conformation of SARS-CoV-2 Spike, and that α- and θ-defensins may be valuable tools in developing SARS-CoV-2 infection prevention strategies.
Collapse
|
55
|
Srivastava S, Verma S, Kamthania M, Agarwal D, Saxena AK, Kolbe M, Singh S, Kotnis A, Rathi B, Nayar SA, Shin HJ, Vashisht K, Pandey KC. Computationally validated SARS-CoV-2 CTL and HTL Multi-Patch vaccines, designed by reverse epitomics approach, show potential to cover large ethnically distributed human population worldwide. J Biomol Struct Dyn 2022; 40:2369-2388. [PMID: 33155524 PMCID: PMC7651196 DOI: 10.1080/07391102.2020.1838329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is responsible for the COVID-19 outbreak. The highly contagious COVID-19 disease has spread to 216 countries in less than six months. Though several vaccine candidates are being claimed, an effective vaccine is yet to come. A novel reverse epitomics approach, 'overlapping-epitope-clusters-to-patches' method is utilized to identify the antigenic regions from the SARS-CoV-2 proteome. These antigenic regions are named as 'Ag-Patch or Ag-Patches', for Antigenic Patch or Patches. The identification of Ag-Patches is based on the clusters of overlapping epitopes rising from SARS-CoV-2 proteins. Further, we have utilized the identified Ag-Patches to design Multi-Patch Vaccines (MPVs), proposing a novel method for the vaccine design. The designed MPVs were analyzed for immunologically crucial parameters, physiochemical properties and cDNA constructs. We identified 73 CTL (Cytotoxic T-Lymphocyte) and 49 HTL (Helper T-Lymphocyte) novel Ag-Patches from the proteome of SARS-CoV-2. The identified Ag-Patches utilized to design MPVs cover 768 overlapping epitopes targeting 55 different HLA alleles leading to 99.98% of world human population coverage. The MPVs and Toll-Like Receptor ectodomain complex shows stable complex formation tendency. Further, the cDNA analysis favors high expression of the MPVs constructs in a human cell line. We identified highly immunogenic novel Ag-Patches from the entire proteome of SARS CoV-2 by a novel reverse epitomics approach and utilized them to design MPVs. We conclude that the novel MPVs could be a highly potential novel approach to combat SARS-CoV-2, with greater effectiveness, high specificity and large human population coverage worldwide. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sukrit Srivastava
- Molecular Medicine Lab., School of Life Science, Jawaharlal Nehru University, New Delhi, India
- Infection Biology Group, Indian Foundation for Fundamental Research, RaeBareli, India
| | - Sonia Verma
- Parasite-Host Biology Group, Protein Biochemistry & Engineering Lab, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Mohit Kamthania
- Infection Biology Group, Indian Foundation for Fundamental Research, RaeBareli, India
| | - Deepa Agarwal
- Infection Biology Group, Indian Foundation for Fundamental Research, RaeBareli, India
| | - Ajay Kumar Saxena
- Molecular Medicine Lab., School of Life Science, Jawaharlal Nehru University, New Delhi, India
| | - Michael Kolbe
- Department for Structural Infection Biology, Centre for Structural Systems Biology (CSSB) & Helmholtz-Centre for Infection Research, Hamburg, Germany
- Faculty of Mathematics, Informatics and Natural Sciences, University of Hamburg, Hamburg, Germany
| | - Sarman Singh
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Ashwin Kotnis
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Brijesh Rathi
- Laboratory For Translational Chemistry and Drug Discovery, Hansraj College, University of Delhi, New Delhi, India
| | - Seema A. Nayar
- Department of Microbiology, Government Medical College, Trivandrum, India
- Department of Microbiology, Sree Gokulam Medical College, Trivandrum, India
| | - Ho-Joon Shin
- Department of Microbiology, School of Medicine, Ajou University, Suwon, South Korea
| | - Kapil Vashisht
- Parasite-Host Biology Group, Protein Biochemistry & Engineering Lab, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Kailash C. Pandey
- Parasite-Host Biology Group, Protein Biochemistry & Engineering Lab, ICMR-National Institute of Malaria Research, New Delhi, India
| |
Collapse
|
56
|
Leannec-Rialland V, Atanasova V, Chereau S, Tonk-Rügen M, Cabezas-Cruz A, Richard-Forget F. Use of Defensins to Develop Eco-Friendly Alternatives to Synthetic Fungicides to Control Phytopathogenic Fungi and Their Mycotoxins. J Fungi (Basel) 2022; 8:229. [PMID: 35330231 PMCID: PMC8950385 DOI: 10.3390/jof8030229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/10/2022] Open
Abstract
Crops are threatened by numerous fungal diseases that can adversely affect the availability and quality of agricultural commodities. In addition, some of these fungal phytopathogens have the capacity to produce mycotoxins that pose a serious health threat to humans and livestock. To facilitate the transition towards sustainable environmentally friendly agriculture, there is an urgent need to develop innovative methods allowing a reduced use of synthetic fungicides while guaranteeing optimal yields and the safety of the harvests. Several defensins have been reported to display antifungal and even-despite being under-studied-antimycotoxin activities and could be promising natural molecules for the development of control strategies. This review analyses pioneering and recent work addressing the bioactivity of defensins towards fungal phytopathogens; the details of approximately 100 active defensins and defensin-like peptides occurring in plants, mammals, fungi and invertebrates are listed. Moreover, the multi-faceted mechanism of action employed by defensins, the opportunity to optimize large-scale production procedures such as their solubility, stability and toxicity to plants and mammals are discussed. Overall, the knowledge gathered within the present review strongly supports the bright future held by defensin-based plant protection solutions while pointing out the obstacles that still need to be overcome to translate defensin-based in vitro research findings into commercial products.
Collapse
Affiliation(s)
- Valentin Leannec-Rialland
- Université de Bordeaux, UR1264 Mycology and Food Safety (MycSA), INRAE, 33882 Villenave d’Ornon, France;
| | - Vessela Atanasova
- UR1264 Mycology and Food Safety (MycSA), INRAE, 33882 Villenave d’Ornon, France; (V.A.); (S.C.)
| | - Sylvain Chereau
- UR1264 Mycology and Food Safety (MycSA), INRAE, 33882 Villenave d’Ornon, France; (V.A.); (S.C.)
| | - Miray Tonk-Rügen
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- Institute of Nutritional Sciences, Justus Liebig University, Wilhelmstrasse 20, 35392 Giessen, Germany
| | - Alejandro Cabezas-Cruz
- Anses, Ecole Nationale Vétérinaire d’Alfort, UMR Parasitic Molecular Biology and Immunology (BIPAR), Laboratoire de Santé Animale, INRAE, 94700 Maison-Alfort, France
| | - Florence Richard-Forget
- UR1264 Mycology and Food Safety (MycSA), INRAE, 33882 Villenave d’Ornon, France; (V.A.); (S.C.)
| |
Collapse
|
57
|
Kotlyarov S. Involvement of the Innate Immune System in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2022; 23:985. [PMID: 35055174 PMCID: PMC8778852 DOI: 10.3390/ijms23020985] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 01/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common, socially significant disease characterized by progressive airflow limitation due to chronic inflammation in the bronchi. Although the causes of COPD are considered to be known, the pathogenesis of the disease continues to be a relevant topic of study. Mechanisms of the innate immune system are involved in various links in the pathogenesis of COPD, leading to persistence of chronic inflammation in the bronchi, their bacterial colonization and disruption of lung structure and function. Bronchial epithelial cells, neutrophils, macrophages and other cells are involved in the development and progression of the disease, demonstrating multiple compromised immune mechanisms.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
58
|
Ballarini S, Ardusso L, Ortega Martell JA, Sacco O, Feleszko W, Rossi GA. Can bacterial lysates be useful in prevention of viral respiratory infections in childhood? The results of experimental OM-85 studies. Front Pediatr 2022; 10:1051079. [PMID: 36479289 PMCID: PMC9720385 DOI: 10.3389/fped.2022.1051079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 11/22/2022] Open
Abstract
Respiratory tract infections (RTI) are mainly viral in origin and among the leading cause of childhood morbidity globally. Associated wheezing illness and asthma are still a clear unmet medical need. Despite the continuous progress in understanding the processes involved in their pathogenesis, preventive measures and treatments failed to demonstrate any significant disease-modifying effect. However, in the last decades it was understood that early-life exposure to microbes, may reduce the risk of infectious and allergic disorders, increasing the immune response efficacy. These results suggested that treatment with bacterial lysates (BLs) acting on gut microbiota, could promote a heterologous immunomodulation useful in the prevention of recurrent RTIs and of wheezing inception and persistence. This hypothesis has been supported by clinical and experimental studies showing the reduction of RTI frequency and severity in childhood after oral BL prophylaxis and elucidating the involved mechanisms. OM-85 is the product whose anti-viral effects have been most extensively studied in vitro, animal, and human cell studies and in translational animal infection/disease models. The results of the latter studies, describing the potential immune training-based activities of such BL, leading to the protection against respiratory viruses, will be reported. In response to human rhinovirus, influenza virus, respiratory syncytial virus and severe acute respiratory coronavirus-2, OM-85 was effective in modulating the structure and the functions of a large numbers of airways epithelial and immune cells, when administered both orally and intranasally.
Collapse
Affiliation(s)
| | - Ledit Ardusso
- Allergy and Immunology Department, Rosario School of Medicine, National University of Rosario, Rosario, Argentina
| | | | - Oliviero Sacco
- Department of Pediatrics, Pulmonary and Allergy Disease Unit, G. Gaslini University Hospital, Genoa, Italy
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, The Medical University Children's Hospital, Warszawa, Poland
| | - Giovanni A Rossi
- Department of Pediatrics, Unit of Pediatrics Pulmonology and Respiratory Endoscopy, G. Gaslini Hospital, Genoa, Italy
| |
Collapse
|
59
|
Wei PL, Lin JC, Hung CS, Makondi PT, Batzorig U, Chang TC, Huang CY, Chang YJ. Human α-defensin 6 (HD6) suppresses CRC proliferation and metastasis through abolished EGF/EGFR signaling pathway. Int J Med Sci 2022; 19:34-46. [PMID: 34975297 PMCID: PMC8692109 DOI: 10.7150/ijms.64850] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
The incidence of colorectal cancer (CRC) has increased significantly in the past decade. Early diagnosis and new therapeutics are still urgently needed for CRC in clinical practice. Human α-defensin 6 (HD6) plays a defense role against microbes in the gastrointestinal tract. However, the role and mechanism of HD6 in CRC is still unresolved. Specimens from CRC patients with higher HD6 showed better outcomes. Overexpressed HD6 in CRC cells caused a reduction of cell proliferative, migratory, and invasive ability in vitro and in vivo. HD6-overexpressed caused S phase arrest through changes in cyclin-A and B and CDK2 levels. In addition, serpine-1 may be negatively regulated by HD6 altering the translocation of c-Jun N-terminal kinases (JNK), extracellular regulated protein kinases (ERK), and p38. Higher HD6 and lower serpine-1 levels in CRC patients reflected better outcomes. Finally, we found that HD6 interacts directly with epidermal growth factor receptor (EGFR) by co-immunoprecipitated assay. EGF treatment caused an increase of the level of serpine-1 and pEGFR levels and then increased growth activity in HD6 overexpressing cells. Together, our study shows that HD6 may compete with EGF to bind to EGFR and interrupt cancer progression in CRC. We believe these findings may give new insights for HD6 in CRC therapy.
