1
|
Nagib M, Sayed AM, Korany AH, Abdelkader K, Shari FH, Mackay WG, Rateb ME. Human Defensins: Structure, Function, and Potential as Therapeutic Antimicrobial Agents with Highlights Against SARS CoV-2. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10436-8. [PMID: 39693007 DOI: 10.1007/s12602-024-10436-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
The human defensins are a group of cationic antimicrobial peptides that range in size from 2 to 5 kDa and share a common structural motif of six disulphide-linked cysteines. Several naturally occurring human α- and β-defensins have been identified over the past two decades. They have a wide variety of antimicrobial effects, and their potential to avoid the development of resistance to antimicrobial treatment makes them attractive as therapeutic agents. Human defensins have recently been the focus of medical and molecular biology studies due to their promising application in medicine and the pharmaceutical industry. This work aims to provide a comprehensive summary of the current developments of human defensins, including their identification, categorization, molecular features, expression, modes of action, and potential application in medical settings. Current obstacles and future opportunities for using human defensins are also covered. Furthermore, we shed light on the potential of this class as an antiviral agent, particularly against SARS CoV-2, by providing an in silico-based investigation of their plausible mechanisms of action.
Collapse
Affiliation(s)
- Maryam Nagib
- School of Computing Engineering and Physical Sciences, University of the West of Scotland, Paisley, Scotland, PA12BE, UK
| | - Ahmed M Sayed
- Department of Pharmacognosy, College of Pharmacy, Almaaqal University, Basrah, 61014, Iraq
| | - Ahmed H Korany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Nahda University, Beni Suef, 62513, Egypt
| | - Karim Abdelkader
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62511, Egypt
| | - Falah H Shari
- Department of Clinical Biochemistry, College of Pharmacy, Almaaqal University, Basrah, 61014, Iraq
| | - William G Mackay
- School of Health and Life Sciences, University of the West of Scotland, Blantyre, Glasgow, G72 0LH, UK
| | - Mostafa E Rateb
- School of Computing Engineering and Physical Sciences, University of the West of Scotland, Paisley, Scotland, PA12BE, UK.
| |
Collapse
|
2
|
Zhou D, Zhang C, Sun J, Yuan M. Neutrophils in oncolytic virus immunotherapy. Front Immunol 2024; 15:1490414. [PMID: 39697335 PMCID: PMC11652357 DOI: 10.3389/fimmu.2024.1490414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Oncolytic viruses have emerged as a highly promising modality for cancer treatment due to their ability to replicate specifically within tumors, carry therapeutic genes, and modulate the immunosuppressive tumor microenvironment through various mechanisms. Additionally, they show potential synergy with immune checkpoint inhibitors. A study report indicates that from 2000 to 2020, 49.5% of oncolytic viruses were administered intratumorally and 35% intravenously during clinical trials. However, both administration methods face significant challenges, particularly with intravenous delivery, which encounters issues such as non-specific tissue uptake, neutralizing antibody responses, and antiviral effects mediated by various immune cells. Despite extensive research into the antiviral roles of CD8+ T cells and NK cells in oncolytic virus therapy, neutrophils-constituting approximately 50% to 70% of human peripheral blood leukocytes-have received relatively little attention. Neutrophils are the most abundant leukocyte subset in peripheral circulation, known for their phagocytic activity. Beyond their traditional roles in bacterial and fungal infections, emerging literature suggests that neutrophils also play a critical role in the body's antiviral responses. Given the gaps in understanding the role of neutrophils in oncolytic virus therapy, this article reviews current literature on this topic. It aims to provide a theoretical foundation for developing oncolytic virus-based cancer therapies and enhancing their anti-tumor efficacy in future clinical treatments.
Collapse
Affiliation(s)
- Danya Zhou
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University) Ministry of Education, Hefei, Anhui, China
| | - Chenglin Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingyi Sun
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Yuan
- Huayao Kangming Biopharmaceutical Co., Ltd, Shenzhen, China
| |
Collapse
|
3
|
Di YP, Kuhn JM, Mangoni ML. Lung antimicrobial proteins and peptides: from host defense to therapeutic strategies. Physiol Rev 2024; 104:1643-1677. [PMID: 39052018 PMCID: PMC11495187 DOI: 10.1152/physrev.00039.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 06/11/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Representing severe morbidity and mortality globally, respiratory infections associated with chronic respiratory diseases, including complicated pneumonia, asthma, interstitial lung disease, and chronic obstructive pulmonary disease, are a major public health concern. Lung health and the prevention of pulmonary disease rely on the mechanisms of airway surface fluid secretion, mucociliary clearance, and adequate immune response to eradicate inhaled pathogens and particulate matter from the environment. The antimicrobial proteins and peptides contribute to maintaining an antimicrobial milieu in human lungs to eliminate pathogens and prevent them from causing pulmonary diseases. The predominant antimicrobial molecules of the lung environment include human α- and β-defensins and cathelicidins, among numerous other host defense molecules with antimicrobial and antibiofilm activity such as PLUNC (palate, lung, and nasal epithelium clone) family proteins, elafin, collectins, lactoferrin, lysozymes, mucins, secretory leukocyte proteinase inhibitor, surfactant proteins SP-A and SP-D, and RNases. It has been demonstrated that changes in antimicrobial molecule expression levels are associated with regulating inflammation, potentiating exacerbations, pathological changes, and modifications in chronic lung disease severity. Antimicrobial molecules also display roles in both anticancer and tumorigenic effects. Lung antimicrobial proteins and peptides are promising alternative therapeutics for treating and preventing multidrug-resistant bacterial infections and anticancer therapies.
Collapse
Affiliation(s)
- Yuanpu Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jenna Marie Kuhn
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
4
|
Ziu T, Sambur E, Ruzsics Z, Hengel H, Grabherr R, Höfinger S, Harant H. In Vitro Profiling of the Antiviral Peptide TAT-I24. Int J Mol Sci 2024; 25:10463. [PMID: 39408791 PMCID: PMC11477294 DOI: 10.3390/ijms251910463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The synthetic peptide TAT-I24 (GRKKRRQRRRPPQCLAFYACFC) exerts antiviral activity against several double-stranded (ds) DNA viruses, including herpes simplex viruses, cytomegalovirus, some adenoviruses, vaccinia virus and SV40 polyomavirus. In the present study, in vitro profiling of this peptide was performed with the aim of characterizing and improving its properties for further development. As TAT-I24 contains three free cysteine residues, a potential disadvantageous feature, peptide variants with replacements or deletions of specific residues were generated and tested in various cell systems and by biochemical analyses. Some cysteine replacements had no impact on the antiviral activity, such as the deletion of cysteine 14, which also showed improved biochemical properties, while the cyclization of cysteines 14 and 20 had the most detrimental effect on antiviral activity. At concentrations below 20 µM, TAT-I24 and selected variants did not induce hemolysis in red blood cells (RBCs) nor modulated lipopolysaccharide (LPS)-induced release of cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), in human peripheral blood mononuclear cells (PBMCs). These data indicate that TAT-I24 or its peptide variants are not expected to cause unwanted effects on blood cells.
Collapse
Affiliation(s)
- Theodhora Ziu
- Pivaris BioScience GmbH, Media Quarter Marx 3.4, Maria-Jacobi-Gasse 1, 1030 Vienna, Austria;
| | - Ezgi Sambur
- VSC Research Center, Technical University of Vienna, Operngasse 11/E057-09, 1040 Vienna, Austria; (E.S.); or (S.H.)
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Hermann-Herder-Str.11, 79104 Freiburg, Germany; (Z.R.); (H.H.)
| | - Hartmut Hengel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Hermann-Herder-Str.11, 79104 Freiburg, Germany; (Z.R.); (H.H.)
| | - Reingard Grabherr
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria;
| | - Siegfried Höfinger
- VSC Research Center, Technical University of Vienna, Operngasse 11/E057-09, 1040 Vienna, Austria; (E.S.); or (S.H.)
- Department of Physics, Michigan Technological University, Houghton, MI 49931, USA
| | - Hanna Harant
- Pivaris BioScience GmbH, Media Quarter Marx 3.4, Maria-Jacobi-Gasse 1, 1030 Vienna, Austria;
| |
Collapse
|
5
|
Porter JM, Oswald MS, Busuttil K, Emmanuel SN, Bennett A, McKenna R, Smith JG. Mechanisms of AAV2 neutralization by human alpha-defensins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614754. [PMID: 39386661 PMCID: PMC11463608 DOI: 10.1101/2024.09.25.614754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Antiviral immunity compromises the efficacy of adeno-associated virus (AAV) vectors used for gene therapy. This is well understood for the adaptive immune response. However, innate immune effectors like alpha-defensin antimicrobial peptides also block AAV infection, although their mechanisms of action are unknown. To address this gap in knowledge, we investigated AAV2 neutralization by human neutrophil peptide 1 (HNP1), a myeloid alpha-defensin, and human defensin 5 (HD5), an enteric alpha-defensin. We found that both defensins bind to AAV2 and inhibit infection at low micromolar concentrations. While HD5 prevents AAV2 from binding to cells, HNP1 does not. However, AAV2 exposed to HD5 after binding to cells is still neutralized, indicating an additional block to infection. Accordingly, both HD5 and HNP1 inhibit externalization of the VP1 unique domain, which contains a phospholipase A 2 enzyme required for endosome escape and nuclear localization signals required for nuclear entry. Consequently, both defensins prevent AAV2 from reaching the nucleus. Disruption of intracellular trafficking of the viral genome to the nucleus is reminiscent of how alpha-defensins neutralize other non-enveloped viruses, suggesting a common mechanism of inhibition. These results will inform the development of vectors capable of overcoming these hurdles to improve the efficiency of gene therapy. Author Summary AAVs are commonly used as gene therapy vectors due to their broad tropism and lack of disease association; however, host innate immune factors, such as human alpha-defensin antimicrobial peptides, can hinder gene delivery. Although it is becoming increasingly evident that human alpha-defensins can block infection by a wide range of nonenveloped viruses, including AAVs, their mechanism of action remains poorly understood. In this study, we describe for the first time how two types of abundant human alpha-defensins neutralize a specific AAV serotype, AAV2. We found that one defensin prevents AAV2 from binding to cells, the first step in infection, while both defensins block a critical later step in AAV2 entry. Our findings support the emerging idea that defensins use a common strategy to block infection by DNA viruses that replicate in the nucleus. Through understanding how innate immune effectors interact with and impede AAV infection, vectors can be developed to bypass these interventions and allow more efficient gene delivery.
Collapse
|
6
|
Li S, Mu R, Guo X. Defensins regulate cell cycle: Insights of defensins on cellular proliferation and division. Life Sci 2024; 349:122740. [PMID: 38777302 DOI: 10.1016/j.lfs.2024.122740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024]
Abstract
Defensins are a class of small antimicrobial peptides that play a crucial role against pathogens. However, recent research has highlighted defensins exhibit the ability to influence cell cycle checkpoints, promoting or inhibiting specific phases such as G1 arrest or S/M transition. By regulating the cell cycle, defensins impact the proliferation of normal and cancerous cells, with implications for cancer development and progression. Dysregulation of defensin expression can disrupt the delicate balance of cell cycle regulation, leading to uncontrolled cell growth and an increased risk of tumor formation. Defensins contribute to the resolution of inflammation, stimulate angiogenesis, and enhance the migration and proliferation of cells involved in tissue repair. Furthermore, The ability of defensins to respond to microenvironmental changes further demonstrates the significance of these peptides in host defense mechanisms and immune function. By adjusting their expression, defensins continue to combat pathogens effectively and maintain homeostasis within the body. This review highlights the multifaceted role of defensins in regulating the cell cycle and their broader implications in cancer progression, tissue repair, and microenvironmental response.
Collapse
Affiliation(s)
- Shuang Li
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China.
| | - Rongrong Mu
- Affiliated Hospital of Sichuan Nursing Vocational College, The Third People's Hospital of Sichuan Province, China
| | - Xueqin Guo
- Department of Pathology, Gaomi City People's Hospital, Gaomi 261500, China
| |
Collapse
|
7
|
Pace E, Di Vincenzo S, Ferraro M, Lanata L, Scaglione F. Role of airway epithelium in viral respiratory infections: Can carbocysteine prevent or mitigate them? Immunology 2024; 172:329-342. [PMID: 38354831 DOI: 10.1111/imm.13762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Alterations in airway epithelial homeostasis increase viral respiratory infections risk. Viral infections frequently are associated with chronic obstructive pulmonary disease (COPD) exacerbations, events that dramatically promote disease progression. Mechanism promoting the main respiratory viruses entry and virus-evocated innate and adaptive immune responses have now been elucidated, and an oxidative stress central role in these pathogenic processes has been recognized. Presence of reactive oxygen species in macrophages and other cells allows them to eliminate virus, but its excess alters the balance between innate and adaptive immune responses and proteases/anti-proteases and leads to uncontrolled inflammation, tissue damage, and hypercoagulability. Different upper and lower airway cell types also play a role in viral entry and infection. Carbocysteine is a muco-active drug with anti-oxidant and anti-inflammatory properties used for the management of several chronic respiratory diseases. Although the use of anti-oxidants has been proposed as an effective strategy in COPD exacerbations management, the molecular mechanisms that explain carbocysteine efficacy have not yet been fully clarified. The present review describes the most relevant features of the common respiratory virus pathophysiology with a focus on epithelial cells and oxidative stress role and reports data supporting a putative role of carbocysteine in viral respiratory infections.