Collapse
Affiliation(s)
- Po-Li Wei
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan.,Cancer Research Center and Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Jang-Chun Lin
- Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Sheng Hung
- Department of Surgery, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Uyanga Batzorig
- Department of Surgery, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tung-Cheng Chang
- Department of Surgery, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Colon and Rectal, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chien-Yu Huang
- Department of Surgery, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Colon and Rectal, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
60
|
Reijnders TDY, Schuurman AR, van der Poll T. The Immune Response to Respiratory Viruses: From Start to Memory. Semin Respir Crit Care Med 2021; 42:759-770. [PMID: 34918319 DOI: 10.1055/s-0041-1736459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Biomedical research has long strived to improve our understanding of the immune response to respiratory viral infections, an effort that has become all the more important as we live through the consequences of a pandemic. The disease course of these infections is shaped in large part by the actions of various cells of the innate and adaptive immune systems. While these cells are crucial in clearing viral pathogens and establishing long-term immunity, their effector mechanisms may also escalate into excessive, tissue-destructive inflammation detrimental to the host. In this review, we describe the breadth of the immune response to infection with respiratory viruses such as influenza and respiratory syncytial virus. Throughout, we focus on the host rather than the pathogen and try to describe shared patterns in the host response to different viruses. We start with the local cells of the airways, onto the recruitment and activation of innate and adaptive immune cells, followed by the establishment of local and systemic memory cells key in protection against reinfection. We end by exploring how respiratory viral infections can predispose to bacterial superinfection.
Collapse
Affiliation(s)
- Tom D Y Reijnders
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex R Schuurman
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
61
|
Tosta E. The seven constitutive respiratory defense barriers against SARS-CoV-2 infection. Rev Soc Bras Med Trop 2021; 54:e04612021. [PMID: 34932765 PMCID: PMC8687496 DOI: 10.1590/0037-8682-0461-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/17/2021] [Indexed: 12/18/2022] Open
Abstract
Before eliciting an adaptive immune response, SARS-CoV-2 must overcome seven constitutive respiratory defense barriers. The first is the mucus covering the respiratory tract's luminal surface, which entraps inhaled particles, including infectious agents, and eliminates them by mucociliary clearance. The second barrier comprises various components present in the airway lining fluid, the surfactants. Besides providing low surface tension that allows efficient gas exchange at the alveoli, surfactants inhibit the invasion of epithelial cells by respiratory viruses, enhance pathogen uptake by phagocytes, and regulate immune cells' functions. The respiratory tract microbiota constitutes the third defense barrier against SARS-CoV-2. It activates the innate and adaptive immune cells and elicits anti-infectious molecules such as secretory IgA antibodies, defensins, and interferons. The fourth defense barrier comprises the antimicrobial peptides defensins, and lactoferrin. They show direct antiviral activity, inhibit viral fusion, and modulate the innate and adaptive immune responses. Secretory IgA antibodies, the fifth defense barrier, besides protecting the local microbiota against noxious agents, also inhibit SARS-CoV-2 cell invasion. If the virus overcomes this barrier, it reaches its target, the respiratory epithelial cells. However, these cells also act as a defense barrier, the sixth one, since they hinder the virus' access to receptors and produce antiviral and immunomodulatory molecules such as interferons, lactoferrin, and defensins. Finally, the sensing of the virus by the cells of innate immunity, the last constitutive defense barrier, elicits a cascade of signals that activate adaptive immune cells and may inhibit the development of productive infection. The subject of the present essay is discussing these mechanisms.
Collapse
Affiliation(s)
- Eduardo Tosta
- Professor Emeritus, Faculdade de Medicina, Universidade de Brasília, Brasília, DF , Brasil
| |
Collapse
|
62
|
Gao X, Ding J, Liao C, Xu J, Liu X, Lu W. Defensins: The natural peptide antibiotic. Adv Drug Deliv Rev 2021; 179:114008. [PMID: 34673132 DOI: 10.1016/j.addr.2021.114008] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/28/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
Defensins are a family of cationic antimicrobial peptides active against a broad range of infectious microbes including bacteria, viruses and fungi, playing important roles as innate effectors and immune modulators in immunological control of microbial infection. Their antibacterial properties and unique mechanisms of action have garnered considerable interest in developing defensins into a novel class of natural antibiotic peptides to fend off pathogenic infection by bacteria, particularly those resistant to conventional antibiotics. However, serious pharmacological and technical obstacles, some of which are unique to defensins and others are common to peptide drugs in general, have hindered the development and clinical translation of defensins as anti-infective therapeutics. To overcome them, several technologies have been developed, aiming for improved functionality, prolonged circulation time, enhanced proteolytic stability and bioavailability, and efficient and controlled delivery and release of defensins to the site of infection. Additional challenges include the alleviation of potential toxicity of defensins and their cost-effective manufacturing. In this review, we briefly introduce defensin biology, focus on various transforming strategies and practical techniques developed for defensins and their derivatives as antibacterial therapeutics, and conclude with a summation of future challenges and possible solutions.
Collapse
|
63
|
Kuroki A, Tay J, Lee GH, Yang YY. Broad-Spectrum Antiviral Peptides and Polymers. Adv Healthc Mater 2021; 10:e2101113. [PMID: 34599850 DOI: 10.1002/adhm.202101113] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/13/2021] [Indexed: 12/18/2022]
Abstract
As the human cost of the pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still being witnessed worldwide, the development of broad-spectrum antiviral agents against emerging and re-emerging viruses is seen as a necessity to hamper the spread of infections. Various targets during the viral life-cycle can be considered to inhibit viral infection, from viral attachment to viral fusion or replication. Macromolecules represent a particularly attractive class of therapeutics due to their multivalency and versatility. Although several antiviral macromolecules hold great promise in clinical applications, the emergence of resistance after prolonged exposure urges the need for improved solutions. In the present article, the recent advancement in the discovery of antiviral peptides and polymers with diverse structural features and antiviral mechanisms is reviewed. Future perspectives, such as, the development of virucidal peptides/polymers and their coatings against SARS-CoV-2 infection, standardization of antiviral testing protocols, and use of artificial intelligence or machine learning as a tool to accelerate the discovery of antiviral macromolecules, are discussed.
Collapse
Affiliation(s)
- Agnès Kuroki
- Yong Loo Lin School of Medicine National University of Singapore Singapore 117597 Singapore
- Institute of Bioengineering and Bioimaging 31 Biopolis Ways, The Nanos Singapore 138669 Singapore
| | - Joyce Tay
- Institute of Bioengineering and Bioimaging 31 Biopolis Ways, The Nanos Singapore 138669 Singapore
| | - Guan Huei Lee
- Yong Loo Lin School of Medicine National University of Singapore Singapore 117597 Singapore
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging 31 Biopolis Ways, The Nanos Singapore 138669 Singapore
| |
Collapse
|
64
|
Lin B, Li R, Handley TN, Wade JD, Li W, O’Brien-Simpson NM. Cationic Antimicrobial Peptides Are Leading the Way to Combat Oropathogenic Infections. ACS Infect Dis 2021; 7:2959-2970. [PMID: 34587737 DOI: 10.1021/acsinfecdis.1c00424] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oral dental infections are one of the most common diseases affecting humans, with caries and periodontal disease having the highest incidence. Caries and periodontal disease arise from infections caused by oral bacterial pathogens. Current misuse and overuse of antibiotic treatments have led to the development of antimicrobial resistance. However, recent studies have shown that cationic antimicrobial peptides are a promising family of antibacterial agents that are active against oral pathogenic bacteria and also possess less propensity for development of antimicrobial resistance. This timely Review has a focus on two primary subjects: (i) the oral bacterial pathogens associated with dental infections and (ii) the current development of antimicrobial peptides targeting oral pathogens.
Collapse
Affiliation(s)
- Bruce Lin
- The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Rong Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
- Department of Biochemistry & Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Thomas N.G. Handley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - John D. Wade
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
- School of Chemistry, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Wenyi Li
- The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Neil M. O’Brien-Simpson
- The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
65
|
Al-Bayatee NT, Ad'hiah AH. Human beta-defensins 2 and 4 are dysregulated in patients with coronavirus disease 19. Microb Pathog 2021; 160:105205. [PMID: 34547411 PMCID: PMC8450228 DOI: 10.1016/j.micpath.2021.105205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
Antimicrobial peptides (AMPs) have recently been proposed as significant immunological factors involved in pathogenesis of coronavirus disease 19 (COVID-19). Human β-defensins (hBDs) are among these AMPs, but the evidence is not well detailed. Therefore, this case-control study analyzed levels of hBD1, hBD2, hBD3 and hBD4 in serum of 103 patients with severe COVID-19 and 105 healthy controls. Most patients were older than 45 years (80.6%), and more than 50% suffered from chronic diseases (cardiovascular and diabetes). Results revealed that median levels of hBD1 and hBD3 did not show significant differences between patients and controls. On the contrary, HBD2 levels were significantly decreased in patients compared to controls (1036 vs. 1289 ng/L; p < 0.001), while HBD4 levels were significantly increased (4.04 vs. 2.43 ng/L; p < 0.001). Receiver operating characteristic curve analysis demonstrated the predictive significance of hBD2 (area under the curve [AUC] = 0.795; 95% confidence interval [CI] = 0.729–0.861; p < 0.001) and hBD4 (AUC = 0.816; 95% CI = 0.756–0.876; p < 0.001) in discriminating between COVID-19 patients and controls. Logistic regression analysis (adjusted for age, gender and body mass index) confirmed the significance of hBD2 (odds ratio [OR] = 0.996; corrected p = 0.004) and hBD4 (OR = 4.948; corrected p < 0.001) in susceptibility to COVID-19. In conclusion, the study indicated that hBD2 showed low levels in serum of patients infected with severe COVID-19, while hBD4 showed elevated levels. These differences in HBDs were not influenced by age, gender, body mass index, or chronic disease.
Collapse
Affiliation(s)
- Noor T Al-Bayatee
- Biotechnology Department, College of Science, University of Baghdad, Baghdad, Iraq
| | - Ali H Ad'hiah
- Tropical-Biological Research Unit, College of Science, University of Baghdad, Baghdad, Iraq.
| |
Collapse
|
66
|
Rivas-Santiago B, Jacobo-Delgado Y, Rodriguez-Carlos A. Are Host Defense Peptides and Their Derivatives Ready to be Part of the Treatment of the Next Coronavirus Pandemic? Arch Immunol Ther Exp (Warsz) 2021; 69:25. [PMID: 34529143 PMCID: PMC8444179 DOI: 10.1007/s00005-021-00630-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
The term host defense peptides arose at the beginning to refer to those peptides that are part of the host's immunity. Because of their broad antimicrobial capacity and immunomodulatory activity, nowadays, they emerge as a hope to combat resistant multi-drug microorganisms and emerging viruses, such as the case of coronaviruses. Since the beginning of this century, coronaviruses have been part of different outbreaks and a pandemic, and they will be surely part of the next pandemics, this review analyses whether these peptides and their derivatives are ready to be part of the treatment of the next coronavirus pandemic.
Collapse
Affiliation(s)
- Bruno Rivas-Santiago
- Biomedical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Col. Centro Zacatecas, Interior of Alameda #45, Zacatecas, Mexico.
| | - Yolanda Jacobo-Delgado
- Biomedical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Col. Centro Zacatecas, Interior of Alameda #45, Zacatecas, Mexico
| | - Adrian Rodriguez-Carlos
- Biomedical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Col. Centro Zacatecas, Interior of Alameda #45, Zacatecas, Mexico
| |
Collapse
|
67
|
Abdelraouf AMN, Naguib DM. Nano Defensin: A Promising Antibacterial Agent Against Colorectal Cancer Related Bacteria. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10237-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
68
|
Johnston RA, Rheinwald JG, vonHoldt BM, Stahler DR, Lowry W, Tung J, Wayne RK. K Locus Effects in Gray Wolves: Experimental Assessment of TLR3 Signaling and the Gene Expression Response to Canine Distemper Virus. J Hered 2021; 112:458-468. [PMID: 34132805 PMCID: PMC8567826 DOI: 10.1093/jhered/esab029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/04/2021] [Accepted: 05/06/2021] [Indexed: 12/21/2022] Open
Abstract
In North American gray wolves, black coat color is dominantly inherited via a 3 base pair coding deletion in the canine beta defensin 3 (CBD103) gene. This 3 base pair deletion, called the KB allele, was introduced through hybridization with dogs and subsequently underwent a selective sweep that increased its frequency in wild wolves. Despite apparent positive selection, KBB wolves have lower fitness than wolves with the KyB genotype, even though the 2 genotypes show no observable differences in black coat color. Thus, the KB allele is thought to have pleiotropic effects on as-yet unknown phenotypes. Given the role of skin-expressed CBD103 in innate immunity, we hypothesized that the KB allele influences the keratinocyte gene expression response to TLR3 pathway stimulation and/or infection by canine distemper virus (CDV). To test this hypothesis, we developed a panel of primary epidermal keratinocyte cell cultures from 24 wild North American gray wolves of both Kyy and KyB genotypes. In addition, we generated an immortalized Kyy line and used CRISPR/Cas9 editing to produce a KyB line on the same genetic background. We assessed the transcriptome-wide responses of wolf keratinocytes to the TLR3 agonist polyinosinic:polycytidylic acid (polyI:C), and to live CDV. K locus genotype did not predict the transcriptional response to either challenge, suggesting that variation in the gene expression response does not explain pleiotropic effects of the KB allele on fitness. This study supports the feasibility of using cell culture methods to investigate the phenotypic effects of naturally occurring genetic variation in wild mammals.
Collapse
Affiliation(s)
- Rachel A Johnston
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
| | - James G Rheinwald
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Bridgett M vonHoldt
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - Daniel R Stahler
- Yellowstone Center for Resources, National Park Service, Yellowstone National Park, WY, USA
| | - William Lowry
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jenny Tung
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
- Department of Biology, Duke University, Durham, NC, USA
- Duke Population Research Institute, Duke University, Durham, NC, USA
| | - Robert K Wayne
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
69
|
Gisina A, Novikova S, Kim Y, Sidorov D, Bykasov S, Volchenko N, Kaprin A, Zgoda V, Yarygin K, Lupatov A. CEACAM5 overexpression is a reliable characteristic of CD133-positive colorectal cancer stem cells. Cancer Biomark 2021; 32:85-98. [PMID: 34092615 DOI: 10.3233/cbm-203187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND CD133 (prominin-1) is the most commonly used molecular marker of the cancer stem cells (CSCs) that maintain tumor progression and recurrence in colorectal cancer. However, the proteome of CSCs directly isolated from colorectal tumors based on CD133 expression has never been investigated. OBJECTIVE To reveal biomarkers of CD133-positive colorectal CSCs. METHODS Thirty colorectal tumor samples were collected from patients undergoing bowel resection. CD133-positive and CD133-negative cells were isolated by FACS. Comparative proteomic profiling was performed by LC-MS/MS analysis combined with label-free quantification. Verification of differentially expressed proteins was performed by flow cytometry or ELISA. CD133-knockout Caco-2 and HT-29 cell lines were generated using CRISPR-Cas9 gene editing. RESULTS LC-MS/MS analysis identified 29 proteins with at least 2.5-fold higher expression in CD133-positive cells versus CD133-negative cells. Flow cytometry confirmed CEACAM5 overexpression in CD133-positive cells in all clinical samples analyzed. S100A8, S100A9, and DEFA1 were differentially expressed in only a proportion of the samples. CD133 knockout in the colon cancer cell lines Caco-2 and HT-29 did not affect the median level of CEACAM5 expression, but led to higher variance of the percentage of CEACAM5-positive cells. CONCLUSIONS High CEACAM5 expression in colorectal cancer cells is firmly associated with the CD133-positive colorectal CSC phenotype, but it is unlikely that CD133 directly regulates CEACAM5 expression.
Collapse
Affiliation(s)
- Alisa Gisina
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow, Russia
| | - Svetlana Novikova
- Laboratory of Systems Biology, Institute of Biomedical Chemistry, Moscow, Russia
| | - Yan Kim
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow, Russia
| | - Dmitry Sidorov
- Department of Abdominal Surgery, P. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Stanislav Bykasov
- Department of Abdominal Surgery, P. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Nadezhda Volchenko
- Department of Oncomorphology, P. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Andrey Kaprin
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Victor Zgoda
- Laboratory of Systems Biology, Institute of Biomedical Chemistry, Moscow, Russia
| | - Konstantin Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow, Russia
| | - Alexey Lupatov
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
70
|
Damour A, Garcia M, Cho HS, Larivière A, Lévêque N, Park C, Bodet C. Characterisation of Antiviral Activity of Cathelicidins from Naked Mole Rat and Python bivittatus on Human Herpes Simplex Virus 1. Pharmaceuticals (Basel) 2021; 14:ph14080715. [PMID: 34451812 PMCID: PMC8398704 DOI: 10.3390/ph14080715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Hg-CATH and Pb-CATH4 are cathelicidins from Heterocephalus glaber and Python bivittatus that have been previously identified as potent antibacterial peptides. However, their antiviral properties were not previously investigated. In this study, their activity against the herpes simplex virus (HSV)-1 was evaluated during primary human keratinocyte infection. Both of them significantly reduced HSV-1 DNA replication and production of infectious viral particles in keratinocytes at noncytotoxic concentrations, with the stronger activity of Pb-CATH4. These peptides did not show direct virucidal activity and did not exhibit significant immunomodulatory properties, except for Pb-CATH4, which exerted a moderate proinflammatory action. All in all, our results suggest that Hg-CATH and Pb-CATH4 could be potent candidates for the development of new therapies against HSV-1.
Collapse
Affiliation(s)
- Alexia Damour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
| | - Magali Garcia
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, 86021 Poitiers, France
| | - Hye-Sun Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (H.-S.C.); (C.P.)
| | - Andy Larivière
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, 86021 Poitiers, France
| | - Nicolas Lévêque
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, 86021 Poitiers, France
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (H.-S.C.); (C.P.)
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC EA 4331), Université de Poitiers, CEDEX 9, 86073 Poitiers, France; (A.D.); (M.G.); (A.L.); (N.L.)
- Correspondence:
| |
Collapse
|
71
|
Liu L, Deng CJ, Duan YL, Ye CJ, Gong DH, Guo XL, Lee WH, Zhou J, Li SA, Zhang Y. An Aerolysin-like Pore-Forming Protein Complex Targets Viral Envelope to Inactivate Herpes Simplex Virus Type 1. THE JOURNAL OF IMMUNOLOGY 2021; 207:888-901. [PMID: 34290105 DOI: 10.4049/jimmunol.2001056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/20/2021] [Indexed: 01/12/2023]
Abstract
Because most of animal viruses are enveloped, cytoplasmic entry of these viruses via fusion with cellular membrane initiates their invasion. However, the strategies in which host cells counteract cytoplasmic entry of such viruses are incompletely understood. Pore-forming toxin aerolysin-like proteins (ALPs) exist throughout the animal kingdom, but their functions are mostly unknown. In this study, we report that βγ-crystallin fused aerolysin-like protein and trefoil factor complex (βγ-CAT), an ALP and trefoil factor complex from the frog Bombina maxima, directly blocks enveloped virus invasion by interfering with cytoplasmic entry. βγ-CAT targeted acidic glycosphingolipids on the HSV type 1 (HSV-1) envelope to induce pore formation, as indicated by the oligomer formation of protein and potassium and calcium ion efflux. Meanwhile, βγ-CAT formed ring-like oligomers of ∼10 nm in diameter on the liposomes and induced dye release from liposomes that mimic viral envelope. Unexpectedly, transmission electron microscopy analysis showed that the βγ-CAT-treated HSV-1 was visibly as intact as the vehicle-treated HSV-1, indicating that βγ-CAT did not lyse the viral envelope. However, the cytoplasmic entry of the βγ-CAT-treated HSV-1 into HeLa cells was totally hindered. In vivo, topical application of βγ-CAT attenuated the HSV-1 corneal infection in mice. Collectively, these results uncovered that βγ-CAT possesses the capacity to counteract enveloped virus invasion with its featured antiviral-acting manner. Our findings will also largely help to illustrate the putative antiviral activity of animal ALPs.
Collapse
Affiliation(s)
- Long Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Cheng-Jie Deng
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ya-Li Duan
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chen-Jun Ye
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dao-Hua Gong
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao-Long Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Wen-Hui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jumin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China;
| | - Sheng-An Li
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China; and
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Science/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
72
|
El-Dirany R, Shahrour H, Dirany Z, Abdel-Sater F, Gonzalez-Gaitano G, Brandenburg K, Martinez de Tejada G, Nguewa PA. Activity of Anti-Microbial Peptides (AMPs) against Leishmania and Other Parasites: An Overview. Biomolecules 2021; 11:984. [PMID: 34356608 PMCID: PMC8301979 DOI: 10.3390/biom11070984] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-microbial peptides (AMPs), small biologically active molecules, produced by different organisms through their innate immune system, have become a considerable subject of interest in the request of novel therapeutics. Most of these peptides are cationic-amphipathic, exhibiting two main mechanisms of action, direct lysis and by modulating the immunity. The most commonly reported activity of AMPs is their anti-bacterial effects, although other effects, such as anti-fungal, anti-viral, and anti-parasitic, as well as anti-tumor mechanisms of action have also been described. Their anti-parasitic effect against leishmaniasis has been studied. Leishmaniasis is a neglected tropical disease. Currently among parasitic diseases, it is the second most threating illness after malaria. Clinical treatments, mainly antimonial derivatives, are related to drug resistance and some undesirable effects. Therefore, the development of new therapeutic agents has become a priority, and AMPs constitute a promising alternative. In this work, we describe the principal families of AMPs (melittin, cecropin, cathelicidin, defensin, magainin, temporin, dermaseptin, eumenitin, and histatin) exhibiting a potential anti-leishmanial activity, as well as their effectiveness against other microorganisms.
Collapse
Affiliation(s)
- Rima El-Dirany
- ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), University of Navarra, c/Irunlarrea 1, 31008 Pamplona, Navarra, Spain;
- Faculty of Sciences I, Lebanese University, Hadath 1003, Lebanon; (H.S.); (F.A.-S.)
| | - Hawraa Shahrour
- Faculty of Sciences I, Lebanese University, Hadath 1003, Lebanon; (H.S.); (F.A.-S.)
- Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), University of Navarra, 31008 Pamplona, Navarra, Spain;
| | - Zeinab Dirany
- Department of Chemistry, Faculty of Sciences, University of Navarra, 31080 Pamplona, Navarra, Spain; (Z.D.); (G.G.-G.)
| | - Fadi Abdel-Sater
- Faculty of Sciences I, Lebanese University, Hadath 1003, Lebanon; (H.S.); (F.A.-S.)
| | - Gustavo Gonzalez-Gaitano
- Department of Chemistry, Faculty of Sciences, University of Navarra, 31080 Pamplona, Navarra, Spain; (Z.D.); (G.G.-G.)
| | - Klaus Brandenburg
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Leibniz Lungenzentrum, 23845 Borstel, Germany;
| | - Guillermo Martinez de Tejada
- Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), University of Navarra, 31008 Pamplona, Navarra, Spain;
| | - Paul A. Nguewa
- ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), University of Navarra, c/Irunlarrea 1, 31008 Pamplona, Navarra, Spain;
| |
Collapse
|
73
|
Bharucha JP, Sun L, Lu W, Gartner S, Garzino-Demo A. Human Beta-Defensin 2 and 3 Inhibit HIV-1 Replication in Macrophages. Front Cell Infect Microbiol 2021; 11:535352. [PMID: 34277460 PMCID: PMC8281893 DOI: 10.3389/fcimb.2021.535352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/17/2021] [Indexed: 12/26/2022] Open
Abstract
Human beta-defensins (hBDs) are broad-spectrum antimicrobial peptides, secreted by epithelial cells of the skin and mucosae, and astrocytes, which we and others have shown to inhibit HIV-1 in primary CD4+ T cells. Although loss of CD4+ T cells contributes to mucosal immune dysfunction, macrophages are a major source of persistence and spread of HIV and also contribute to the development of various HIV-associated complications. We hypothesized that, besides T cells, hBDs could protect macrophages from HIV. Our data in primary human monocyte-derived macrophages (MDM) in vitro show that hBD2 and hBD3 inhibit HIV replication in a dose-dependent manner. We determined that hBD2 neither alters surface expression of HIV receptors nor induces expression of anti-HIV cytokines or beta-chemokines in MDM. Studies using a G-protein signaling antagonist in a single-cycle reporter virus system showed that hBD2 suppresses HIV at an early post-entry stage via G-protein coupled receptor (GPCR)-mediated signaling. We find that MDM express the shared chemokine-hBD receptors CCR2 and CCR6, albeit at variable levels among donors. However, cell surface expression analyses show that neither of these receptors is necessary for hBD2-mediated HIV inhibition, suggesting that hBD2 can signal via additional receptor(s). Our data also illustrate that hBD2 treatment was associated with increased expression of APOBEC3A and 3G antiretroviral restriction factors in MDM. These findings suggest that hBD2 inhibits HIV in MDM via more than one CCR thus adding to the potential of using β-defensins in preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Jennifer P Bharucha
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Lingling Sun
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Wuyuan Lu
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Suzanne Gartner
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alfredo Garzino-Demo
- Division of Virology, Pathogenesis, and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
74
|
Ghosh SK, Weinberg A. Ramping Up Antimicrobial Peptides Against Severe Acute Respiratory Syndrome Coronavirus-2. Front Mol Biosci 2021; 8:620806. [PMID: 34235176 PMCID: PMC8255374 DOI: 10.3389/fmolb.2021.620806] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 05/27/2021] [Indexed: 12/17/2022] Open
Abstract
Human-derived antimicrobial peptides (AMPs), such as defensins and cathelicidin LL-37, are members of the innate immune system and play a crucial role in early pulmonary defense against viruses. These AMPs achieve viral inhibition through a variety of mechanisms including, but not limited to, direct binding to virions, binding to and modulating host cell-surface receptors, blocking viral replication, and aggregation of viral particles and indirectly by functioning as chemokines to enhance or curb adaptive immune responses. Given the fact that we are in a pandemic of unprecedented severity and the urgent need for therapeutic options to combat severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), naturally expressed AMPs and their derivatives have the potential to combat coronavirus disease 2019 (COVID-19) and impede viral infectivity in various ways. Provided the fact that development of effective treatments is an urgent public health priority, AMPs and their derivatives are being explored as potential prophylactic and therapeutic candidates. Additionally, cell-based platforms such as human mesenchymal stem cell (hMSC) therapy are showing success in saving the lives of severely ill patients infected with SARS-CoV-2. This could be partially due to AMPs released from hMSCs that also act as immunological rheostats to modulate the host inflammatory response. This review highlights the utilization of AMPs in strategies that could be implemented as novel therapeutics, either alone or in combination with other platforms, to treat CoV-2-infected individuals.
Collapse
Affiliation(s)
| | - Aaron Weinberg
- Department of Biological Sciences, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
75
|
The Peptide TAT-I24 with Antiviral Activity against DNA Viruses Binds Double-Stranded DNA with High Affinity. Biologics 2021. [DOI: 10.3390/biologics1010003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The peptide TAT-I24, composed of the 9-mer peptide I24 and the TAT (48-60) peptide, exerts broad-spectrum antiviral activity against several DNA viruses. The current model of the mode of action suggests a reduction of viral entry and also a possible interaction with the viral DNA upon virus entry. To further support this model, the present study investigates the DNA binding properties of TAT-I24. DNA binding was analysed by gel retardation of a peptide-complexed DNA, fluorescence reduction of DNA labelled with intercalating dyes and determination of binding kinetics by surface plasmon resonance. Molecular dynamics simulations of DNA-peptide complexes predict high-affinity binding and destabilization of the DNA by TAT-I24. The effect on viral DNA levels of infected cells were studied by real-time PCR and staining of viral DNA by bromodeoxyuridine. TAT-I24 binds double-stranded DNA with high affinity, leading to inhibition of polymerase binding and thereby blocking of de novo nucleic acid synthesis. Analysis of early steps of virus entry using a bromodeoxyuridine-labelled virus as well as quantification of viral genomes in the cells indicate direct binding of the peptide to the viral DNA. Saturation of the peptide with exogenous DNA can fully neutralize the inhibitory effect. The antiviral activity of TAT-I24 is linked to its ability to bind DNA with high affinity. This mechanism could be the basis for the development of novel antiviral agents.
Collapse
|
76
|
Sahoo A, Swain SS, Behera A, Sahoo G, Mahapatra PK, Panda SK. Antimicrobial Peptides Derived From Insects Offer a Novel Therapeutic Option to Combat Biofilm: A Review. Front Microbiol 2021; 12:661195. [PMID: 34248873 PMCID: PMC8265172 DOI: 10.3389/fmicb.2021.661195] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022] Open
Abstract
Biofilms form a complex layer with defined structures, that attach on biotic or abiotic surfaces, are tough to eradicate and tend to cause some resistance against most antibiotics. Several studies confirmed that biofilm-producing bacteria exhibit higher resistance compared to the planktonic form of the same species. Antibiotic resistance factors are well understood in planktonic bacteria which is not so in case of biofilm producing forms. This may be due to the lack of available drugs with known resistance mechanisms for biofilms. Existing antibiotics cannot eradicate most biofilms, especially of ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). Insects produce complex and diverse set of chemicals for survival and defense. Antimicrobial peptides (AMPs), produced by most insects, generally have a broad spectrum of activity and the potential to bypass the resistance mechanisms of classical antibiotics. Besides, AMPs may well act synergistically with classical antibiotics for a double-pronged attack on infections. Thus, AMPs could be promising alternatives to overcome medically important biofilms, decrease the possibility of acquired resistance and treatment of multidrug-resistant pathogens including ESKAPE. The present review focuses on insect-derived AMPs with special reference to anti-biofilm-based strategies. It covers the AMP composition, pathways and mechanisms of action, the formation of biofilms, impact of biofilms on human diseases, current strategies as well as therapeutic options to combat biofilm with antimicrobial peptides from insects. In addition, the review also illustrates the importance of bioinformatics tools and molecular docking studies to boost the importance of select bioactive peptides those can be developed as drugs, as well as suggestions for further basic and clinical research.
Collapse
Affiliation(s)
- Alaka Sahoo
- Department of Skin & VD, Institute of Medical Sciences, SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, India
| | - Shasank Sekhar Swain
- Division of Microbiology & NCDs, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Ayusman Behera
- Department of Zoology, Maharaja Sriram Chandra Bhanja Deo University, Baripada, India
| | - Gunanidhi Sahoo
- Department of Zoology, Utkal University, Vani Vihar, Bhubaneswar, India
| | | | - Sujogya Kumar Panda
- Centre of Environment, Climate Change and Public Health, RUSA 2.0, Utkal University, Vani Vihar, Bhubaneswar, India
| |
Collapse
|
77
|
Yacouba A, Olowo-Okere A, Yunusa I. Repurposing of antibiotics for clinical management of COVID-19: a narrative review. Ann Clin Microbiol Antimicrob 2021; 20:37. [PMID: 34020659 PMCID: PMC8139224 DOI: 10.1186/s12941-021-00444-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 05/11/2021] [Indexed: 12/22/2022] Open
Abstract
Background Drug repurposing otherwise known as drug repositioning or drug re-profiling is a time-tested approach in drug discovery through which new medical uses are being established for already known drugs. Antibiotics are among the pharmacological agents being investigated for potential anti-SARS-COV-2 activities. The antibiotics are used either to resolve bacterial infections co-existing with COVID-19 infections or exploitation of their potential antiviral activities. Herein, we aimed to review the various antibiotics that have been repositioned for the management of COVID-19. Methods This literature review was conducted from a methodical search on PubMed and Web of Science regarding antibiotics used in patients with COVID-19 up to July 5, 2020. Results Macrolide and specifically azithromycin is the most common antibiotic used in the clinical management of COVID-19. The other antibiotics used in COVID-19 includes teicoplanin, clarithromycin, doxycycline, tetracyclines, levofloxacin, moxifloxacin, ciprofloxacin, and cefuroxime. In patients with COVID-19, antibiotics are used for their immune-modulating, anti-inflammatory, and antiviral properties. The precise antiviral mechanism of most of these antibiotics has not been determined. Moreover, the use of some of these antibiotics against SARS-CoV-2 infection remains highly controversial and not widely accepted. Conclusion The heavy use of antibiotics during the COVID-19 pandemic would likely worsen antibiotic resistance crisis. Consequently, antibiotic stewardship should be strengthened in order to prevent the impacts of COVID-19 on the antibiotic resistance crisis.