Collapse
Affiliation(s)
- Elisabetta Pace
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Serena Di Vincenzo
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Maria Ferraro
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Palermo, Italy
| | | | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Yu C, Wu Q, Xin J, Yu Q, Ma Z, Xue M, Xu Q, Zheng C. Designing a smallpox B-cell and T-cell multi-epitope subunit vaccine using a comprehensive immunoinformatics approach. Microbiol Spectr 2024; 12:e0046524. [PMID: 38700327 PMCID: PMC11237557 DOI: 10.1128/spectrum.00465-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Smallpox is a highly contagious human disease caused by the variola virus. Although the disease was eliminated in 1979 due to its highly contagious nature and historical pathogenicity, with a mortality rate of up to 30%, this virus is an important candidate for biological weapons. Currently, vaccines are the critical measures to prevent this virus infection and spread. In this study, we designed a peptide vaccine using immunoinformatics tools, which have the potential to activate human immunity against variola virus infection efficiently. The design of peptides derives from vaccine-candidate proteins showing protective potential in vaccinia WR strains. Potential non-toxic and nonallergenic T-cell and B-cell binding and cytokine-inducing epitopes were then screened through a priority prediction using special linkers to connect B-cell epitopes and T-cell epitopes, and an appropriate adjuvant was added to the vaccine construction to enhance the immunogenicity of the peptide vaccine. The 3D structure display, docking, and free energy calculation analysis indicate that the binding affinity between the vaccine peptide and Toll-like receptor 3 is high, and the vaccine receptor complex is highly stable. Notably, the vaccine we designed is obtained from the protective protein of the vaccinia and combined with preventive measures to avoid side effects. This vaccine is highly likely to produce an effective and safe immune response against the variola virus infection in the body. IMPORTANCE In this work, we designed a vaccine with a cluster of multiple T-cell/B-cell epitopes, which should be effective in inducing systematic immune responses against variola virus infection. Besides, this work also provides a reference in vaccine design for preventing monkeypox virus infection, which is currently prevalent.
Collapse
Affiliation(s)
- Changqing Yu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Engineering Center of Agricultural Biosafety Assessment and Biotechnology, School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, China
| | - Qi Wu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jiuqing Xin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Qiujuan Yu
- Department of Dermatology, The First People's Hospital of Mudanjiang, Mudanjiang, China
| | - Zhixin Ma
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingyuan Xu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infection Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
9
|
Zhao C, Porter JM, Burke PC, Arnberg N, Smith JG. Alpha-defensin binding expands human adenovirus tropism. PLoS Pathog 2024; 20:e1012317. [PMID: 38900833 PMCID: PMC11230588 DOI: 10.1371/journal.ppat.1012317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/08/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Mammalian α-defensins are a family of abundant effector peptides of the mucosal innate immune system. Although primarily considered to be antimicrobial, α-defensins can increase rather than block infection by certain prominent bacterial and viral pathogens in cell culture and in vivo. We have shown previously that exposure of mouse and human adenoviruses (HAdVs) to α-defensins is able to overcome competitive inhibitors that block cell binding, leading us to hypothesize a defensin-mediated binding mechanism that is independent of known viral receptors. To test this hypothesis, we used genetic approaches to demonstrate that none of several primary receptors nor integrin co-receptors are needed for human α-defensin-mediated binding of HAdV to cells; however, infection remains integrin dependent. Thus, our studies have revealed a novel pathway for HAdV binding to cells that bypasses viral primary receptors. We speculate that this pathway functions in parallel with receptor-mediated entry and contributes to α-defensin-enhanced infection of susceptible cells. Remarkably, we also found that in the presence of α-defensins, HAdV tropism is expanded to non-susceptible cells, even when viruses are exposed to a mixture of both susceptible and non-susceptible cells. Therefore, we propose that in the presence of sufficient concentrations of α-defensins, such as in the lung or gut, integrin expression rather than primary receptor expression will dictate HAdV tropism in vivo. In summary, α-defensins may contribute to tissue tropism not only through the neutralization of susceptible viruses but also by allowing certain defensin-resistant viruses to bind to cells independently of previously described mechanisms.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jessica M. Porter
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Phillip C. Burke
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Niklas Arnberg
- Department of Clinical Microbiology, Division of Virology and Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Jason G. Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
10
|
Chen M, Hu Z, Shi J, Xie Z. Human β-defensins and their synthetic analogs: Natural defenders and prospective new drugs of oral health. Life Sci 2024; 346:122591. [PMID: 38548013 DOI: 10.1016/j.lfs.2024.122591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/08/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
As a family of cationic host defense peptides, human β-defensins (HBDs) are ubiquitous in the oral cavity and are mainly synthesized primarily by epithelial cells, serving as the primary barrier and aiming to prevent microbial invasion, inflammation, and disease while maintaining physiological homeostasis. In recent decades, there has been great interest in their biological functions, structure-activity relationships, mechanisms of action, and therapeutic potential in oral diseases. Meanwhile, researchers are dedicated to improving the properties of HBDs for clinical application. In this review, we first describe the classification, structural characteristics, functions, and mechanisms of HBDs. Next, we cover the role of HBDs and their synthetic analogs in oral diseases, including dental caries and pulp infections, periodontitis, peri-implantitis, fungal/viral infections and oral mucosal diseases, and oral squamous cell carcinoma. Finally, we discuss the limitations and challenges of clinical translation of HBDs and their synthetic analogs, including, but not limited to, stability, bioavailability, antimicrobial activity, resistance, and toxicity. Above all, this review summarizes the biological functions, mechanisms of action, and therapeutic potential of both natural HBDs and their synthetic analogs in oral diseases, as well as the challenges associated with clinical translation, thus providing substantial insights into the laboratory development and clinical application of HBDs in oral diseases.
Collapse
Affiliation(s)
- Mumian Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zihe Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Jue Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
11
|
Zhao C, Porter JM, Burke PC, Arnberg N, Smith JG. Alpha-defensin binding expands human adenovirus tropism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596681. [PMID: 38854108 PMCID: PMC11160700 DOI: 10.1101/2024.05.30.596681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Mammalian α-defensins are a family of abundant effector peptides of the mucosal innate immune system. Although primarily considered to be antimicrobial, α-defensins can increase rather than block infection by certain prominent bacterial and viral pathogens in cell culture and in vivo . We have shown previously that exposure of mouse and human adenoviruses (HAdVs) to α-defensins is able to overcome competitive inhibitors that block cell binding, leading us to hypothesize a defensin-mediated binding mechanism that is independent of known viral receptors. To test this hypothesis, we used genetic approaches to demonstrate that none of several primary receptors nor integrin co-receptors are needed for human α-defensin-mediated binding of HAdV to cells; however, infection remains integrin dependent. Thus, our studies have revealed a novel pathway for HAdV binding to cells that bypasses viral primary receptors. We speculate that this pathway functions in parallel with receptor-mediated entry and contributes to α-defensin-enhanced infection of susceptible cells. Remarkably, we also found that in the presence of α-defensins, HAdV tropism is expanded to non-susceptible cells, even when viruses are exposed to a mixture of both susceptible and non-susceptible cells. Therefore, we propose that in the presence of sufficient concentrations of α-defensins, such as in the lung or gut, integrin expression rather than primary receptor expression will dictate HAdV tropism in vivo . In summary, α-defensins may contribute to tissue tropism not only through the neutralization of susceptible viruses but also by allowing certain defensin-resistant viruses to bind to cells independently of previously described mechanisms. Author Summary In this study, we demonstrate a novel mechanism for binding of human adenoviruses (HAdVs) to cells that is dependent upon interactions with α-defensin host defense peptides but is independent of known viral receptors and co-receptors. To block normal receptor-mediated HAdV infection, we made genetic changes to both host cells and HAdVs. Under these conditions, α-defensins restored cell binding; however, infection still required the function of HAdV integrin co-receptors. This was true for multiple types of HAdVs that use different primary receptors and for cells that are either naturally devoid of HAdV receptors or were engineered to be receptor deficient. These observations suggest that in the presence of concentrations of α-defensins that would be found naturally in the lung or intestine, there are two parallel pathways for HAdV binding to cells that converge on integrins for productive infection. Moreover, these binding pathways function independently, and both operate in mixed culture. Thus, we have found that viruses can co-opt host defense molecules to expand their tropism.
Collapse
|
12
|
Schwartz J, Capistrano KJ, Gluck J, Hezarkhani A, Naqvi AR. SARS-CoV-2, periodontal pathogens, and host factors: The trinity of oral post-acute sequelae of COVID-19. Rev Med Virol 2024; 34:e2543. [PMID: 38782605 PMCID: PMC11260190 DOI: 10.1002/rmv.2543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/04/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
COVID-19 as a pan-epidemic is waning but there it is imperative to understand virus interaction with oral tissues and oral inflammatory diseases. We review periodontal disease (PD), a common inflammatory oral disease, as a driver of COVID-19 and oral post-acute-sequelae conditions (PASC). Oral PASC identifies with PD, loss of teeth, dysgeusia, xerostomia, sialolitis-sialolith, and mucositis. We contend that PD-associated oral microbial dysbiosis involving higher burden of periodontopathic bacteria provide an optimal microenvironment for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These pathogens interact with oral epithelial cells activate molecular or biochemical pathways that promote viral adherence, entry, and persistence in the oral cavity. A repertoire of diverse molecules identifies this relationship including lipids, carbohydrates and enzymes. The S protein of SARS-CoV-2 binds to the ACE2 receptor and is activated by protease activity of host furin or TRMPSS2 that cleave S protein subunits to promote viral entry. However, PD pathogens provide additional enzymatic assistance mimicking furin and augment SARS-CoV-2 adherence by inducing viral entry receptors ACE2/TRMPSS, which are poorly expressed on oral epithelial cells. We discuss the mechanisms involving periodontopathogens and host factors that facilitate SARS-CoV-2 infection and immune resistance resulting in incomplete clearance and risk for 'long-haul' oral health issues characterising PASC. Finally, we suggest potential diagnostic markers and treatment avenues to mitigate oral PASC.