Collapse
Affiliation(s)
- Abdourahamane Yacouba
- Faculté des Sciences de la Santé, Université Abdou Moumouni, P.M.B. 10896, Niamey, Niger.
| | - Ahmed Olowo-Okere
- Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, P.M.B. 2346, Sokoto, Nigeria
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| |
Collapse
|
78
|
Tonk M, Růžek D, Vilcinskas A. Compelling Evidence for the Activity of Antiviral Peptides against SARS-CoV-2. Viruses 2021; 13:v13050912. [PMID: 34069206 PMCID: PMC8156787 DOI: 10.3390/v13050912] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple outbreaks of epidemic and pandemic viral diseases have occurred in the last 20 years, including those caused by Ebola virus, Zika virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The emergence or re-emergence of such diseases has revealed the deficiency in our pipeline for the discovery and development of antiviral drugs. One promising solution is the extensive library of antimicrobial peptides (AMPs) produced by all eukaryotic organisms. AMPs are widely known for their activity against bacteria, but many possess additional antifungal, antiparasitic, insecticidal, anticancer, or antiviral activities. AMPs could therefore be suitable as leads for the development of new peptide-based antiviral drugs. Sixty therapeutic peptides had been approved by the end of 2018, with at least another 150 in preclinical or clinical development. Peptides undergoing clinical trials include analogs, mimetics, and natural AMPs. The advantages of AMPs include novel mechanisms of action that hinder the evolution of resistance, low molecular weight, low toxicity toward human cells but high specificity and efficacy, the latter enhanced by the optimization of AMP sequences. In this opinion article, we summarize the evidence supporting the efficacy of antiviral AMPs and discuss their potential to treat emerging viral diseases including COVID-19.
Collapse
Affiliation(s)
- Miray Tonk
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
| | - Daniel Růžek
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic;
- Biology Centre of the Czech Academy of Sciences, Institute of Parasitology, Branisovska 31, 37005 Ceske Budejovice, Czech Republic
| | - Andreas Vilcinskas
- Institute for Insect Biotechnology, Justus Liebig University of Giessen, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
79
|
Liu X, Cao Y, Xie X, Qin X, He X, Shi C, Zeng W, Guo Y, Lin Y. Association between vaginal microbiota and risk of early pregnancy miscarriage. Comp Immunol Microbiol Infect Dis 2021; 77:101669. [PMID: 34116388 DOI: 10.1016/j.cimid.2021.101669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 10/21/2022]
Abstract
Approximately 15 % of clinically recognized pregnancies end in miscarriage. To explore the vaginal microbiota profile in women diagnosed with early pregnancy miscarriage (including missed miscarriage [M] or empty-sac miscarriage [E]), the microbial community structure in vaginal fluid was evaluated by Illumina MiSeq sequencing of the 16S rRNA gene V4 region and compared with that in women with normal pregnancy (P). Taxa identified in samples from the P, E, and M groups formed distinct clusters. The M group had the highest bacterial species richness and diversity, with lower Lactobacillus levels and higher Bacteroides, Halomonas, Miscellaneous-Crenarchaeota, Bacillus, Staphylococcus, Escherichia/Shigella, and Acetobacter levels than in the other two groups. The vaginal community-state types differed significantly among the three groups (P = 0.02) but were similar between the P and E groups (P = 0.21). Moreover, we identified an optimal marker set composed of 12 operational taxonomic units based on a random forest model that distinguished the M and P groups, with areas under the receiver-operating characteristic curve of 86.76 % and 93.33 % in the training and test groups, respectively. In conclusion, this study highlights that patients with early pregnancy miscarriage had a significantly different vaginal microbiota profile. Microbial markers analyzed by RT-qPCR may be applied for the etiological diagnosis of miscarriage. Further studies should be performed to elucidate the possible mechanism of special strains affecting miscarriage during early pregnancy.
Collapse
Affiliation(s)
- Xiaorui Liu
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yunyun Cao
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xianjing Xie
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiaoli Qin
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiaoying He
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chunlei Shi
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai, 200240, China; School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weihong Zeng
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yuna Guo
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yi Lin
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
80
|
Julio-Pieper M, López-Aguilera A, Eyzaguirre-Velásquez J, Olavarría-Ramírez L, Ibacache-Quiroga C, Bravo JA, Cruz G. Gut Susceptibility to Viral Invasion: Contributing Roles of Diet, Microbiota and Enteric Nervous System to Mucosal Barrier Preservation. Int J Mol Sci 2021; 22:ijms22094734. [PMID: 33946994 PMCID: PMC8125429 DOI: 10.3390/ijms22094734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/08/2023] Open
Abstract
The gastrointestinal lumen is a rich source of eukaryotic and prokaryotic viruses which, together with bacteria, fungi and other microorganisms comprise the gut microbiota. Pathogenic viruses inhabiting this niche have the potential to induce local as well as systemic complications; among them, the viral ability to disrupt the mucosal barrier is one mechanism associated with the promotion of diarrhea and tissue invasion. This review gathers recent evidence showing the contributing effects of diet, gut microbiota and the enteric nervous system to either support or impair the mucosal barrier in the context of viral attack.
Collapse
Affiliation(s)
- Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
- Correspondence:
| | - Alejandra López-Aguilera
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
| | - Johana Eyzaguirre-Velásquez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
| | | | - Claudia Ibacache-Quiroga
- Centro de Micro-Bioinnovación (CMBi), Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2340000, Chile;
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile; (A.L.-A.); (J.E.-V.); (J.A.B.)
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile;
| |
Collapse
|
81
|
Solanki SS, Singh P, Kashyap P, Sansi MS, Ali SA. Promising role of defensins peptides as therapeutics to combat against viral infection. Microb Pathog 2021; 155:104930. [PMID: 33933603 PMCID: PMC8084285 DOI: 10.1016/j.micpath.2021.104930] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022]
Abstract
Antimicrobial peptides (AMPs) are ubiquitously present small peptides, which play a critical function in the innate immune system. The defensin class of AMPs represented an evolutionarily ancient family containing cationic cysteine residue and frequently expressed in epithelial or neutrophils cells. It plays myriad functions in host innate immune responses against various infection. Defensin has a broad spectrum of antimicrobial activities, including anti-bacteria, anti-viruses (AVPs), anti-fungi, anti-cancers, and also overcoming bacterial drug resistance. In this review, we compiled the progress on defensin, particularly incorporating the mechanism of action, their application as an antiviral agent, prospects in different areas, and limitations to be solved as an antiviral peptide. Defensins were explored, in particular, their capacity to stimulate innate and adaptive immunity by trigging as anti-coronavirus (COVID-19) peptides. The present review summarised its immunomodulatory and immunoenhancing properties and predominantly focused on its promising therapeutic adjuvant choices for combat against viral infection.
Collapse
Affiliation(s)
| | - Parul Singh
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Poonam Kashyap
- Animal Genomics Lab, National Dairy Research Institute, Karnal, 132001, India
| | - Manish Singh Sansi
- Animal Biochemistry Division, National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, 132001, Haryana, India; Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
82
|
Bakovic A, Risner K, Bhalla N, Alem F, Chang TL, Weston WK, Harness JA, Narayanan A. Brilacidin Demonstrates Inhibition of SARS-CoV-2 in Cell Culture. Viruses 2021; 13:271. [PMID: 33572467 PMCID: PMC7916214 DOI: 10.3390/v13020271] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the newly emergent causative agent of coronavirus disease-19 (COVID-19), has resulted in more than two million deaths worldwide since it was first detected in 2019. There is a critical global need for therapeutic intervention strategies that can be deployed to safely treat COVID-19 disease and reduce associated morbidity and mortality. Increasing evidence shows that both natural and synthetic antimicrobial peptides (AMPs), also referred to as Host Defense Proteins/Peptides (HDPs), can inhibit SARS-CoV-2, paving the way for the potential clinical use of these molecules as therapeutic options. In this manuscript, we describe the potent antiviral activity exerted by brilacidin-a de novo designed synthetic small molecule that captures the biological properties of HDPs-on SARS-CoV-2 in a human lung cell line (Calu-3) and a monkey cell line (Vero). These data suggest that SARS-CoV-2 inhibition in these cell culture models is likely to be a result of the impact of brilacidin on viral entry and its disruption of viral integrity. Brilacidin demonstrated synergistic antiviral activity when combined with remdesivir. Collectively, our data demonstrate that brilacidin exerts potent inhibition of SARS-CoV-2 against different strains of the virus in cell culture.
Collapse
Affiliation(s)
- Allison Bakovic
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Kenneth Risner
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Nishank Bhalla
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Farhang Alem
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| | - Theresa L. Chang
- Public Health Research Institute, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ 07103, USA;
| | - Warren K. Weston
- Innovation Pharmaceuticals Inc., Wakefield, MA 01880, USA; (W.K.W.); (J.A.H.)
| | - Jane A. Harness
- Innovation Pharmaceuticals Inc., Wakefield, MA 01880, USA; (W.K.W.); (J.A.H.)
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA 20110, USA; (A.B.); (K.R.); (N.B.); (F.A.)
| |
Collapse
|
83
|
Scudiero O, Lombardo B, Brancaccio M, Mennitti C, Cesaro A, Fimiani F, Gentile L, Moscarella E, Amodio F, Ranieri A, Gragnano F, Laneri S, Mazzaccara C, Di Micco P, Caiazza M, D’Alicandro G, Limongelli G, Calabrò P, Pero R, Frisso G. Exercise, Immune System, Nutrition, Respiratory and Cardiovascular Diseases during COVID-19: A Complex Combination. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:904. [PMID: 33494244 PMCID: PMC7908487 DOI: 10.3390/ijerph18030904] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Coronaviruses (CoVs) represent a large family of RNA viruses that can infect different living species, posing a global threat to human health. CoVs can evade the immune response, replicate within the host, and cause a rapid immune compromise culminating in severe acute respiratory syndrome. In humans, the immune system functions are influenced by physical activity, nutrition, and the absence of respiratory or cardiovascular diseases. This review provides an in-depth study between the interactions of the immune system and coronaviruses in the host to defend against CoVs disease.
Collapse
Affiliation(s)
- Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (O.S.); (B.L.); (C.M.); (C.M.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
- Ceinge Biotecnologie Avanzate S. C. a R. L., 80131 Naples, Italy; (L.G.); (A.R.)
| | - Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (O.S.); (B.L.); (C.M.); (C.M.)
- Ceinge Biotecnologie Avanzate S. C. a R. L., 80131 Naples, Italy; (L.G.); (A.R.)
| | - Mariarita Brancaccio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Naples, Italy;
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (O.S.); (B.L.); (C.M.); (C.M.)
| | - Arturo Cesaro
- Department of Translational Medical Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (A.C.); (E.M.); (F.A.); (F.G.)
- Division of Clinical Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, 81100 Caserta, Italy
| | - Fabio Fimiani
- Unit of Inherited and Rare Cardiovascular Diseases, Azienda Ospedaliera di Rilievo Nazionale AORN Dei Colli, “V.Monaldi”, 80122 Naples, Italy;
| | - Luca Gentile
- Ceinge Biotecnologie Avanzate S. C. a R. L., 80131 Naples, Italy; (L.G.); (A.R.)
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (A.C.); (E.M.); (F.A.); (F.G.)
- Division of Clinical Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, 81100 Caserta, Italy
| | - Federica Amodio
- Department of Translational Medical Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (A.C.); (E.M.); (F.A.); (F.G.)
| | - Annaluisa Ranieri
- Ceinge Biotecnologie Avanzate S. C. a R. L., 80131 Naples, Italy; (L.G.); (A.R.)
| | - Felice Gragnano
- Department of Translational Medical Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (A.C.); (E.M.); (F.A.); (F.G.)
- Division of Clinical Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, 81100 Caserta, Italy
| | - Sonia Laneri
- Department of Pharmacy, University of Naples Federico II Via Montesano, 80131 Naples, Italy;
| | - Cristina Mazzaccara
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (O.S.); (B.L.); (C.M.); (C.M.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Pierpaolo Di Micco
- Department of Internal Medicine and Emergency Room, Ospedale Buon Consiglio Fatebenefratelli, 80123 Naples, Italy;
| | - Martina Caiazza
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 81100 Naples, Italy;
| | - Giovanni D’Alicandro
- Department of Neuroscience and Rehabilitation, Center of Sports Medicine and Disability, AORN, Santobono-Pausillipon, 80122 Naples, Italy;
| | - Giuseppe Limongelli
- Department of Cardio-Thoracic and Respiratory Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy;
| | - Paolo Calabrò
- Department of Translational Medical Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (A.C.); (E.M.); (F.A.); (F.G.)
- Division of Clinical Cardiology, A.O.R.N. “Sant’Anna e San Sebastiano”, 81100 Caserta, Italy
| | - Raffaela Pero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (O.S.); (B.L.); (C.M.); (C.M.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (O.S.); (B.L.); (C.M.); (C.M.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| |
Collapse
|
84
|
Damour A, Garcia M, Seneschal J, Lévêque N, Bodet C. Eczema Herpeticum: Clinical and Pathophysiological Aspects. Clin Rev Allergy Immunol 2021; 59:1-18. [PMID: 31836943 DOI: 10.1007/s12016-019-08768-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease in the world. AD is a complex pathology mainly characterized by an impaired skin barrier, immune response dysfunction, and unbalanced skin microbiota. Moreover, AD patients exhibit an increased risk of developing bacterial and viral infections. One of the most current, and potentially life-threatening, viral infection is caused by herpes simplex virus (HSV), which occurs in about 3% of AD patients under the name of eczema herpeticum (EH). Following a first part dedicated to the clinical features, virological diagnosis, and current treatments of EH, this review will focus on the description of the pathophysiology and, more particularly, the presently known predisposing factors to herpetic complications in AD patients. These factors include those related to impairment of the skin barrier such as deficit in filaggrin and anomalies in tight and adherens junctions. In addition, low production of the antimicrobial peptides cathelicidin LL-37 and human β-defensins; overexpression of cytokines such as interleukin (IL)-4, IL-13, IL-25, IL-33, and thymic stromal lymphopoietin (TSLP); or downregulation of type I to III interferons as well as defect in functions of immune cells such as dendritic, natural killer, and regulatory T cells have been involved. Otherwise, genetic polymorphisms and AD topical calcineurin inhibitor treatments have been associated with an increased risk of EH. Finally, dysbiosis of skin microbiota characterized in AD patients by Staphylococcus aureus colonization and toxin secretion, such as α-toxin, has been described as promoting HSV replication and could therefore contribute to EH.
Collapse
Affiliation(s)
- Alexia Damour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France
| | - Magali Garcia
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.,Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Julien Seneschal
- INSERM U1035, BMGIC, Immuno-dermatologie ATIP-AVENIR, Bordeaux, France.,Département de Dermatologie and Dermatologie Pédiatrique, Centre national de référence pour les maladies rares de la peau, Hôpital Saint-André, Bordeaux, France
| | - Nicolas Lévêque
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.,Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.
| |
Collapse
|
85
|
Pedan H, Janosova V, Hajtman A, Calkovsky V. Non-Reflex Defense Mechanisms of Upper Airway Mucosa: Possible Clinical Application. Physiol Res 2021; 69:S55-S67. [PMID: 32228012 DOI: 10.33549/physiolres.934404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The sinonasal mucosa has an essential role in defense mechanisms of the upper respiratory tract. The innate immune system presents the primary defense against noxious microorganisms followed by induction of the adaptive immune mechanisms as a consequence of the presence of pathogens. This well-known activation of adaptive immune system in response to presence of the antigen on mucosal surfaces is now broadly applicated in vaccinology research. Prevention of infectious diseases belongs to substantial challenges in maintaining the population health. Non-invasive, easily applicable mucosal vaccination purposes various research opportunities that could be usable in daily practice. However, the existence of multiple limitations such as rapid clearance of vaccine from nasal mucosa by means of mucociliary transport represents a great challenge in development of safe and efficient vaccines. Here we give an updated view on nasal functions with focus on nasal mucosal immunity and its potential application in vaccination in nearly future.
Collapse
Affiliation(s)
- H Pedan
- Clinic of Otorhinolaryngology and Head and Neck Surgery, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, and Martin University Hospital, Martin, Slovak
| | | | | | | |
Collapse
|
86
|
Murugan NA, Raja KMP, Saraswathi NT. Peptide-Based Antiviral Drugs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:261-284. [PMID: 34258744 DOI: 10.1007/978-981-16-0267-2_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Three types of chemical entities, namely, small organic molecules (organics), peptides, and biologics, are mainly used as drug candidates for the treatment of various diseases. Even though the peptide drugs are known since 1920 in association with the clinical use of insulin, only a limited number of peptides are currently used for therapeutics due to various disadvantages associated with them such as limited serum and blood stability, oral bioavailability, and permeability. Since, through chemical modifications and structure tuning, many of these limitations can be overcome, peptide-based drugs are gaining attention in pharmaceutical research. As of today, there are more than 60 peptide-based drugs approved by FDA, and over 150 peptides are in the advanced clinical studies. In this book chapter, the peptide-based lead compounds and drugs available for treating various viral diseases and their advantages and disadvantages when compared to small molecules drugs are discussed.
Collapse
Affiliation(s)
- N Arul Murugan
- Department of Theoretical Chemistry and Biology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - K Muruga Poopathi Raja
- Chemical Biology and Biophysics Laboratory, Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, Tamilnadu, India.
| | - N T Saraswathi
- School of Chemical & Biotechnology, Sastra Deemed University, Thanjavur, Tamil Nadu, India
| |
Collapse
|
87
|
Emamifar S, Abolmaali S, Mohsen Sohrabi S, Mohammadi M, Shahmohammadi M. Molecular characterization and evaluation of the antibacterial activity of a plant defensin peptide derived from a gene of oat (Avena sativa L.). PHYTOCHEMISTRY 2021; 181:112586. [PMID: 33232862 DOI: 10.1016/j.phytochem.2020.112586] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 06/11/2023]
Abstract
Plant defensins are a group of small disulfide-rich cationic peptides that exhibit a broad spectrum of antimicrobial activities. In the present study, an antibacterial plant defensin peptide was successfully identified and characterized from the transcriptome of the oat (Avena sativa L.), and called AsDef1. The complete nucleotide sequence of AsDef1 was determined (321 bp) and found to contain an open reading frame (ORF) encoding a peptide of 77 aa with a putative 22 aa signal peptide sequence that addresses the mature defensin to the apoplast. Further in silico analyses revealed that the structure of the identified defensin (AsDef1) consists of the Knot1 functional domain with eight conserved cysteine residues and four disulfide bonds. The highest expression of AsDef1 was observed in the developing seeds of the A. sativa plant. AsDef1 also showed antibacterial activity against both Gram-positive and Gram-negative bacteria. The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values ranged from 0.15625 μM to 0.625 μM. In this study, we identified and characterized an antibacterial defensin from A. sativa for the first time. The findings of the present study offer insights that can be used in producing pathogen-resistant transgenic plants and in developing potential antibacterial agents in the future using AsDef1 from A. sativa.
Collapse
Affiliation(s)
- Sara Emamifar
- Department of Cell and Molecular Biology, Faculty of Basic Sciences, Semnan University, Semnan, Iran.
| | - Shamsozoha Abolmaali
- Department of Cell and Molecular Biology, Faculty of Basic Sciences, Semnan University, Semnan, Iran.
| | - Seyyed Mohsen Sohrabi
- Young Researchers and Elite Club, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran.
| | - Mohsen Mohammadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Maasume Shahmohammadi
- Razi Herbal Medicines Research Center and Department of Medical Biotechnology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
88
|
Liu K, Hou F, Wahaab A, Kang L, Xie F, Ma X, Xia Q, Xiao C, Shao D, Li B, Wei J, Qiu Y, Zhu H, Ma Z. Mosquito defensin facilitates Japanese encephalitis virus infection by downregulating the C6/36 cell-surface antiviral protein HSC70B. Vet Microbiol 2020; 253:108971. [PMID: 33385886 DOI: 10.1016/j.vetmic.2020.108971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/20/2020] [Indexed: 12/31/2022]
Abstract
Japanese encephalitis virus (JEV) is a viral zoonosis that can cause viral encephalitis, death and disability whose primary vector is the Culex mosquito. Viral infection induces a series of antimicrobial peptide responses in mosquitoes, and the effector defensin enhances JEV replication in mosquitoes. However, the underlying mechanisms by which defensin enhances JEV are not fully understood. Here, we found that mosquito defensin could downregulate the antiviral protein HSC70B and enhance virus infection in mosquitoes. The cell-surface protein HSC70B was significantly downregulated by JEV infection and defensin treatment. Low levels of HSC70B were beneficial to JEV infection in mosquitoes. Taken together, these findings show that defensin and HSC70B axis facilitates JEV infection in the mosquito.
Collapse
Affiliation(s)
- Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Fengxiang Hou
- Wenzhou Vocational College of Science & Technology (Wenzhou Academy of Agricultural Sciences), Wenzhou, Zhejiang Province, PR China
| | - Abdul Wahaab
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Lei Kang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Fengyu Xie
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Xiaochun Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Qiqi Xia
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Changguang Xiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Huaimin Zhu
- Department of Pathogen Biology. Second Military Medical University. Shanghai, 200433, PR China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China.
| |
Collapse
|
89
|
Tahamtan A, Besteman S, Samadizadeh S, Rastegar M, Bont L, Salimi V. Neutrophils in respiratory syncytial virus infection: From harmful effects to therapeutic opportunities. Br J Pharmacol 2020; 178:515-530. [PMID: 33169387 DOI: 10.1111/bph.15318] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is an important infectious agent in infants and young children. In most cases, RSV infection only causes mild disease, but in some, it requires invasive ventilation. Although antiviral drugs are obvious candidates to treat viral illness, and some have shown antiviral effects in humans, antivirals such as GS-5806, ALX-0171 and ALS-8176 have not yet met their expectations. Since the inappropriate or dysregulated immune response against RSV leads to harmful immune pathology, a robust immune cascade is probably underway by the time patients reach the hospital. RSV infection is associated with a strong neutrophil influx into the airway. It not clear if these cells contribute to antiviral defence or to lung pathology. This article discusses the protective and harmful roles of neutrophils during RSV infection and provides an overview of mechanisms by which neutrophil function could be targeted to prevent tissue injury and preserve homeostasis.