Collapse
Affiliation(s)
- Joel Schwartz
- Department of Oral Medicine and Diagnostic Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | | | - Joseph Gluck
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Armita Hezarkhani
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Afsar R. Naqvi
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| |
Collapse
|
13
|
Chen G, Zheng Y, Wu N, Yang X, Qu S. Human beta defensin 3 knockdown inhibits the proliferation and migration of airway smooth muscle cells through regulating the PI3K/AKT signaling pathway. Mol Immunol 2024; 168:38-46. [PMID: 38422885 DOI: 10.1016/j.molimm.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/04/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Asthma, a common pediatric pulmonary disease, significantly affects children's healthy development. This study aimed to investigate the functions of human β defensin-3 (HBD-3) in asthma progression. For this purpose, blood samples from asthmatic and healthy children were collected. Moreover, the airway smooth muscle cells (ASMCs) were treated with platelet-derived growth factor BB (PDGF-BB) to develop an in vitro asthma model, then evaluated cell viability and migration via CCK-8 and transwell assays. The mRNA levels of interferon γ (INF-γ), interleukin 4 (IL-4), interleukin 10 (IL-10), alpha-smooth muscle actin (α-SMA), HBD-3, and the protein levels of phosphatidylinositol 3-kinase (PI3K) along with protein kinase B (AKT) were detected. Similarly, the N6-methyladenosine (m6A) content in the ASMCs and m6A levels of HBD-3 were also measured. Results indicated an upregulated HBD-3 in the asthmatic children. The ASMCs were found to be stimulated by PDGF-BB, in addition to the promotion of cell viability and migration. The INF-γ, IL-4, and α-SMA levels were reduced, while IL-10 was elevated in PDGF-BB-stimulated ASMCs. Silencing HBD-3 in PDGF-BB stimulated ASMCs was found to exert the opposite effect by inhibiting cell viability and migration, enhancing the levels of INF-γ, IL-4, and α-SMA, while the IL-10 levels were found to decline. PDGF-BB stimulation of ASMCs resulted in activation of the PI3K/AKT signaling pathway, which was blocked post HBD-3 silencing, while the role of si-hBD in PDGF-BB stimulated ASMCs was neutralized post-treatment with IGF-1. Finally, it was found that METTL3 overexpression prominently upregulated the m6A levels of HBD-3 and decreased the mRNA expression and stability of HBD-3 in the PDGF-BB-stimulated ASMCs. The study concluded that METTL3-mediated HBD-3 participates in the progression of asthma through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Guiying Chen
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, China; Department of Emergency, Sixth Affiliated Hospital of Harbin Medical University(Jiangnan Courtyard), China
| | - Yuling Zheng
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, China
| | - Nan Wu
- Department of Emergency, Sixth Affiliated Hospital of Harbin Medical University(Jiangnan Courtyard), China
| | - Xia Yang
- Department of Respiratory, Sixth Affiliated Hospital of Harbin Medical University (Jiangnan Courtyard), China
| | - Shuqiang Qu
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, China.
| |
Collapse
|
14
|
Han X, Song X, Xiao Z, Zhu G, Gao R, Ni B, Li J. Study on the mechanism of MDSC-platelets and their role in the breast cancer microenvironment. Front Cell Dev Biol 2024; 12:1310442. [PMID: 38404689 PMCID: PMC10884319 DOI: 10.3389/fcell.2024.1310442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key immunosuppressive cells in the tumor microenvironment (TME) that play critical roles in promoting tumor growth and metastasis. Tumor-associated platelets (TAPs) help cancer cells evade the immune system and promote metastasis. In this paper, we describe the interaction between MDSCs and TAPs, including their generation, secretion, activation, and recruitment, as well as the effects of MDSCs and platelets on the generation and changes in the immune, metabolic, and angiogenic breast cancer (BC) microenvironments. In addition, we summarize preclinical and clinical studies, traditional Chinese medicine (TCM) therapeutic approaches, and new technologies related to targeting and preventing MDSCs from interacting with TAPs to modulate the BC TME, discuss the potential mechanisms, and provide perspectives for future development. The therapeutic strategies discussed in this review may have implications in promoting the normalization of the BC TME, reducing primary tumor growth and distant lung metastasis, and improving the efficiency of anti-tumor therapy, thereby improving the overall survival (OS) and progression-free survival (PFS) of patients. However, despite the significant advances in understanding these mechanisms and therapeutic strategies, the complexity and heterogeneity of MDSCs and side effects of antiplatelet agents remain challenging. This requires further investigation in future prospective cohort studies.
Collapse
Affiliation(s)
- Xinpu Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Hematology-Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotong Song
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhigang Xiao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Ni
- Department of Oncology, First Hospital of Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Gao X, Feng J, Wei L, Dong P, Chen J, Zhang L, Yang Y, Xu L, Wang H, Luo J, Qin M. Defensins: A novel weapon against Mycobacterium tuberculosis? Int Immunopharmacol 2024; 127:111383. [PMID: 38118315 DOI: 10.1016/j.intimp.2023.111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 12/22/2023]
Abstract
Tuberculosis (TB) is a serious airborne communicable disease caused by organisms of the Mycobacterium tuberculosis (Mtb) complex. Although the standard treatment antimicrobials, including isoniazid, rifampicin, pyrazinamide, and ethambutol, have made great progress in the treatment of TB, problems including the rising incidence of multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant tuberculosis (XDR-TB), the severe toxicity and side effects of antimicrobials, and the low immunity of TB patients have become the bottlenecks of the current TB treatments. Therefore, both safe and effective new strategies to prevent and treat TB have become a top priority. As a subfamily of cationic antimicrobial peptides, defensins are rich in cysteine and play a vital role in resisting the invasion of microorganisms and regulating the immune response. Inspired by studies on the roles of defensins in host defence, we describe their research history and then review their structural features and antimicrobial mechanisms, specifically for fighting Mtb in detail. Finally, we discuss the clinical relevance, therapeutic potential, and potential challenges of defensins in anti-TB therapy. We further debate the possible solutions of the current application of defensins to provide new insights for eliminating Mtb.
Collapse
Affiliation(s)
- Xuehan Gao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jihong Feng
- Department of Oncology, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China
| | - Linna Wei
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Pinzhi Dong
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jin Chen
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Langlang Zhang
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yuhan Yang
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Lin Xu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Haiyan Wang
- Department of Epidemiology and Health Statistics, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Junmin Luo
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Ming Qin
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Special Key Laboratory of Gene Detection & Therapy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
16
|
Haratian K, Borjian Boroujeni P, Sabbaghian M, Maghareh Abed E, Moazenchi M, Mohseni Meybodi A. DEFB126 2-nt Deletion (rs11467417) as a Potential Risk Factor for Chlamydia Trachomatis Infection and Subsequent Infertility in Iranian Men. J Reprod Infertil 2024; 25:20-27. [PMID: 39157277 PMCID: PMC11330205 DOI: 10.18502/jri.v25i1.15195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/27/2024] [Indexed: 08/20/2024] Open
Abstract
Background Chlamydia trachomatis (CT) is one of the most prevalent sexually transmitted infections, causing genital tract infections and infertility. Defensins have an immunomodulatory function and play an important role in sperm maturation, motility, and fertilization. DEFB126 is present on ejaculated spermatozoa and is essential for them to pass through the female reproductive tract. The purpose of the study was to determine the frequency of the 2-nt deletion of the DEFB126 (rs11467417) in Iranian infertile males with a recurrent history of CT. Methods Semen samples of 1080 subfertile males were investigated. Among patients who had CT-positive results, sperm DNA from 50 symptomatic and 50 asymptomatic patients were collected for the DEFB126 genotype analysis. Additionally, a control group comprising 100 DNA samples from individuals with normal spermogram and testing negative for CT was included in the study. The PCR-sequencing for detecting the 2-nt deletion of the second exon of the DEFB126 was performed. Results The Chi-squared test comparing all three groups revealed no significant difference across the different genotypes. Moreover, no significant difference between the symptomatic and asymptomatic groups was seen. However, analysis within CT-positive patients and controls demonstrated significant difference between the frequencies of homozygous del/del. Conclusion The higher frequency of the 2-nt deletion of the DEFB126 in CT- positive patients suggests that the occurrence of mutations in the DEFB-126 may cause the impairment of the antimicrobial activity of the DEFB126 protein and consequently makes individuals more susceptible to infections such as CT.
Collapse
Affiliation(s)
- Kaveh Haratian
- Department of Pathology and Laboratory Medicine, Western University, Ontario, Canada
| | - Parnaz Borjian Boroujeni
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Marjan Sabbaghian
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Elham Maghareh Abed
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maedeh Moazenchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Anahita Mohseni Meybodi
- Department of Pathology and Laboratory Medicine, Western University, Ontario, Canada
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
17
|
Santacroce L, Magrone T. Molluscum Contagiosum Virus: Biology and Immune Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:151-170. [PMID: 38801577 DOI: 10.1007/978-3-031-57165-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Molluscum contagiosum virus is a poxvirus belonging to the Poxviridae family, which includes Orthopoxvirus, Parapoxvirus, Yantapoxvirus, Molluscipoxvirus, Smallpox virus, Cowpox virus and Monkeypox virus. MCV belongs to the genus Molluscipoxvirus and has a tropism for skin tissue. MCV infects keratinocytes and, after an incubation period of 2 weeks to 6 weeks, causes a breakdown of the skin barrier with the development of papules of variable size depending on the proper functioning of the immune response (both adaptive and acquired). MCV only infects humans and does not cause viraemia. MCV encodes for several inhibitory proteins responsible to circumvent the immune response through different signalling pathways. Individuals who can be infected with MCV are children, immunocompromised individuals such as organ transplant recipients and Human Immunodeficiency Virus (HIV)-infected individuals. Current treatments to manage MCV-induced lesions are different and include the use of immunomodulators, which, however, do not provide an effective response.
Collapse
Affiliation(s)
- Luigi Santacroce
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari, Bari, Italy.
| | - Thea Magrone
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari, Bari, Italy
| |
Collapse
|
18
|
Hackert NS, Radtke FA, Exner T, Lorenz HM, Müller-Tidow C, Nigrovic PA, Wabnitz G, Grieshaber-Bouyer R. Human and mouse neutrophils share core transcriptional programs in both homeostatic and inflamed contexts. Nat Commun 2023; 14:8133. [PMID: 38065997 PMCID: PMC10709367 DOI: 10.1038/s41467-023-43573-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Neutrophils are frequently studied in mouse models, but the extent to which findings translate to humans remains poorly defined. In an integrative analysis of 11 mouse and 13 human datasets, we find a strong correlation of neutrophil gene expression across species. In inflammation, neutrophils display substantial transcriptional diversity but share a core inflammation program. This program includes genes encoding IL-1 family members, CD14, IL-4R, CD69, and PD-L1. Chromatin accessibility of core inflammation genes increases in blood compared to bone marrow and further in tissue. Transcription factor enrichment analysis implicates members of the NF-κB family and AP-1 complex as important drivers, and HoxB8 neutrophils with JunB knockout show a reduced expression of core inflammation genes in resting and activated cells. In independent single-cell validation data, neutrophil activation by type I or type II interferon, G-CSF, and E. coli leads to upregulation in core inflammation genes. In COVID-19 patients, higher expression of core inflammation genes in neutrophils is associated with more severe disease. In vitro treatment with GM-CSF, LPS, and type II interferon induces surface protein upregulation of core inflammation members. Together, we demonstrate transcriptional conservation in neutrophils in homeostasis and identify a core inflammation program shared across heterogeneous inflammatory conditions.
Collapse
Affiliation(s)
- Nicolaj S Hackert
- Division of Rheumatology, Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Felix A Radtke
- Division of Rheumatology, Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Tarik Exner
- Division of Rheumatology, Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), University of Heidelberg, Heidelberg, Germany
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Guido Wabnitz
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ricardo Grieshaber-Bouyer
- Division of Rheumatology, Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany.
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), University of Heidelberg, Heidelberg, Germany.
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich Alexander Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
19
|
Hu CT, Diaz K, Yang LC, Sharma A, Greenberg HB, Smith JG. Corrected and republished from: "VP4 Is a Determinant of Alpha-Defensin Modulation of Rotaviral Infection". J Virol 2023; 97:e0096223. [PMID: 37787534 PMCID: PMC10617384 DOI: 10.1128/jvi.00962-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 10/04/2023] Open
Abstract
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 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, Stanford School of Medicine, Stanford, California, USA
- 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
|
20
|
Urbańska DM, Pawlik M, Korwin-Kossakowska A, Czopowicz M, Rutkowska K, Kawecka-Grochocka E, Mickiewicz M, Kaba J, Bagnicka E. Effect of Supplementation with Curcuma longa and Rosmarinus officinalis Extract Mixture on Acute Phase Protein, Cathelicidin, Defensin and Cytolytic Protein Gene Expression in the Livers of Young Castrated Polish White Improved Bucks. Genes (Basel) 2023; 14:1932. [PMID: 37895281 PMCID: PMC10606746 DOI: 10.3390/genes14101932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Goats are an excellent animal model for research on some physiological and pathophysiological processes in humans. The search for supplements that prevent homeostasis disorders and strengthen the immune system is necessary to reduce the risk of many diseases in both humans and animals. The aim of the study was to analyze the effect of supplementation with a mixture of dried extracts of Curcuma longa and Rosmarinus officinalis on the expression of acute-phase protein (SAA, HP, CRP, LALBA, AGP, CP, FGA, FGB, and FGG), cathelicidin (BAC5, BAC7.5, BAC3.4, MAP28, MAP34, and HEPC), beta-defensin-1 (GBD1, DEFB1), and beta-defensin-2, and cytolytic protein (LIZ and LF) genes in the livers of young castrated bucks of the Polish White Improved breed. The higher expression of LF in the control group suggests that it is important for the first line of hepatic immune defense and its expression is downregulated by the mixture of turmeric and rosemary extracts; thus, the spice-herb mixture mutes its activity. The lower expression of FGB and the higher expression of BAC5 genes in the livers of healthy, young castrated bucks who were administered the supplement suggest the silencing effects of the mixture on the acute-phase response and the stimulating effect on the antimicrobial activity of the immune system.
Collapse
Affiliation(s)
- Daria M. Urbańska
- Department of Animal Improvement and Nutrigenomics, Institute of Genetics and Animal Breeding Polish Academy of Sciences, Postepu 36A Str., 05-552 Jastrzębiec, Poland; (A.K.-K.); (E.B.)
| | - Marek Pawlik
- Department of Neurotoxicology, Mossakowski Medical Research Institute Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
| | - Agnieszka Korwin-Kossakowska
- Department of Animal Improvement and Nutrigenomics, Institute of Genetics and Animal Breeding Polish Academy of Sciences, Postepu 36A Str., 05-552 Jastrzębiec, Poland; (A.K.-K.); (E.B.)
| | - Michał Czopowicz
- Division of Veterinary Epidemiology and Economics, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska Str. 159c, 02-776 Warsaw, Poland; (M.C.); (M.M.); (J.K.)
| | - Karolina Rutkowska
- Department of Medical Genetics, Medical University of Warsaw, Pawińskiego 3c Str., 02-106 Warsaw, Poland;
| | - Ewelina Kawecka-Grochocka
- Department of Animal Improvement and Nutrigenomics, Institute of Genetics and Animal Breeding Polish Academy of Sciences, Postepu 36A Str., 05-552 Jastrzębiec, Poland; (A.K.-K.); (E.B.)
| | - Marcin Mickiewicz
- Division of Veterinary Epidemiology and Economics, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska Str. 159c, 02-776 Warsaw, Poland; (M.C.); (M.M.); (J.K.)
| | - Jarosław Kaba
- Division of Veterinary Epidemiology and Economics, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska Str. 159c, 02-776 Warsaw, Poland; (M.C.); (M.M.); (J.K.)
| | - Emilia Bagnicka
- Department of Animal Improvement and Nutrigenomics, Institute of Genetics and Animal Breeding Polish Academy of Sciences, Postepu 36A Str., 05-552 Jastrzębiec, Poland; (A.K.-K.); (E.B.)
| |
Collapse
|
21
|
Srivastava S, Kolbe M. Novel "GaEl Antigenic Patches" Identified by a "Reverse Epitomics" Approach to Design Multipatch Vaccines against NIPAH Infection, a Silent Threat to Global Human Health. ACS OMEGA 2023; 8:31698-31713. [PMID: 37692250 PMCID: PMC10483669 DOI: 10.1021/acsomega.3c01909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/01/2023] [Indexed: 09/12/2023]
Abstract
Nipah virus (NiV) is a zoonotic virus that causes lethal encephalitis and respiratory disease with the symptom of endothelial cell-cell fusion. Several NiV outbreaks have been reported since 1999 with nearly annual occurrences in Bangladesh. The outbreaks had high mortality rates ranging from 40 to 90%. No specific vaccine has yet been reported against NiV. Recently, several vaccine candidates and different designs of vaccines composed of epitopes against NiV were proposed. Most of the vaccines target single protein or protein complex subunits of the pathogen. The multiepitope vaccines proposed also cover a largely limited number of epitopes, and hence, their efficiency is still uncertain. To address the urgent need for a specific and effective vaccine against NiV infection, in the present study, we have utilized the "reverse epitomics" approach ("overlapping-epitope-clusters-to-patches" method) to identify "antigenic patches" (Ag-Patches) and utilize them as immunogenic composition for multipatch vaccine (MPV) design. The designed MPVs were analyzed for immunologically crucial parameters, physiochemical properties, and interaction with Toll-like receptor 3 ectodomain. In total, 30 CTL (cytotoxic T lymphocyte) and 27 HTL (helper T lymphocyte) antigenic patches were identified from the entire NiV proteome based on the clusters of overlapping epitopes. These identified Ag-Patches cover a total of discrete 362 CTL and 414 HTL epitopes from the entire proteome of NiV. The antigenic patches were utilized as immunogenic composition for the design of two CTL and two HTL multipatch vaccines. The 57 antigenic patches utilized here cover 776 overlapping epitopes targeting 52 different HLA class I and II alleles, providing a global ethnically distributed human population coverage of 99.71%. Such large number of epitope coverage resulting in large human population coverage cannot be reached with single-protein/subunit or multiepitope based vaccines. The reported antigenic patches also provide potential immunogenic composition for early detection diagnostic kits for NiV infection. Further, all the MPVs and Toll-like receptor ectodomain complexes show a stable nature of molecular interaction with numerous hydrogen bonds, salt bridges, and nonbounded contact formation and acceptable root mean square deviation and fluctuation. The cDNA analysis shows a favorable large-scale expression of the MPV constructs in a human cell line. By utilizing the novel "reverse epitomics" approach, highly immunogenic novel "GaEl antigenic patches" (GaEl Ag-Patches), a synonym term for "antigenic patches", were identified and utilized as immunogenic composition to design four MPVs against NiV. We conclude that the novel multipatch vaccines are potential candidates to combat NiV, with greater effectiveness, high specificity, and large human population coverage worldwide.
Collapse
Affiliation(s)
- Sukrit Srivastava
- Infection
Biology Group, Indian Foundation for Fundamental
Research Trust, Raebareli, Uttar Pradesh 229316, India
- Department
for Structural Infection Biology, Centre
for Structural Systems Biology (CSSB) & Helmholtz-Centre for Infection
Research, Notkestraße 85, 22607 Hamburg, Germany
| | - Michael Kolbe
- Department
for Structural Infection Biology, Centre
for Structural Systems Biology (CSSB) & Helmholtz-Centre for Infection
Research, Notkestraße 85, 22607 Hamburg, Germany
- Faculty
of Mathematics, Informatics and Natural Sciences, University of Hamburg, Rothenbaumchaussee 19, 20148 Hamburg, Germany
| |
Collapse
|
22
|
Govender Y, Morrison CS, Chen PL, Gao X, Yamamoto H, Chipato T, Anderson S, Barbieri R, Salata R, Doncel GF, Fichorova RN. Cervical and systemic innate immunity predictors of HIV risk linked to genital herpes acquisition and time from HSV-2 seroconversion. Sex Transm Infect 2023; 99:311-316. [PMID: 36104248 PMCID: PMC10011014 DOI: 10.1136/sextrans-2022-055458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/25/2022] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES To examine innate immunity predictors of HIV-1 acquisition as biomarkers of HSV-2 risk and biological basis for epidemiologically established HIV-1 predisposition in HSV-2 infected women. METHODS We analysed longitudinal samples from HIV-1 negative visits of 1019 women before and after HSV-2 acquisition. We measured cervical and serum biomarkers of inflammation and immune activation previously linked to HIV-1 risk. Protein levels were Box-Cox transformed and ORs for HSV-2 acquisition were calculated based on top quartile or below/above median levels for all HSV-2 negative visits. Bivariate analysis determined the likelihood of HSV-2 acquisition by biomarker levels preceding infection. Linear mixed-effects models evaluated if biomarkers differed by HSV-2 status defined as negative, incident or established infections with an established infection cut-off starting at 6 months. RESULTS In the cervical compartment, two biomarkers of HIV-1 risk (low SLPI and high BD-2) also predicted HSV-2 acquisition. In addition, HSV-2 acquisition was associated with IL-1β, IL-6, IL-8, MIP-3α, ICAM-1 and VEGF when below median levels. Systemic immunity predictors of HSV-2 acquisition were high sCD14 and IL-6, with highest odds when concomitantly increased (OR=2.23, 1.49-3.35). Concomitant systemic and mucosal predictors of HSV-2 acquisition risk included (1) serum top quartile sCD14 with cervical low SLPI, VEGF and ICAM-1, or high BD-2; (2) serum high IL-6 with cervical low VEGF and ICAM-1, SLPI, IL-1β and IL-6; and (3) serum low C reactive protein with cervical high BD-2 (the only combination also predictive of HIV-1 acquisition). Most cervical biomarkers were decreased after HSV-2 acquisition compared with the HSV-2 negative visits, with incident infections associated with a larger number of suppressed cervical biomarkers and lower serum IL-6 levels compared with established infections. CONCLUSIONS A combination of systemic immunoinflammatory and cervical immunosuppressed states predicts HSV-2 acquisition. A persistently suppressed innate immunity during incident HSV-2 infection may add to the increased HIV-1 susceptibility.
Collapse
Affiliation(s)
- Yashini Govender
- Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Charles S Morrison
- Global Health and Population Research, FHI 360, Durham, North Carolina, USA
| | - Pai-Lien Chen
- Global Health and Population Research, FHI 360, Durham, North Carolina, USA
| | - Xiaoming Gao
- Global Health and Population Research, FHI 360, Durham, North Carolina, USA
| | - Hidemi Yamamoto
- Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tsungai Chipato
- Obstetrics and Gynecology, University of Zimbabwe, Harare, Zimbabwe
| | - Sharon Anderson
- Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Robert Barbieri
- Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Salata
- Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gustavo F Doncel
- Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, USA
- CONRAD, Arlington, Virginia, USA
| | - Raina Nakova Fichorova
- Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Rizzi A, Di Gioacchino M, Gammeri L, Inchingolo R, Chini R, Santilli F, Nucera E, Gangemi S. The Emerging Role of Innate Lymphoid Cells (ILCs) and Alarmins in Celiac Disease: An Update on Pathophysiological Insights, Potential Use as Disease Biomarkers, and Therapeutic Implications. Cells 2023; 12:1910. [PMID: 37508573 PMCID: PMC10378400 DOI: 10.3390/cells12141910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Celiac disease (CD) is an intestinal disease that develops in genetically predisposed individuals and is triggered by the ingestion of gluten. CD was considered a Th1-disease. Today, the role of Th17, IL-21, and IL-17A lymphocytes is well known. Inflammation is regulated by the activity of gluten-specific CD4+ T lymphocytes that produce pro-inflammatory cytokines, including IFN-γ, TNF-α, and IL-21, perpetuating the Th1 response. These cytokines determine an inflammatory state of the small intestine, with consequent epithelial infiltration of lymphocytes and an alteration of the architecture of the duodenal mucosa. B cells produce antibodies against tissue transglutaminase and against deamidated gliadin. Although the role of the adaptive immune response is currently known, the evidence about the role of innate immunity cells is still poorly understood. Epithelial damage determines the release of damage-associated molecular patterns (DAMPs), also known as alarmins. Together with the intestinal epithelial cells and the type 1 innate lymphoid cells (ILC1s), alarmins like TSLP, IL-33, and HMGB1 could have a fundamental role in the genesis and maintenance of inflammation. Our study aims to evaluate the evidence in the literature about the role of ILCs and alarmins in celiac disease, evaluating the possible future diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Angela Rizzi
- UOSD Allergologia e Immunologia Clinica, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Mario Di Gioacchino
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University, 66100 Chieti, Italy
| | - Luca Gammeri
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| | - Riccardo Inchingolo
- Pulmonary Medicine Unit, Department of Neurosciences, Sense Organs and Thorax, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Raffaella Chini
- UOSD Allergologia e Immunologia Clinica, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesca Santilli
- Center for Advanced Studies and Technology, G. d'Annunzio University, 66100 Chieti, Italy
| | - Eleonora Nucera
- UOSD Allergologia e Immunologia Clinica, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
24
|
Hardy E, Sarker H, Fernandez-Patron C. Could a Non-Cellular Molecular Interactome in the Blood Circulation Influence Pathogens' Infectivity? Cells 2023; 12:1699. [PMID: 37443732 PMCID: PMC10341357 DOI: 10.3390/cells12131699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
We advance the notion that much like artificial nanoparticles, relatively more complex biological entities with nanometric dimensions such as pathogens (viruses, bacteria, and other microorganisms) may also acquire a biomolecular corona upon entering the blood circulation of an organism. We view this biomolecular corona as a component of a much broader non-cellular blood interactome that can be highly specific to the organism, akin to components of the innate immune response to an invading pathogen. We review published supporting data and generalize these notions from artificial nanoparticles to viruses and bacteria. Characterization of the non-cellular blood interactome of an organism may help explain apparent differences in the susceptibility to pathogens among individuals. The non-cellular blood interactome is a candidate therapeutic target to treat infectious and non-infectious conditions.