Collapse
Affiliation(s)
- Alireza Tahamtan
- Infectious Diseases Research Centre, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sjanna Besteman
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands.,Center for Translation Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Saeed Samadizadeh
- Infectious Diseases Research Centre, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mostafa Rastegar
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Louis Bont
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
90
|
Nikoloudakis N, Pappi P, Markakis EA, Charova SN, Fanourakis D, Paschalidis K, Delis C, Tzortzakakis EA, Tsaniklidis G. Structural Diversity and Highly Specific Host-Pathogen Transcriptional Regulation of Defensin Genes Is Revealed in Tomato. Int J Mol Sci 2020; 21:ijms21249380. [PMID: 33317090 PMCID: PMC7764197 DOI: 10.3390/ijms21249380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 01/17/2023] Open
Abstract
Defensins are small and rather ubiquitous cysteine-rich anti-microbial peptides. These proteins may act against pathogenic microorganisms either directly (by binding and disrupting membranes) or indirectly (as signaling molecules that participate in the organization of the cellular defense). Even though defensins are widespread across eukaryotes, still, extensive nucleotide and amino acid dissimilarities hamper the elucidation of their response to stimuli and mode of function. In the current study, we screened the Solanum lycopersicum genome for the identification of defensin genes, predicted the relating protein structures, and further studied their transcriptional responses to biotic (Verticillium dahliae, Meloidogyne javanica, Cucumber Mosaic Virus, and Potato Virus Y infections) and abiotic (cold stress) stimuli. Tomato defensin sequences were classified into two groups (C8 and C12). Our data indicate that the transcription of defensin coding genes primarily depends on the specific pathogen recognition patterns of V. dahliae and M. javanica. The immunodetection of plant defensin 1 protein was achieved only in the roots of plants inoculated with V. dahliae. In contrast, the almost null effects of viral infections and cold stress, and the failure to substantially induce the gene transcription suggest that these factors are probably not primarily targeted by the tomato defensin network.
Collapse
Affiliation(s)
- Nikolaos Nikoloudakis
- Department of Agricultural Science, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus
- Correspondence: (N.N.); (G.T.)
| | - Polyxeni Pappi
- Department of Viticulture, Vegetable Crops, Floriculture and Plant Protection, Institute of Olive Tree, Subtropical Crops and Viticulture, Hellenic Agricultural Organization ELGO-DIMITRA, Mesa Katsabas, 71307 Heraklion, Crete, Greece; (P.P.); (E.A.M.); (E.A.T.)
| | - Emmanouil A. Markakis
- Department of Viticulture, Vegetable Crops, Floriculture and Plant Protection, Institute of Olive Tree, Subtropical Crops and Viticulture, Hellenic Agricultural Organization ELGO-DIMITRA, Mesa Katsabas, 71307 Heraklion, Crete, Greece; (P.P.); (E.A.M.); (E.A.T.)
| | - Spyridoula N. Charova
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), 70013 Heraklion, Crete, Greece;
- Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Dimitrios Fanourakis
- Giannakakis SA, Export Fruits and Vegetables, 70200 Tympaki, Crete, Greece;
- School of Agricultural Sciences, Hellenic Mediterranean University, Estavromenos, 71004 Heraklion, Crete, Greece;
| | - Konstantinos Paschalidis
- School of Agricultural Sciences, Hellenic Mediterranean University, Estavromenos, 71004 Heraklion, Crete, Greece;
| | - Costas Delis
- Department of Agricultural Technology, School of Agricultural Technology and Food Technology and Nutrition, University of Peloponnese, 24100 Antikalamos, Kalamata, Greece;
| | - Emmanuel A. Tzortzakakis
- Department of Viticulture, Vegetable Crops, Floriculture and Plant Protection, Institute of Olive Tree, Subtropical Crops and Viticulture, Hellenic Agricultural Organization ELGO-DIMITRA, Mesa Katsabas, 71307 Heraklion, Crete, Greece; (P.P.); (E.A.M.); (E.A.T.)
| | - Georgios Tsaniklidis
- Department of Viticulture, Vegetable Crops, Floriculture and Plant Protection, Institute of Olive Tree, Subtropical Crops and Viticulture, Hellenic Agricultural Organization ELGO-DIMITRA, Mesa Katsabas, 71307 Heraklion, Crete, Greece; (P.P.); (E.A.M.); (E.A.T.)
- Correspondence: (N.N.); (G.T.)
| |
Collapse
|
91
|
Xiao B, Fu Q, Niu S, Zhu P, He J, Li C. Penaeidins restrict white spot syndrome virus infection by antagonizing the envelope proteins to block viral entry. Emerg Microbes Infect 2020; 9:390-412. [PMID: 32397950 PMCID: PMC7048182 DOI: 10.1080/22221751.2020.1729068] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Emerging studies have indicated that some penaeidins restrict virus infection; however, the mechanism(s) involved are poorly understood. In the present study, we uncovered that penaeidins are a novel family of antiviral effectors against white spot syndrome virus (WSSV), which antagonize the envelope proteins to block viral entry. We found that the expression levels of four identified penaeidins from Litopenaeus vannamei, including BigPEN, PEN2, PEN3, and PEN4, were significantly induced in hemocytes during the early stage of WSSV infection. Knockdown of each penaeidin in vivo via RNA interference resulted in elevated viral loads and rendered shrimp more susceptible to WSSV, while the survival rate was rescued via the injection of recombinant penaeidins. All penaeidins, except PEN4, were shown to interact with several envelope proteins of WSSV, and all four penaeidins were observed to be located on the outer surface of the WSSV virion. Co-incubation of each recombinant penaeidin with WSSV inhibited virion internalization into hemocytes. More importantly, we found that PEN2 competitively bound to the envelope protein VP24 to release it from polymeric immunoglobulin receptor (pIgR), the cellular receptor required for WSSV infection. Moreover, we also demonstrated that BigPEN was able to bind to VP28 of WSSV, which disrupted the interaction between VP28 and Rab7 – the Rab GTPase that contributes to viral entry by binding with VP28. Taken together, our results demonstrated that penaeidins interact with the envelope proteins of WSSV to block multiple viral infection processes, thereby protecting the host against WSSV.
Collapse
Affiliation(s)
- Bang Xiao
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China.,State Key Laboratory of Biocontrol/ School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China
| | - Qihui Fu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China.,State Key Laboratory of Biocontrol/ School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China
| | - Shengwen Niu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China.,State Key Laboratory of Biocontrol/ School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China
| | - Peng Zhu
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, Beibu Gluf University, Qinzhou, P. R. People's Republic of China
| | - Jianguo He
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China.,State Key Laboratory of Biocontrol/ School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China
| | - Chaozheng Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China.,State Key Laboratory of Biocontrol/ School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. People's Republic of China
| |
Collapse
|
92
|
Schütz D, Ruiz-Blanco YB, Münch J, Kirchhoff F, Sanchez-Garcia E, Müller JA. Peptide and peptide-based inhibitors of SARS-CoV-2 entry. Adv Drug Deliv Rev 2020; 167:47-65. [PMID: 33189768 PMCID: PMC7665879 DOI: 10.1016/j.addr.2020.11.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022]
Abstract
To date, no effective vaccines or therapies are available against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative pandemic agent of the coronavirus disease 2019 (COVID-19). Due to their safety, efficacy and specificity, peptide inhibitors hold great promise for the treatment of newly emerging viral pathogens. Based on the known structures of viral proteins and their cellular targets, antiviral peptides can be rationally designed and optimized. The resulting peptides may be highly specific for their respective targets and particular viral pathogens or exert broad antiviral activity. Here, we summarize the current status of peptides inhibiting SARS-CoV-2 entry and outline the strategies used to design peptides targeting the ACE2 receptor or the viral spike protein and its activating proteases furin, transmembrane serine protease 2 (TMPRSS2), or cathepsin L. In addition, we present approaches used against related viruses such as SARS-CoV-1 that might be implemented for inhibition of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Desiree Schütz
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Yasser B Ruiz-Blanco
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, 45117 Essen, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, 45117 Essen, Germany.
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany.
| |
Collapse
|
93
|
Nikolenko VN, Oganesyan MV, Sankova MV, Bulygin KV, Vovkogon AD, Rizaeva NA, Sinelnikov MY. Paneth cells: Maintaining dynamic microbiome-host homeostasis, protecting against inflammation and cancer. Bioessays 2020; 43:e2000180. [PMID: 33244814 DOI: 10.1002/bies.202000180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022]
Abstract
The human intestines are constantly under the influence of numerous pathological factors: enteropathogenic microorganisms, food antigens, physico-chemical stress associated with digestion and bacterial metabolism, therefore it must be provided with a system of protection against adverse impact. Recent studies have shown that Paneth cells play a crucial role in maintaining homeostasis of the small intestines. Paneth cells perform many vital functions aimed at maintaining a homeostatic balance between normal microbiota, infectious pathogens and the human body, regulate the qualitative composition and number of intestinal microorganisms, prevent the introduction of potentially pathogenic species, and protect stem cells from damage. Paneth cells take part in adaptive and protective-inflammatory reactions. Paneth cells maintain dynamic balance between microbial populations, and the macroorganism, preventing the development of intestinal infections and cancer. They play a crucial role in gastrointestinal homeostasis and may be key factors in the etiopathological progression of intestinal diseases.
Collapse
Affiliation(s)
- Vladimir N Nikolenko
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia.,Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Marine V Oganesyan
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Maria V Sankova
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia.,Department of Normal and Topographic Anatomy, Lomonosov Moscow State University, Moscow, Russia
| | - Andzhela D Vovkogon
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | - Negoriya A Rizaeva
- Department of Human Anatomy, First Moscow State Medical University named after I.M.Sechenov (Sechenov University), Moscow, Russia
| | | |
Collapse
|
94
|
Diaz K, Hu CT, Sul Y, Bromme BA, Myers ND, Skorohodova KV, Gounder AP, Smith JG. Defensin-driven viral evolution. PLoS Pathog 2020; 16:e1009018. [PMID: 33232373 PMCID: PMC7723274 DOI: 10.1371/journal.ppat.1009018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 12/08/2020] [Accepted: 10/02/2020] [Indexed: 01/06/2023] Open
Abstract
Enteric alpha-defensins are potent effectors of innate immunity that are abundantly expressed in the small intestine. Certain enteric bacteria and viruses are resistant to defensins and even appropriate them to enhance infection despite neutralization of closely related microbes. We therefore hypothesized that defensins impose selective pressure during fecal-oral transmission. Upon passaging a defensin-sensitive serotype of adenovirus in the presence of a human defensin, mutations in the major capsid protein hexon accumulated. In contrast, prior studies identified the vertex proteins as important determinants of defensin antiviral activity. Infection and biochemical assays suggest that a balance between increased cell binding and a downstream block in intracellular trafficking mediated by defensin interactions with all of the major capsid proteins dictates the outcome of infection. These results extensively revise our understanding of the interplay between defensins and non-enveloped viruses. Furthermore, they provide a feasible rationale for defensins shaping viral evolution, resulting in differences in infection phenotypes of closely related viruses. Defensins are potent antimicrobial peptides that are found on human mucosal surfaces and can directly neutralize viruses. They are abundant in the small intestine, which is constantly challenged by ingested viral pathogens. Interestingly, non-enveloped viruses, such as adenovirus, that infect the gastrointestinal system are unaffected by defensins or can even appropriate defensins to enhance their infection. In contrast, respiratory adenoviruses are neutralized by the same defensins. How enteric viruses overcome defensin neutralization is not well understood. Our studies are the first to show that defensins can drive the evolution of non-enveloped viruses. Furthermore, we identify important components within human adenovirus that dictate sensitivity to defensins. This new insight into defensin-virus interactions informs our understanding of mucosal immunity to viral infections.