Collapse
Affiliation(s)
- Eugenio Hardy
- Center of Molecular Immunology, P.O. Box 16040, Havana 11600, Cuba
| | - Hassan Sarker
- Department of Biochemistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2H7, Canada;
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
25
|
Lyu Z, Yang P, Lei J, Zhao J. Biological Function of Antimicrobial Peptides on Suppressing Pathogens and Improving Host Immunity. Antibiotics (Basel) 2023; 12:1037. [PMID: 37370356 DOI: 10.3390/antibiotics12061037] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The emergence of drug-resistant genes and concerns about food safety caused by the overuse of antibiotics are becoming increasingly prominent. There is an urgent need for effective alternatives to antibiotics in the fields of livestock production and human medicine. Antimicrobial peptides can effectively replace antibiotics to kill pathogens and enhance the immune functions of the host, and pathogens cannot easily produce genes that are resistant to them. The ability of antimicrobial peptides (AMPs) to kill pathogens is associated with their structure and physicochemical properties, such as their conformation, electrical charges, hydrophilicity, and hydrophobicity. AMPs regulate the activity of immunological cells and stimulate the secretion of inflammatory cytokines via the activation of the NF-κB and MAPK signaling pathways. However, there are still some limitations to the application of AMPs in the fields of livestock production and human medicine, including a restricted source base, high costs of purification and expression, and the instability of the intestines of animals and humans. This review summarizes the information on AMPs as effective antibiotic substitutes to improve the immunological functions of the host through suppressing pathogens and regulating inflammatory responses. Potential challenges for the commercial application of AMPs in animal husbandry and human medicine are discussed.
Collapse
Affiliation(s)
- Zhiqian Lyu
- Guangdong Haid Group Co., Ltd., Guangzhou 511400, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Pan Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jian Lei
- Guangdong Haid Group Co., Ltd., Guangzhou 511400, China
- Qingyuan Haibei BIO-TECH Co., Ltd., Qingyuan 511853, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
26
|
Qi X, Feng T, Ma Z, Zheng L, Liu H, Shi Z, Shen C, Li P, Wu P, Ru Y, Li D, Zhu Z, Tian H, Wu S, Zheng H. Deletion of DP148R, DP71L, and DP96R Attenuates African Swine Fever Virus, and the Mutant Strain Confers Complete Protection against Homologous Challenges in Pigs. J Virol 2023; 97:e0024723. [PMID: 37017515 PMCID: PMC10134827 DOI: 10.1128/jvi.00247-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/10/2023] [Indexed: 04/06/2023] Open
Abstract
The African swine fever virus (ASFV) has caused a devastating pandemic in domestic and wild swine, causing economic losses to the global swine industry. Recombinant live attenuated vaccines are an attractive option for ASFV treatment. However, safe and effective vaccines against ASFV are still scarce, and more high-quality experimental vaccine strains need to be developed. In this study, we revealed that deletion of the ASFV genes DP148R, DP71L, and DP96R from the highly virulent isolate ASFV CN/GS/2018 (ASFV-GS) substantially attenuated virulence in swine. Pigs infected with 104 50% hemadsorbing doses of the virus with these gene deletions remained healthy during the 19-day observation period. No ASFV infection was detected in contact pigs under the experimental conditions. Importantly, the inoculated pigs were protected against homologous challenges. Additionally, RNA sequence analysis showed that deletion of these viral genes induced significant upregulation of the host histone H3.1 gene (H3.1) and downregulation of the ASFV MGF110-7L gene. Knocking down the expression of H3.1 resulted in high levels of ASFV replication in primary porcine macrophages in vitro. These findings indicate that the deletion mutant virus ASFV-GS-Δ18R/NL/UK is a novel potential live attenuated vaccine candidate and one of the few experimental vaccine strains reported to induce full protection against the highly virulent ASFV-GS virus strain. IMPORTANCE Ongoing outbreaks of African swine fever (ASF) have considerably damaged the pig industry in affected countries. Thus, a safe and effective vaccine is important to control African swine fever spread. Here, an ASFV strain with three gene deletions was developed by knocking out the viral genes DP148R (MGF360-18R), NL (DP71L), and UK (DP96R). The results showed that the recombinant virus was completely attenuated in pigs and provided strong protection against parental virus challenge. Additionally, no viral genomes were detected in the sera of pigs housed with animals infected with the deletion mutant. Furthermore, transcriptome sequencing (RNA-seq) analysis revealed significant upregulation of histone H3.1 in virus-infected macrophage cultures and downregulation of the ASFV MGF110-7L gene after viral DP148R, UK, and NL deletion. Our study provides a valuable live attenuated vaccine candidate and potential gene targets for developing strategies for anti-ASFV treatment.
Collapse
Affiliation(s)
- Xiaolan Qi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Tao Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhao Ma
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Linlin Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Huanan Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhengwang Shi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chaochao Shen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pan Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Panxue Wu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yi Ru
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Dan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zixiang Zhu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hong Tian
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Sen Wu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
27
|
Hrynkiewicz R, Niedźwiedzka-Rystwej P. Etiology of viral induced acute liver failure and defensins as potential therapeutic agents in ALF treatment. Front Immunol 2023; 14:1153528. [PMID: 37153560 PMCID: PMC10160486 DOI: 10.3389/fimmu.2023.1153528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
Acute liver failure (ALF) is a rare and severe disease, which, despite continuous advances in medicine, is still characterized by high mortality (65-85%). Very often, a liver transplant is the only effective treatment for ALF. Despite the implementation of prophylactic vaccinations in the world, the viral background of ALF is still a problem and leads to many deaths. Depending on the cause of ALF, it is sometimes possible to reverse this condition with appropriate therapies, which is why the search for effective antiviral agents seems to be a very desirable direction of research. Defensins, which are our natural antimicrobial peptides, have a very high potential to be used as therapeutic agents for infectious liver diseases. Previous studies on the expression of human defensins have shown that increased expression of human α and β-defensins in HCV and HBV infections is associated with a better response to treatment. Unfortunately, conducting clinical trials for ALF is very difficult due to the severity of the disease and the low incidence, therefore animal models are important for the development of new therapeutic strategies. One of the best animal models that has real reference to research on acute liver failure (ALF) is rabbit hemorrhagic disease in rabbits caused by the Lagovirus europaeus virus. So far, there have been no studies on the potential of defensins in rabbits infected with Lagovirus europaeus virus.
Collapse
|
28
|
Li K, Wu Q, Li H, Sun H, Xing Z, Li L, Chen H. Multiomic characterisation of the long-term sequelae of SARS survivors: a clinical observational study. EClinicalMedicine 2023; 58:101884. [PMID: 36873427 PMCID: PMC9969173 DOI: 10.1016/j.eclinm.2023.101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND We aimed to characterise the long-term health outcomes of survivors of severe acute respiratory syndrome (SARS) and determine their recovery status and possible immunological basis. METHODS We performed a clinical observational study on 14 health workers who survived SARS coronavirus infection between Apr 20, 2003 and Jun 6, 2003 in Haihe Hospital (Tianjin, China). Eighteen years after discharge, SARS survivors were interviewed using questionnaires on symptoms and quality of life, and received physical examination, laboratory tests, pulmonary function tests, arterial blood gas analysis, and chest imaging. Plasma samples were collected for metabolomic, proteomic, and single-cell transcriptomic analyses. The health outcomes were compared 18 and 12 years after discharge. Control individuals were also health workers from the same hospital but did not infect with SARS coronavirus. FINDINGS Fatigue was the most common symptom in SARS survivors 18 years after discharge, with osteoporosis and necrosis of the femoral head being the main sequelae. The respiratory function and hip function scores of the SARS survivors were significantly lower than those of the controls. Physical and social functioning at 18 years was improved compared to that after 12 years but still worse than the controls. Emotional and mental health were fully recovered. Lung lesions on CT scans remained consistent at 18 years, especially in the right upper lobe and left lower lobe lesions. Plasma multiomics analysis indicated an abnormal metabolism of amino acids and lipids, promoted host defense immune responses to bacteria and external stimuli, B-cell activation, and enhanced cytotoxicity of CD8+ T cells but impaired antigen presentation capacity of CD4+ T cells. INTERPRETATION Although health outcomes continued to improve, our study suggested that SARS survivors still suffered from physical fatigue, osteoporosis, and necrosis of the femoral head 18 years after discharge, possibly related to plasma metabolic disorders and immunological alterations. FUNDING This study was funded by the Tianjin Haihe Hospital Science and Technology Fund (HHYY-202012) and Tianjin Key Medical Discipline (Specialty) Construction Project (TJYXZDXK-063B, TJYXZDXK-067C).
Collapse
Affiliation(s)
- Kuan Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, China
| | - Qian Wu
- Department of Respiratory Medicine, Haihe Hospital, Tianjin Medical University, Tianjin, China
| | - Hongjie Li
- Department of Clinical Laboratory, Haihe Hospital, Tianjin University, Tianjin, China
| | - Haibai Sun
- Department of Clinical Laboratory, Haihe Hospital, Tianjin University, Tianjin, China
- Corresponding author.
| | - Zhiheng Xing
- Department of Radiology, Haihe Hospital, Tianjin University, Tianjin, China
- Corresponding author.
| | - Li Li
- Department of Respiratory Medicine, Haihe Hospital, Tianjin Medical University, Tianjin, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
- Corresponding author.
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
- Corresponding author. Haihe Hospital, Tianjin University, Tianjin 300350, PR China.
| |
Collapse
|
29
|
Porter JM, Oswald MS, Sharma A, Emmanuel S, Kansol A, Bennett A, McKenna R, Smith JG. A Single Surface-Exposed Amino Acid Determines Differential Neutralization of AAV1 and AAV6 by Human Alpha-Defensins. J Virol 2023; 97:e0006023. [PMID: 36916912 PMCID: PMC10062168 DOI: 10.1128/jvi.00060-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Adeno-associated viruses (AAVs) are being developed as gene therapy vectors due to their low pathogenicity and tissue tropism properties. However, the efficacy of these vectors is impeded by interactions with the host immune system. One potential immune barrier to vector transduction is innate immune host defense peptides, such as alpha-defensins, which are potent antiviral agents against other nonenveloped viruses. To investigate the interaction between AAVs and alpha-defensins, we utilized two closely related AAV serotypes, AAV1 and AAV6. Although their capsids differ by only six residues, these two serotypes exhibit markedly different tissue tropisms and transduction efficiencies. Using two abundant human alpha-defensins, enteric human defensin 5 (HD5) and myeloid human neutrophil peptide 1 (HNP1), we found both serotype-specific and defensin-specific effects on AAV infection. AAV6 infection was uniformly neutralized by both defensins at low micromolar concentrations; however, inhibition of AAV1 infection was profoundly influenced by the timing of defensin exposure to the virus relative to viral attachment to the cell. Remarkably, these differences in the defensin-dependent infection phenotype between the viruses are completely dictated by the identity of a single, surface-exposed amino acid (position 531) that varies between the two serotypes. These findings reveal a determinant for defensin activity against a virus with unprecedented precision. Furthermore, they provide a rationale for the investigation of other AAV serotypes not only to understand the mechanism of neutralization of defensins against AAVs but also to design more efficient vectors. IMPORTANCE The ability of adeno-associated viruses (AAVs) to infect and deliver genetic material to a range of cell types makes them favorable gene therapy vectors. However, AAV vectors encounter a wide variety of host immune factors throughout the body, which can impede efficient gene delivery. One such group of factors is the alpha-defensins, which are a key component of the innate immune system that can directly block viral infection. By studying the impact that alpha-defensins have on AAV infection, we found that two similar AAV serotypes (AAV1 and AAV6) have different sensitivities to inhibition. We also identified a single amino acid (position 531) that differs between the two AAV serotypes and is responsible for mediating their defensin sensitivity. By investigating the effects that host immune factors have on AAV infection, more efficient vectors may be developed to evade intervention by the immune system prior to gene delivery.
Collapse
Affiliation(s)
- Jessica M. Porter
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Mackenzi S. Oswald
- 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
| | - Shanan Emmanuel
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Austin Kansol
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Antonette Bennett
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Jason G. Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
30
|
Srivastava S, Verma S, Kamthania M, Saxena AK, Pandey KC, Pande V, Kolbe M. Exploring the structural basis to develop efficient multi-epitope vaccines displaying interaction with HLA and TAP and TLR3 molecules to prevent NIPAH infection, a global threat to human health. PLoS One 2023; 18:e0282580. [PMID: 36920996 PMCID: PMC10016716 DOI: 10.1371/journal.pone.0282580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Nipah virus (NiV) is an emerging zoonotic virus that caused several serious outbreaks in the south asian region with high mortality rates ranging from 40 to 90% since 2001. NiV infection causes lethal encephalitis and respiratory disease with the symptom of endothelial cell-cell fusion. No specific and effective vaccine has yet been reported against NiV. To address the urgent need for a specific and effective vaccine against NiV infection, in the present study, we have designed two Multi-Epitope Vaccines (MEVs) composed of 33 Cytotoxic T lymphocyte (CTL) epitopes and 38 Helper T lymphocyte (HTL) epitopes. Out of those CTL and HTL combined 71 epitopes, 61 novel epitopes targeting nine different NiV proteins were not used before for vaccine design. Codon optimization for the cDNA of both the designed MEVs might ensure high expression potential in the human cell line as stable proteins. Both MEVs carry potential B cell linear epitope overlapping regions, B cell discontinuous epitopes as well as IFN-γ inducing epitopes. Additional criteria such as sequence consensus amongst CTL, HTL and B Cell epitopes was implemented for the design of final constructs constituting MEVs. Hence, the designed MEVs carry the potential to elicit cell-mediated as well as humoral immune response. Selected overlapping CTL and HTL epitopes were validated for their stable molecular interactions with HLA class I and II alleles and in case of CTL epitopes with human Transporter Associated with antigen Processing (TAP) cavity. The structure based epitope cross validation for interaction with TAP cavity was used as another criteria choosing final epitopes for NiV MEVs. Finally, human Beta-defensin 2 and Beta-defensin 3 were used as adjuvants to enhance the immune response of both the MEVs. Molecular dynamics simulation studies of MEVs-TLR3 ectodomain (Human Toll-Like Receptor 3) complex indicated the stable molecular interaction. We conclude that the MEVs designed and in silico validated here could be highly potential vaccine candidates to combat NiV infections, with great effectiveness, high specificity and large human population coverage worldwide.
Collapse
Affiliation(s)
- Sukrit Srivastava
- Infection Biology Group, Indian Foundation for Fundamental Research Trust, RaeBareli, India
- Department for Structural Infection Biology, Centre for Structural Systems Biology (CSSB) & Helmholtz-Centre for Infection Research, Hamburg, Germany
| | - Sonia Verma
- Protein Biochemistry & Engineering Lab, Parasite-Host Biology Group, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Mohit Kamthania
- Infection Biology Group, Indian Foundation for Fundamental Research Trust, RaeBareli, India
| | - Ajay Kumar Saxena
- Molecular Medicine Lab., School of Life Science, Jawaharlal Nehru University, New Delhi, India
| | - Kailash C. Pandey
- Protein Biochemistry & Engineering Lab, Parasite-Host Biology Group, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Veena Pande
- Kumaun University, Bheemtal, Nainital, Uttarakhand, 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
| |
Collapse
|
31
|
Antiviral Peptides in Antimicrobial Surface Coatings—From Current Techniques to Potential Applications. Viruses 2023; 15:v15030640. [PMID: 36992349 PMCID: PMC10051592 DOI: 10.3390/v15030640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
The transmission of pathogens through contact with contaminated surfaces is an important route for the spread of infections. The recent outbreak of COVID-19 highlights the necessity to attenuate surface-mediated transmission. Currently, the disinfection and sanitization of surfaces are commonly performed in this regard. However, there are some disadvantages associated with these practices, including the development of antibiotic resistance, viral mutation, etc.; hence, a better strategy is necessary. In recent years, peptides have been studied to be utilized as a potential alternative. They are part of the host immune defense and have many potential in vivo applications in drug delivery, diagnostics, immunomodulation, etc. Additionally, the ability of peptides to interact with different molecules and membrane surfaces of microorganisms has made it possible to exploit them in ex vivo applications such as antimicrobial (antibacterial and antiviral) coatings. Although antibacterial peptide coatings have been studied extensively and proven to be effective, antiviral coatings are a more recent development. Therefore, this study aims to highlight antiviral coating strategies and the current practices and application of antiviral coating materials in personal protective equipment, healthcare devices, and textiles and surfaces in public settings. Here, we have presented a review on potential techniques to incorporate peptides in current surface coating strategies that will serve as a guide for developing cost-effective, sustainable and coherent antiviral surface coatings. We further our discussion to highlight some challenges of using peptides as a surface coating material and to examine future perspectives.
Collapse
|
32
|
Purewal JS, Doshi GM. Deciphering the Function of New Therapeutic Targets and Prospective Biomarkers in the Management of Psoriasis. Curr Drug Targets 2023; 24:1224-1238. [PMID: 38037998 DOI: 10.2174/0113894501277656231128060242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/29/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Psoriasis is an immune-mediated skin condition affecting people worldwide, presenting at any age, and leading to a substantial burden physically and mentally. The innate and adaptive immune systems interact intricately with the pathomechanisms that underlie disease. T cells can interact with keratinocytes, macrophages, and dendritic cells through the cytokines they secrete. According to recent research, psoriasis flare-ups can cause systemic inflammation and various other co-morbidities, including depression, psoriatic arthritis, and cardio-metabolic syndrome. Additionally, several auto-inflammatory and auto-immune illnesses may be linked to psoriasis. Although psoriasis has no proven treatment, care must strive by treating patients as soon as the disease surfaces, finding and preventing concurrent multimorbidity, recognising and reducing bodily and psychological distress, requiring behavioural modifications, and treating each patient individually. Biomarkers are traits that are assessed at any time along the clinical continuum, from the early stages of a disease through the beginning of treatment (the foundation of precision medicine) to the late stages of treatment (outcomes and endpoints). Systemic therapies that are frequently used to treat psoriasis provide a variety of outcomes. Targeted therapy selection, better patient outcomes, and more cost-effective healthcare would be made possible by biomarkers that reliably predict effectiveness and safety. This review is an attempt to understand the role of Antimicrobial peptides (AMP), Interleukin-38 (IL-38), autophagy 5 (ATG5) protein and squamous cell carcinoma antigen (SCCA) as biomarkers of psoriasis.
Collapse
Affiliation(s)
- Japneet Singh Purewal
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Mahesh Doshi
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| |
Collapse
|
33
|
Roberto de Souza Fonseca R, Valois Laurentino R, Fernando Almeida Machado L, Eduardo Vieira da Silva Gomes C, Oliveira de Alencar Menezes T, Faciola Pessoa O, Branco Oliveira-Filho A, Resque Beckmann Carvalho T, Gabriela Faciola Pessoa de Oliveira P, Brito Tanaka E, Sá Elias Nogueira J, Magno Guimarães D, Newton Carneiro M, Mendes Acatauassú Carneiro P, Ferreira Celestino Junior A, de Almeida Rodrigues P, Augusto Fernandes de Menezes S. HIV Infection and Oral Manifestations: An Update. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.105894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Human immunodeficiency virus (HIV) causes a complete depletion of the immune system; it has been a major health issue around the world since the 1980s, and due to the reduction of CD4+ T lymphocytes levels, it can trigger various opportunistic infections. Oral lesions are usually accurate indicators of immunosuppression because these oral manifestations may occur as a result of the compromised immune system caused by HIV infection; therefore, oral lesions might be initial and common clinical features in people living with HIV. So, it is necessary to evaluate and understand the mechanism, prevalence, and risk factors of oral lesions to avoid the increase morbidity among those with oral diseases.
Collapse
|
34
|
Peng H, Fan P, Hong Y, He J, Zeng X, Xu F, Zhao Y, Jin T. 3' untranslated region variants in DEFA5 gene associated with susceptibility to IgA nephropathy in the Chinese Han population. Biomark Med 2022; 16:1151-1159. [PMID: 36632813 DOI: 10.2217/bmm-2022-0518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aim: To evaluate the association between single-nucleotide polymorphisms in the 3' untranslated region of the DEFA5 gene and IgA nephropathy (IgAN) risk, the authors performed an association study in the Chinese Han population. Materials & methods: The authors recruited 426 IgAN patients and 498 controls. The MassARRAY platform (Agena Bioscience, Inc., CA, USA) was used to genotype single-nucleotide polymorphisms in DEFA5. Odds ratios and 95% CIs were calculated through logistic regression analysis. Results: The authors observed that rs12716641 significantly reduced IgAN risk in the allele (odds ratio: 0.77; p = 0.026) and genotype (odds ratio: 0.75; p = 0.039) models. Stratification analysis revealed that several genotypes of rs12716641 played a protective role against IgAN. Conclusion: The authors' results revealed that single-nucleotide polymorphisms in the 3' untranslated region of DEFA5 were associated with IgAN risk.
Collapse
Affiliation(s)
- Hongwei Peng
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Pingyun Fan
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Yifen Hong
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Jing He
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Xiangxiu Zeng
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Fengguo Xu
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Yunxia Zhao
- Renal Medicine, Hainan Wanning People's Hospital, Wanning, Hainan Province, 571500, China
| | - Tianbo Jin
- Key Laboratory of Resource Biology & Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, Shaanxi, 710000, China.,Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, China
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Paneth cells are specialized, secretory epithelial cells located in the small intestine. Although their existence was first described in 1872, their precise role in the gut remained unclear for over a century. Over the past few decades, elegant studies have shown Paneth cells play a key role enhancing gut barrier function, as niche cells for the intestinal stem cell compartment and via secreting antimicrobial peptides to establish an antimicrobial barrier at the epithelial surface. This review describes what is known about Paneth cell biology from human and animal studies with a focus on their putative role in clinical gastrointestinal disease. RECENT FINDINGS Recent work has demonstrated important associations of dysfunctional Paneth cells with several gastrointestinal disorders. These include Crohn's disease, enteric infections, graft-versus-host disease, necrotizing enterocolitis, and environmental enteric dysfunction. Ongoing studies are examining precisely how Paneth cell biology is altered in these various disease states. SUMMARY By understanding the mechanisms of Paneth cell regulation - and how these processes go awry in specific gastrointestinal diseases - we set the stage for using Paneth cells as biomarkers for disease progression and developing novel therapeutics that augment Paneth cell function to treat a spectrum of gastrointestinal disorders.
Collapse
Affiliation(s)
- Vivian H. Lee
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ajay S. Gulati
- Department of Pediatrics, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Antimicrobial Activity Developed by Scorpion Venoms and Its Peptide Component. Toxins (Basel) 2022; 14:toxins14110740. [PMID: 36355990 PMCID: PMC9693228 DOI: 10.3390/toxins14110740] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/28/2022] [Accepted: 10/23/2022] [Indexed: 01/26/2023] Open
Abstract
Microbial infections represent a problem of great importance at the public health level, with a high rate of morbidity-mortality worldwide. However, treating the different diseases generated by microorganisms requires a gradual increase in acquired resistance when applying or using them against various antibiotic therapies. Resistance is caused by various molecular mechanisms of microorganisms, thus reducing their effectiveness. Consequently, there is a need to search for new opportunities through natural sources with antimicrobial activity. One alternative is using peptides present in different scorpion venoms, specifically from the Buthidae family. Different peptides with biological activity in microorganisms have been characterized as preventing their growth or inhibiting their replication. Therefore, they represent an alternative to be used in the design and development of new-generation antimicrobial drugs in different types of microorganisms, such as bacteria, fungi, viruses, and parasites. Essential aspects for its disclosure, as shown in this review, are the studies carried out on different types of peptides in scorpion venoms with activity against pathogenic microorganisms, highlighting their high therapeutic potential.
Collapse
|
37
|
Pang Y, Zheng Y, Yang N, Zan M, Zhang L, Ding W. Potential novel biomarkers in small intestine for obesity/obesity resistance revealed by multi-omics analysis. Lipids Health Dis 2022; 21:98. [PMID: 36209126 PMCID: PMC9547412 DOI: 10.1186/s12944-022-01711-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Background Although obesity is caused by different factors, individual susceptibility to obesity differs among people under the same circumstances. The microbiota in the caecum or fresh faeces and metabolites in blood or urine contribute to obesity resistance; however, the microbiota or metabolites in the small intestine have not been extensively studied. Methods To investigate the relationship between the microbiota or metabolites in the small intestine and susceptibility to obesity, eighty-eight male C57BL/6 mice were fed a high-fat diet (HFD) for 8 weeks to establish two models of obesity and obesity resistance. For further study, six mice were chosen from among the obesity models, and twelve mice were randomly chosen from among the obesity resistance models. After fasting plasma glucose and behavioural testing, the mice were fed in single cages for another 4 weeks to observe their weight and food intake. All mice were sacrificed at 20 weeks of age. Serum ALT, AST, HDL, LDL, TG and TC levels were measured using an automatic biochemical analyser. The microbiota and metabolites in the small intestine contents were analysed using 16 S sequencing and an ultrahigh-performance liquid chromatographic system, respectively. Transcripts in the jejunum were evaluated using full-length transcriptome sequencing and verified by qPCR. Results The results showed that HFD induced depression and anxiety behaviours and higher fasting plasma glucose, ALT, AST, HDL, LDL, TG and TC levels in the obese mice; however, these levels were improved in obese resistance mice. The correlation analysis showed that the phosphatidylcholine, TG, and phosphatidylethanolamine levels were higher in obese mice and correlated positively with intestinal microflora (Desulfovibrio and Gemella) and the Cxcl10 gene. A higher abundance of Clostridium_sensu_stricto_1 in obesity-resistant mice correlated negatively with the metabolite contents (neuromedin N and enkephalin L) and Pck1 gene expression and correlated positively with certain metabolites (5-hydroxy-L-tryptophan, cinnamyl alcohol and 1 H-indole-3-acetamide) and genes expression (Gdf15, Igfbp6 and Spp1). Conclusion Clostridium_sensu_stricto_1, neuromedin N, enkephalin L, Pck1, 5-hydroxy-L-tryptophan, Cxcl10 and cinnamyl alcohol may be novel biomarkers in the small intestine for obesity/obesity resistance. These might be helpful for obesity prevention or for treating obese patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01711-0.