Collapse
Affiliation(s)
- Karina Diaz
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ciara T. Hu
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Youngmee Sul
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Beth A. Bromme
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Nicolle D. Myers
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ksenia V. Skorohodova
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Anshu P. Gounder
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jason G. Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
95
|
Skeate JG, Segerink WH, Garcia MD, Fernandez DJ, Prins R, Lühen KP, Voss FO, Da Silva DM, Kast WM. Theta-Defensins Inhibit High-Risk Human Papillomavirus Infection Through Charge-Driven Capsid Clustering. Front Immunol 2020; 11:561843. [PMID: 33154746 PMCID: PMC7586039 DOI: 10.3389/fimmu.2020.561843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/24/2020] [Indexed: 11/26/2022] Open
Abstract
Persistent infection with high-risk human papillomavirus (hrHPV) genotypes results in a large number of anogenital and head and neck cancers worldwide. Although prophylactic vaccination coverage has improved, there remains a need to develop methods that inhibit viral transmission toward preventing the spread of HPV-driven disease. Defensins are a class of innate immune effector peptides that function to protect hosts from infection by pathogens such as viruses and bacteria. Previous work utilizing α and β defensins from humans has demonstrated that the α-defensin HD5 is effective at inhibiting the most common high-risk genotype, HPV16. A third class of defensin that has yet to be explored are θ-defensins: small, 18-amino acid cyclic peptides found in old-world monkeys whose unique structure makes them both highly cationic and resistant to degradation. Here we show that the prototype θ-defensin, rhesus theta defensin 1, inhibits hrHPV infection through a mechanism involving capsid clustering that inhibits virions from binding to cell surface receptor complexes.
Collapse
Affiliation(s)
- Joseph G Skeate
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Wouter H Segerink
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Mauricio D Garcia
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Daniel J Fernandez
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ruben Prins
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kim P Lühen
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Féline O Voss
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Diane M Da Silva
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States.,Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - W Martin Kast
- Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States.,Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
96
|
Drab E, Sugihara K. Cooperative Function of LL-37 and HNP1 Protects Mammalian Cell Membranes from Lysis. Biophys J 2020; 119:2440-2450. [PMID: 33157121 DOI: 10.1016/j.bpj.2020.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/08/2020] [Indexed: 01/10/2023] Open
Abstract
LL-37, cleaved from human cathelicidin, and human neutrophil peptide-1 (HNP1) from the defensin family are antimicrobial peptides that are occasionally co-released from neutrophils, which synergistically kill bacteria. We report that this couple presents another type of cooperativity against host eukaryotic cells, in which they antagonistically minimize cytotoxicity by protecting membranes from lysis. Our results describe the potential of the LL-37/HNP1 cooperativity that switches from membrane-destructive to membrane-protective functions, depending on whether the target is an enemy or a host.
Collapse
Affiliation(s)
- Ewa Drab
- Department of Physical Chemistry, University of Geneva, Geneva, Switzerland
| | - Kaori Sugihara
- Institute of Industrial Science, The University of Tokyo, Tokyo, Japan; Department of Physical Chemistry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
97
|
Kerget B, Kerget F, Aksakal A, Aşkın S, Sağlam L, Akgün M. Evaluation of alpha defensin, IL-1 receptor antagonist, and IL-18 levels in COVID-19 patients with macrophage activation syndrome and acute respiratory distress syndrome. J Med Virol 2020; 93:2090-2098. [PMID: 33038012 PMCID: PMC7675303 DOI: 10.1002/jmv.26589] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/22/2020] [Accepted: 10/07/2020] [Indexed: 01/10/2023]
Abstract
Background Many laboratory parameters have been associated with morbidity and mortality in SARS‐CoV‐2 (COVID‐19), which emerged in an animal market in Wuhan, China in December 2019 and has infected over 20 million people. This study investigated the relationship between serum interleukin (IL)‐18, IL‐1 receptor antagonist (IL‐1Ra), and alpha defensin levels and the clinical course and prognosis of COVID‐19. Materials and Methods This study included 100 patients who were admitted to the chest diseases department and intensive care unit of our hospital and diagnosed with COVID‐19 by real‐time polymerase chain reaction (PCR) of nasopharyngeal swab samples between March 24 and May 31, 2020. The control group consisted of 50 nonsymptomatic health workers with negative real‐time PCR results in routine COVID‐19 screening in our hospital. Results Serum alpha defensin, IL‐1Ra, and IL‐18 levels were significantly higher in patients who developed macrophage activation syndrome (MAS) and acute respiratory distress syndrome (ARDS) compared to patients who did not (p < .001 for all). Alpha defensin, IL‐1Ra, and IL‐18 levels were significantly higher in COVID‐19 patients with and without MAS or ARDS when compared to the control group (p < .001 for all). When the 9 patients who died were compared with the 91 surviving patients, IL‐1Ra and IL‐18 levels were found to be significantly higher in the nonsurvivors (p < .001). Conclusion Our findings of correlations between alpha defensin and levels of IL‐1Ra and IL‐18, which were previously shown to be useful in COVID‐19 treatment and follow‐up, indicates that it may also be promising in treatment.
Collapse
Affiliation(s)
- Buğra Kerget
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Ferhan Kerget
- Depertmant of Infection Diseases and Clinical Microbiology, Health Sciences University, Erzurum Regional Education and Research Hospital, Erzurum, Turkey
| | - Alperen Aksakal
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Seda Aşkın
- Department of Biochemistry, Ataturk University School of Medicine, Erzurum, Turkey
| | - Leyla Sağlam
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Metin Akgün
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| |
Collapse
|
98
|
Liu K, Xiao C, Xi S, Hameed M, Wahaab A, Shao D, Li Z, Li B, Wei J, Qiu Y, Miao D, Zhu H, Ma Z. Mosquito Defensins Enhance Japanese Encephalitis Virus Infection by Facilitating Virus Adsorption and Entry within the Mosquito. J Virol 2020; 94:e01164-20. [PMID: 32796073 PMCID: PMC7565626 DOI: 10.1128/jvi.01164-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/02/2020] [Indexed: 12/11/2022] Open
Abstract
Japanese encephalitis virus (JEV) is a viral zoonosis that can cause viral encephalitis, death, and disability. Although the Culex mosquito is the primary vector of JEV, little is known about JEV transmission by this kind of mosquito. Here, we found that mosquito defensin facilitated the adsorption of JEV on target cells via the defensin/lipoprotein receptor-related protein 2 (LRP2) axis. Mosquito defensin bound the ED III domain of the viral envelope (E) protein and directly mediated efficient virus adsorption on the target cell surface; the receptor LRP2, which is expressed on the cell surface, affected defensin-dependent adsorption. As a result, mosquito defensin enhanced JEV infection in the salivary gland, increasing the possibility of viral transmission by mosquitoes. These findings demonstrate the novel role of mosquito defensin in JEV infection and the mechanisms through which the virus exploits mosquito defensin for infection and transmission.IMPORTANCE In this study, we observed the complex roles of mosquito defensin in JEV infection; mosquito defensin exhibited a weak antiviral effect but strongly enhanced binding. In the latter, defensin directly binds the ED III domain of the viral E protein and promotes the adsorption of JEV to target cells by interacting with lipoprotein receptor-related protein 2 (LRP2), thus accelerating virus entry. Together, our results indicate that mosquito defensin plays an important role in facilitating JEV infection and potential transmission.
Collapse
Affiliation(s)
- Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Changguang Xiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Shumin Xi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Muddassar Hameed
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Abdul Wahaab
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Zongjie Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| | - Denian Miao
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, People's Republic of China
| | - Huaimin Zhu
- Department of Pathogen Biology, Second Military Medical University, Shanghai, People's Republic of China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, People's Republic of China
| |
Collapse
|
99
|
Overview of Host Defense Peptides and Their Applications for Plastic and Reconstructive Surgeons. Plast Reconstr Surg 2020; 146:91-103. [PMID: 32590651 DOI: 10.1097/prs.0000000000006910] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Host defense peptides are a family of endogenous short peptides that are found in all living beings and play a critical role in innate immunity against infection. METHODS A nonsystematic review of host defense peptides was conducted with specific interest in properties and applications relevant to plastic and reconstructive surgery. RESULTS In addition to their direct antimicrobial actions against pathogens, including multidrug-resistant bacteria, they also demonstrate important functions in immunomodulation, tumor cell lysis, and tissue regeneration. These properties have made them a topic of clinical interest for plastic surgeons because of their potential applications as novel antibiotics, wound healing medications, and cancer therapies. The rising clinical interest has led to a robust body of literature describing host defense peptides in great depth and breadth. Numerous mechanisms have been observed to explain their diverse functions, which rely on specific structural characteristics. However, these peptides remain mostly experimental, with limited translation to clinical practice because of numerous failures to achieve acceptable results in human trials. CONCLUSIONS Despite the broad ranging potential of these peptides for use in the field of plastic and reconstructive surgery, they are rarely discussed in the literature or at scientific meetings. In this review, the authors provide a summary of the background, structure, function, bacterial resistance, and clinical applications of host defense peptides with the goal of stimulating host defense peptide-based innovation within the field of plastic and reconstructive surgery.
Collapse
|
100
|
Daugavet MA, Shabelnikov SV, Podgornaya OI. Amino acid sequence associated with bacteriophage recombination site helps to reveal genes potentially acquired through horizontal gene transfer. BMC Bioinformatics 2020; 21:305. [PMID: 32703190 PMCID: PMC7379824 DOI: 10.1186/s12859-020-03599-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/04/2022] Open
Abstract
Background Horizontal gene transfer, i.e. the acquisition of genetic material from nonparent organism, is considered an important force driving species evolution. Many cases of horizontal gene transfer from prokaryotes to eukaryotes have been registered, but no transfer mechanism has been deciphered so far, although viruses were proposed as possible vectors in several studies. In agreement with this idea, in our previous study we discovered that in two eukaryotic proteins bacteriophage recombination site (AttP) was adjacent to the regions originating via horizontal gene transfer. In one of those cases AttP site was present inside the introns of cysteine-rich repeats. In the present study we aimed to apply computational tools for finding multiple horizontal gene transfer events in large genome databases. For that purpose we used a sequence of cysteine-rich repeats to identify genes potentially acquired through horizontal transfer. Results HMMER remote similarity search significantly detected 382 proteins containing cysteine-rich repeats. All of them, except 8 sequences, belong to eukaryotes. In 124 proteins the presence of conserved structural domains was predicted. In spite of the fact that cysteine-rich repeats are found almost exclusively in eukaryotic proteins, many predicted domains are most common for prokaryotes or bacteriophages. Ninety-eight proteins out of 124 contain typical prokaryotic domains. In those cases proteins were considered as potentially originating via horizontal transfer. In addition, HHblits search revealed that two domains of the same fungal protein, Glycoside hydrolase and Peptidase M15, have high similarity with proteins of two different prokaryotic species, hinting at independent horizontal gene transfer events. Conclusions Cysteine-rich repeats in eukaryotic proteins are usually accompanied by conserved domains typical for prokaryotes or bacteriophages. These proteins, containing both cysteine-rich repeats, and characteristic prokaryotic domains, might represent multiple independent horizontal gene transfer events from prokaryotes to eukaryotes. We believe that the presence of bacteriophage recombination site inside cysteine-rich repeat coding sequence may facilitate horizontal genes transfer. Thus computational approach, described in the present study, can help finding multiple sequences originated from horizontal transfer in eukaryotic genomes.
Collapse
Affiliation(s)
| | | | - Olga I Podgornaya
- Institute of Cytology, St. Petersburg, Russia, 194064.,School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia, 690090.,Department of Cytology and Histology, St. Pb State University, St. Petersburg, Russia, 199034
| |
Collapse
|