Collapse
Affiliation(s)
- Yueshan Pang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.,The Second Clinical Medical College, North SiChuan Medical College, 637000, Nanchong, China
| | - Yali Zheng
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Ni Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Meng Zan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Lu Zhang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - WeiJun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.
| |
Collapse
|
38
|
Zhai YJ, Feng Y, Ma X, Ma F. Defensins: defenders of human reproductive health. Hum Reprod Update 2022; 29:126-154. [PMID: 36130055 PMCID: PMC9825273 DOI: 10.1093/humupd/dmac032] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/31/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Reproductive tract infection is an important factor leading to male and female infertility. Among female infertility factors, microbial and viral infections are the main factors affecting female reproductive health and causing tubal infertility, ectopic tubal pregnancy and premature delivery. Among male infertility factors, 13-15% of male infertility is related to infection. Defensins are cationic antibacterial and antiviral peptides, classified into α-defensins, β-defensins and θ-defensins. Humans only have α-defensins and β-defensins. Apart from their direct antimicrobial functions, defensins have an immunomodulatory function and are involved in many physiological processes. Studies have shown that defensins are widely distributed in the female reproductive tract (FRT) and male reproductive tract (MRT), playing a dual role of host defence and fertility protection. However, to our knowledge, the distribution, regulation and function of defensins in the reproductive tract and their relation to reproduction have not been reviewed. OBJECTIVE AND RATIONALE This review summarizes the expression, distribution and regulation of defensins in the reproductive tracts to reveal the updated research on the dual role of defensins in host defence and the protection of fertility. SEARCH METHODS A systematic search was conducted in PubMed using the related keywords through April 2022. Related data from original researches and reviews were integrated to comprehensively review the current findings and understanding of defensins in the human reproductive system. Meanwhile, female and male transcriptome data in the GEO database were screened to analyze defensins in the human reproductive tracts. OUTCOMES Two transcriptome databases from the GEO database (GSE7307 and GSE150852) combined with existing researches reveal the expression levels and role of the defensins in the reproductive tracts. In the FRT, a high expression level of α-defensin is found, and the expression levels of defensins in the vulva and vagina are higher than those in other organs. The expression of defensins in the endometrium varies with menstrual cycle stages and with microbial invasion. Defensins also participate in the local immune response to regulate the risk of spontaneous preterm birth. In the MRT, a high expression level of β-defensins is also found. It is mainly highly expressed in the epididymal caput and corpus, indicating that defensins play an important role in sperm maturation. The expression of defensins in the MRT varies with androgen levels, age and the status of microbial invasion. They protect the male reproductive system from bacterial infections by neutralizing lipopolysaccharide and downregulating pro-inflammatory cytokines. In addition, animal and clinical studies have shown that defensins play an important role in sperm maturation, motility and fertilization. WIDER IMPLICATIONS As a broad-spectrum antimicrobial peptide without drug resistance, defensin has great potential for developing new natural antimicrobial treatments for reproductive tract infections. However, increasing evidence has shown that defensins can not only inhibit microbial invasion but can also promote the invasion and adhesion of some microorganisms in certain biological environments, such as human immunodeficiency virus. Therefore, the safety of defensins as reproductive tract anti-infective drugs needs more in-depth research. In addition, the modulatory role of defensins in fertility requires more in-depth research since the current conclusions are based on small-size samples. At present, scientists have made many attempts at the clinical transformation of defensins. However, defensins have problems such as poor stability, low bioavailability and difficulties in their synthesis. Therefore, the production of safe, effective and low-cost drugs remains a challenge.
Collapse
Affiliation(s)
| | | | - Xue Ma
- Correspondence address. Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China. E-mail: https://orcid.org/0000-0002-7781-821X (F.M.); Department of Pediatric Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China. E-mail: https://orcid.org/0000-0002-7650-6214 (X.M.)
| | - Fang Ma
- Correspondence address. Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China. E-mail: https://orcid.org/0000-0002-7781-821X (F.M.); Department of Pediatric Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China. E-mail: https://orcid.org/0000-0002-7650-6214 (X.M.)
| |
Collapse
|
39
|
Tran HTT, Peterburs P, Seibel J, Abramov-Sommariva D, Lamy E. In vitro Screening of Herbal Medicinal Products for Their Supportive Curing Potential in the Context of SARS-CoV-2. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8038195. [PMID: 36110194 PMCID: PMC9470301 DOI: 10.1155/2022/8038195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/06/2022] [Accepted: 07/23/2022] [Indexed: 11/17/2022]
Abstract
COVID-19 herbal medicinal products may have the potential for symptom relief in nonsevere or moderate disease cases. In this in vitro study we screened the five herbal medicinal products Sinupret extract (SINx), Bronchipret thyme-ivy (BRO-TE), Bronchipret thyme-primula (BRO TP), Imupret (IMU), and Tonsipret (TOP) with regard to their potential to (i) interfere with the binding of the human angiotensin-converting enzyme 2 (ACE2) receptor with the SARS-CoV-2 spike S1 protein, (ii) modulate the release of the human defensin HBD1 and cathelicidin LL-37 from human A549 lung cells upon spike S1 protein stimulation, and (iii) modulate the release of IFN-γ from activated human peripheral blood mononuclear cells (PBMC). The effect of the extracts on the interaction of spike S1 protein and the human ACE2 receptor was measured by ELISA. The effects on the intracellular IFN-γ expression in stimulated human PBMC were measured by flow cytometry. Regulation of HBD1 and LL-37 expression and secretion was assessed in 25 d long-term cultured human lung A549 epithelial cells by RT-PCR and ELISA. IMU and BRO-TE concentration-dependently inhibited the interaction between spike S1 protein and the ACE2 receptor. SINx, TOP, and BRO-TE significantly upregulated the intracellular expression of anti-viral IFN-γ from stimulated PBMC. Cotreatment of A549 cells with IMU or BRO TP together with SARS-CoV-2 spike protein significantly upregulated mRNA expression (IMU) and release of HBD1 (IMU and BRO TP) and LL-37 (BRO TP). The in vitro screening results provide first evidence for an immune-activating potential of some of the tested herbal medicinal extracts in the context of SARS-CoV-2. Whether these could be supportive in symptom relief or curing from SARS-CoV-2 infection needs deeper understanding of the observations.
Collapse
Affiliation(s)
- Hoai Thi Thu Tran
- Molecular Preventive Medicine, University of Freiburg, Medical Center and Faculty of Medicine, Engesserstraße 4, 79108 Freiburg, Germany
| | | | - Jan Seibel
- Bionorica SE, Kerschensteinerstraße 11-15, 92318 Neumarkt, Germany
| | | | - Evelyn Lamy
- Molecular Preventive Medicine, University of Freiburg, Medical Center and Faculty of Medicine, Engesserstraße 4, 79108 Freiburg, Germany
| |
Collapse
|
40
|
Identification of antiviral peptide inhibitors for receptor binding domain of SARS-CoV-2 omicron and its sub-variants: an in-silico approach. 3 Biotech 2022; 12:198. [PMID: 35923684 PMCID: PMC9342843 DOI: 10.1007/s13205-022-03258-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/08/2022] [Indexed: 11/01/2022] Open
Abstract
Omicron, a variant of concern (VOC) of SARS-CoV-2, emerged in South Africa in November 2021. Omicron has been continuously acquiring a series of new mutations, especially in the spike (S) protein that led to high infectivity and transmissibility. Peptides targeting the receptor-binding domain (RBD) of the spike protein by which omicron and its variants attach to the host receptor, angiotensin-converting enzyme (ACE2) can block the viral infection at the first step. This study aims to identify antiviral peptides from the Antiviral peptide database (AVPdb) and HIV-inhibitory peptide database (HIPdb) against the RBD of omicron by using a molecular docking approach. The lead RBD binder peptides obtained through molecular docking were screened for allergenicity and physicochemical criteria (isoelectric point (pI) and net charge) required for peptide-based drugs. The binding affinity of the best five peptide inhibitors with the RBD of omicron was validated further by molecular dynamics (MD) simulation. Our result introduces five antiviral peptides, including AVP1056, AVP1059, AVP1225, AVP1801, and HIP755, that may effectively hinder omicron-host interactions. It is worth mentioning that all the three major sub-variants of omicron, BA.1 (B.1.1.529.1), BA.2 (B.1.1.529.2), and BA.3 (B.1.1.529.3), exhibits conserved ACE-2 interacting residues. Hence, the screened antiviral peptides with similar affinity can also interrupt the RBD-mediated invasion of different major sub-variants of omicron. Altogether, these peptides can be considered in the peptide-based therapeutics development for omicron treatment after further experimentation. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03258-4.
Collapse
|
41
|
Zeng F, Wang M, Li J, Li C, Pan X, Meng L, Li L, Wei H, Zhang S. Involvement of Porcine β-Defensin 129 in Sperm Capacitation and Rescue of Poor Sperm in Genital Tract Infection. Int J Mol Sci 2022; 23:ijms23169441. [PMID: 36012708 PMCID: PMC9409293 DOI: 10.3390/ijms23169441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/13/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
In mammals, β-defensins have been reported to play pivotal roles in sperm protection and fertilization. However, the function and mechanism of porcine β-defensin 129 (pBD129) in the sperm remain unclear. Here, we demonstrate that pBD129 is a glycosylated protein and broadly exists in accessory sex glands and coats the sperm surface. We inhibited the pBD129 protein on the sperm surface with an anti-pBD129 antibody and found that sperm motility was not significantly affected; however, sperm acrosome integrity and tyrosine phosphorylation levels increased significantly with time (p < 0.05) during capacitation. These changes were accompanied by an increase in sperm Ca2+ influx, resulting in a significantly reduced in vitro fertilization cleavage rate (p < 0.05). Further investigation revealed that treatment with recombinant pBD129 markedly restored the sperm motility in semen contaminated with Escherichia coli. The results suggest that pBD129 is not only associated with poor sperm motility after genital tract infection but can also protect the spermatozoa from premature capacitation, which may be beneficial for semen preservation.
Collapse
|
42
|
Oral Papillomatosis: Its Relation with Human Papilloma Virus Infection and Local Immunity—An Update. Medicina (B Aires) 2022; 58:medicina58081103. [PMID: 36013570 PMCID: PMC9415166 DOI: 10.3390/medicina58081103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Oral papilloma lesions may appear as a result of HPV infection, or not, and only special molecular methods could differentiate them. Low-risk and high-risk HPV types could induce oral HPV papillomatosis with different natural evolution, clearance and persistence mechanisms. The pathogenic mechanisms are based on the crosstalk between the oral epithelial and immune cells and this very efficient virus. HPV acts as a direct inducer in the process of transforming a benign lesion into a malignant one, the cancerization process being also debated in this paper. According to the degree of malignity, three types of papillomatous lesions can be described in the oral cavity: benign lesions, potential malign disorders and malignant lesions. The precise molecular diagnostic is important to identify the presence of various virus types and also the virus products responsible for its oncogenicity. An accurate diagnostic of oral papilloma can be established through a good knowledge of etiological and epidemiological factors, clinical examination and laboratory tests. This review intends to update the pathogenic mechanisms driving the macroscopic and histological features of oral papillomatosis having HPV infection as the main etiological factor, focusing on its interreference in the local immunity. In the absence of an accurate molecular diagnostic and knowledge of local immunological conditions, the therapeutic strategy could be difficult to decide.
Collapse
|
43
|
León-Sosa A, Castañeda V, Espinosa-Vallejo R, Gómez X, Díaz RF, Cabrera F, Caicedo A. Key points for translating wound regenerative agents from in vivo assays in mice to clinical validation. Cytotherapy 2022; 24:1074-1086. [DOI: 10.1016/j.jcyt.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/07/2022] [Accepted: 07/16/2022] [Indexed: 11/29/2022]
|
44
|
Aggeletopoulou I, Thomopoulos K, Mouzaki A, Triantos C. Vitamin D–VDR Novel Anti-Inflammatory Molecules—New Insights into Their Effects on Liver Diseases. Int J Mol Sci 2022; 23:ijms23158465. [PMID: 35955597 PMCID: PMC9369388 DOI: 10.3390/ijms23158465] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 02/05/2023] Open
Abstract
There is consistent evidence that vitamin D deficiency is strongly associated with liver dysfunction, disease severity, and poor prognosis in patients with liver disease. Vitamin D and its receptor (VDR) contribute to the regulation of innate and adaptive immune responses. The presence of genetic variants of vitamin D- and VDR-associated genes has been associated with liver disease progression. In our recent work, we summarized the progress in understanding the molecular mechanisms involved in vitamin D–VDR signaling and discussed the functional significance of VDR signaling in specific cell populations in liver disease. The current review focuses on the complex interaction between immune and liver cells in the maintenance of liver homeostasis and the development of liver injury, the interplay of vitamin D and VDR in the development and outcome of liver disease, the role of vitamin D- and VDR-associated genetic variants in modulating the occurrence and severity of liver disease, and the therapeutic value of vitamin D supplementation in various liver diseases. The association of the vitamin D–VDR complex with liver dysfunction shows great potential for clinical application and supports its use as a prognostic index and diagnostic tool.
Collapse
Affiliation(s)
- Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, GR-26504 Patras, Greece; (I.A.); (C.T.)
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, GR-26504 Patras, Greece;
| | - Konstantinos Thomopoulos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, GR-26504 Patras, Greece; (I.A.); (C.T.)
- Correspondence:
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, GR-26504 Patras, Greece;
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, GR-26504 Patras, Greece; (I.A.); (C.T.)
| |
Collapse
|
45
|
Antiviral Effect of hBD-3 and LL-37 during Human Primary Keratinocyte Infection with West Nile Virus. Viruses 2022; 14:v14071552. [PMID: 35891533 PMCID: PMC9319560 DOI: 10.3390/v14071552] [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: 05/18/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 02/01/2023] Open
Abstract
West Nile virus (WNV) is an emerging flavivirus transmitted through mosquito bites and responsible for a wide range of clinical manifestations. Following their inoculation within the skin, flaviviruses replicate in keratinocytes of the epidermis, inducing an innate immune response including the production of antimicrobial peptides (AMPs). Among them, the cathelicidin LL-37 and the human beta-defensin (hBD)-3 are known for their antimicrobial and immunomodulatory properties. We assessed their role during WNV infection of human primary keratinocytes. LL-37 reduced the viral load in the supernatant of infected keratinocytes and of the titer of a viral inoculum incubated in the presence of the peptide, suggesting a direct antiviral effect of this AMP. Conversely, WNV replication was not inhibited by hBD-3. The two peptides then demonstrated immunomodulatory properties whether in the context of keratinocyte stimulation by poly(I:C) or infection by WNV, but not alone. This study demonstrates the immunostimulatory properties of these two skin AMPs at the initial site of WNV replication and the ability of LL-37 to directly inactivate West Nile viral infectious particles. The results provide new information on the multiple functions of these two peptides and underline the potential of AMPs as new antiviral strategies in the fight against flaviviral infections.
Collapse
|
46
|
Laude V, Nuño M, Moses RC, Guthrie D. Evaluation of unexpected protecting group removal in solid-phase peptide synthesis - quantified using continuous flow methods. J Pept Sci 2022; 28:e3441. [PMID: 35785412 DOI: 10.1002/psc.3441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/09/2022]
Abstract
As peptides gained interest as new drugs in the past years, their synthetic routes had been the subject of review and improvement. Fmoc/tBu-based solid-phase peptide synthesis (SPPS) is the most convenient technique, and the implementation in continuous flow has allowed for single-pass coupling and deprotection reactions. The focus of this research is to evaluate the relationship between undesired solvent-promoted reactions and final crude purity, by studying volume changes of a variable bed flow reactor through the synthesis. Based on the temperature, typical solvents for SPPS such as dimethylformamide (DMF) or N-methyl-2-pyrrolidone (NMP) can cause unwanted Fmoc removal during wash steps. It was found that for every millilitre of DMF used at 80 °C, up to 1 μmol of Fmoc-protected peptide is deprotected, leading to additional impurities. This effect can, however, be minimised by adding additives such as HOBt; which reduces such unwanted deprotection to just 0.1 μmol/ml.
Collapse
|
47
|
Liew YJM, Ibrahim PAS, Ong HM, Chong CN, Tan CT, Schee JP, Gómez Román R, Cherian NG, Wong WF, Chang LY. The Immunobiology of Nipah Virus. Microorganisms 2022; 10:microorganisms10061162. [PMID: 35744680 PMCID: PMC9228579 DOI: 10.3390/microorganisms10061162] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/23/2022] Open
Abstract
Nipah virus (NiV) is a highly lethal zoonotic paramyxovirus that emerged in Malaysia in 1998. It is a human pathogen capable of causing severe respiratory infection and encephalitis. The natural reservoir of NiV, Pteropus fruit bats, remains a continuous virus source for future outbreaks, although infection in the bats is largely asymptomatic. NiV provokes serious disease in various mammalian species. In the recent human NiV outbreaks in Bangladesh and India, both bats-to-human and human-to-human transmissions have been observed. NiV has been demonstrated to interfere with the innate immune response via interferon type I signaling, promoting viral dissemination and preventing antiviral response. Studies of humoral immunity in infected NiV patients and animal models have shown that NiV-specific antibodies were produced upon infection and were protective. Studies on cellular immunity response to NiV infection in human and animal models also found that the adaptive immune response, specifically CD4+ and CD8+ T cells, was stimulated upon NiV infection. The experimental vaccines and therapeutic strategies developed have provided insights into the immunological requirements for the development of successful medical countermeasures against NiV. This review summarizes the current understanding of NiV pathogenesis and innate and adaptive immune responses induced upon infection.
Collapse
Affiliation(s)
- Yvonne Jing Mei Liew
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
- Deputy Vice Chancellor’s Office (Research & Innovation), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Puteri Ainaa S. Ibrahim
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Hui Ming Ong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Chee Ning Chong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Chong Tin Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (C.T.T.); (J.P.S.)
| | - Jie Ping Schee
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (C.T.T.); (J.P.S.)
| | - Raúl Gómez Román
- Vaccine Research and Development, Coalition for Epidemic Preparedness Innovation (CEPI), Askekroken 11, 0277 Oslo, Norway; (R.G.R.); (N.G.C.)
| | - Neil George Cherian
- Vaccine Research and Development, Coalition for Epidemic Preparedness Innovation (CEPI), Askekroken 11, 0277 Oslo, Norway; (R.G.R.); (N.G.C.)
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Li-Yen Chang
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
- Correspondence:
| |
Collapse
|
48
|
Zhang J, Zhang J, Wang Y, Zhang X, Nie T, Liu Y. Strategies to Improve the Activity and Biocompatibility: Modification of Peptide Antibiotics. Foodborne Pathog Dis 2022; 19:376-385. [PMID: 35713924 DOI: 10.1089/fpd.2021.0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
As host defense peptides, peptide antibiotics exist in almost all organisms. Many of their activities come from their inactivation of bacteria, yeast, fungi, and even cancer cells. However, natural peptide antibiotics are relatively poor in stability and penetration, and have high hemolytic properties, which makes them difficult to directly apply. Therefore, natural peptide antibiotics can be modified to enhance their activity and biocompatibility. Based on the characteristics of amino acids, amino acid substitutions can be performed to study the effect of amino acid types on the activity of peptide antibiotics. The design of ultrashort peptides, cyclic peptides, and self-assembling peptides is also a way to improve the activity of peptide antibiotics. In addition, antibacterial peptides can also be conjugated with antibiotics, lipids, or metal ions to prepare antibacterial peptides with special activities. This review introduces several methods for modifying peptide antibiotics and their specific applications, providing a theoretical basis for the further application of peptide antibiotics.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Jin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Yitong Wang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Ting Nie
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| |
Collapse
|
49
|
Hu J, Brendle SA, Li JJ, Walter V, Cladel NM, Cooper T, Shearer DA, Balogh KK, Christensen ND. Depo Medroxyprogesterone (DMPA) Promotes Papillomavirus Infections but Does Not Accelerate Disease Progression in the Anogenital Tract of a Mouse Model. Viruses 2022; 14:v14050980. [PMID: 35632722 PMCID: PMC9147738 DOI: 10.3390/v14050980] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Contraceptives such as Depo-medroxyprogesterone (DMPA) are used by an estimated 34 million women worldwide. DMPA has been associated with increased risk of several viral infections including Herpes simplex virus-2 (HSV-2) and Human immunodeficiency virus (HIV). In the current study, we used the mouse papillomavirus (MmuPV1) anogenital infection model to test two hypotheses: (1) contraceptives such as DMPA increase the susceptibility of the anogenital tract to viral infection and (2) long-term contraceptive administration induces more advanced disease at the anogenital tract. DMPA treatments of both athymic nude mice and heterozygous NU/J (Foxn1nu/+) but ovariectomized mice led to a significantly increased viral load at the anogenital tract, suggesting that endogenous sex hormones were involved in increased viral susceptibility by DMPA treatment. Consistent with previous reports, DMPA treatment suppressed host anti-viral activities at the lower genital tract. To test the impact of long-term contraceptive treatment on the MmuPV1-infected lower genital tract, we included two other treatments in addition to DMPA: 17β-estradiol and a non-hormone based contraceptive Cilostazol (CLZ, Pletal). Viral infections were monitored monthly up to nine months post infection by qPCR. The infected vaginal and anal tissues were harvested and further examined by histological, virological, and immunological analyses. Surprisingly, we did not detect a significantly higher grade of histology in animals in the long-term DMPA and 17β-estradiol treated groups when compared to the control groups in the athymic mice we tested. Therefore, although DMPA promotes initial papillomavirus infections in the lower genital tract, the chronic administration of DMPA does not promote cancer development in the infected tissues in our mouse model.
Collapse
Affiliation(s)
- Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
- Correspondence:
| | - Sarah A. Brendle
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Jingwei J. Li
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Vonn Walter
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA;
- Department of Biochemistry and Molecular Biology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Nancy M. Cladel
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Timothy Cooper
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, NIH, Fort Detrick, Frederick, MD 21702, USA;
| | - Debra A. Shearer
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Karla K. Balogh
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Neil D. Christensen
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (S.A.B.); (J.J.L.); (N.M.C.); (D.A.S.); (K.K.B.); (N.D.C.)
- Department of Pathology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
50
|
Charoenkwan P, Schaduangrat N, Mahmud SMH, Thinnukool O, Shoombuatong W. Recent development of machine learning-based methods for the prediction of defensin family and subfamily. EXCLI JOURNAL 2022; 21:757-771. [PMID: 35949489 PMCID: PMC9360473 DOI: 10.17179/excli2022-4913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/03/2022] [Indexed: 11/05/2022]
Abstract
Nearly all living species comprise of host defense peptides called defensins, that are crucial for innate immunity. These peptides work by activating the immune system which kills the microbes directly or indirectly, thus providing protection to the host. Thus far, numerous preclinical and clinical trials for peptide-based drugs are currently being evaluated. Although, experimental methods can help to precisely identify the defensin peptide family and subfamily, these approaches are often time-consuming and cost-ineffective. On the other hand, machine learning (ML) methods are able to effectively employ protein sequence information without the knowledge of a protein's three-dimensional structure, thus highlighting their predictive ability for the large-scale identification. To date, several ML methods have been developed for the in silico identification of the defensin peptide family and subfamily. Therefore, summarizing the advantages and disadvantages of the existing methods is urgently needed in order to provide useful suggestions for the development and improvement of new computational models for the identification of the defensin peptide family and subfamily. With this goal in mind, we first provide a comprehensive survey on a collection of six state-of-the-art computational approaches for predicting the defensin peptide family and subfamily. Herein, we cover different important aspects, including the dataset quality, feature encoding methods, feature selection schemes, ML algorithms, cross-validation methods and web server availability/usability. Moreover, we provide our thoughts on the limitations of existing methods and future perspectives for improving the prediction performance and model interpretability. The insights and suggestions gained from this review are anticipated to serve as a valuable guidance for researchers for the development of more robust and useful predictors.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, Thailand, 50200
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700
| | - S. M. Hasan Mahmud
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700,Department of Computer Science, American International University-Bangladesh (AIUB), Kuratoli, Dhaka 1229, Bangladesh
| | - Orawit Thinnukool
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, Thailand, 50200
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700,*To whom correspondence should be addressed: Watshara Shoombuatong, Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand, 10700; Phone: +66 2 441 4371, Fax: +66 2 441 4380, E-mail:
| |
Collapse
|