1
|
Rao AS, Ugwu N, Onufer AP, Kumova O, Carey AJ. Cathelicidin-related antimicrobial peptide (CRAMP) is toxic during neonatal murine influenza virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:1022-1031. [PMID: 40101750 PMCID: PMC12123214 DOI: 10.1093/jimmun/vkae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/13/2024] [Indexed: 03/20/2025]
Abstract
Respiratory viral infections are a major contributor to mortality in children under 5 years of age, and disproportionately affect preterm neonates. Previously, using our established 3-day-old neonatal murine model of influenza virus infection, we demonstrated that treatment of neonatal mice with intranasal Lactobacillus rhamnosus GG (LGG) prior to influenza viral infection improved survival. Transcriptional analysis revealed expression of the mouse cathelicidin-related antimicrobial peptide (CRAMP, encoded by CRAMP) was downregulated in LGG-treated neonates. Mouse CRAMP is a key effector protein secreted by infected epithelial cells and resident and infiltrating immune cells, but the role of CRAMP in neonatal defense to respiratory viruses is unknown. Neonatal mice with a deleted CRAMP gene (CRAMP-/-) were intranasally infected with influenza virus. CRAMP-/- neonates had improved survival over C57BL/6 neonates after influenza viral infection (75% vs. 14%, p < 0.05). Next, immune cell recruitment to the lung of infected neonates was determined. Surprisingly, at 3-days postinfection, there was increased recruitment of neutrophils, inflammatory monocytes, and alveolar macrophages, coupled with increased proinflammatory cytokine and chemokine production in CRAMP-/- compared to C57BL/6 neonates. However, this changed over the first week of infection. C57BL/6 neonatal mice increased CRAMP production significantly, in direct contrast to their adult counterparts. Inflammatory cytokine production increased that indicated CRAMP amplified the innate immune response later in the infection. Furthermore, we identified pulmonary nonimmune cells as an important source of increased CRAMP levels as the infection progressed and CRAMP production drove mortality. These insights emphasize the age-specific role of CRAMP in influenza viral pathogenesis.
Collapse
Affiliation(s)
- Abhishek S Rao
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nneka Ugwu
- Department of Pediatrics, St. Christopher’s Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, United States
- Current affiliation: Department of Pediatrics, School of Medicine & Health Sciences, University of North Dakota, Bismarck, ND, United States
| | - Abigail P Onufer
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ogan Kumova
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Current affiliation: National Academies of Science, Engineering, and Medicine, Washington, DC, United States
| | - Alison J Carey
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Pediatrics, St. Christopher’s Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
2
|
Tanneti NS, Stillwell HA, Weiss SR. Human coronaviruses: activation and antagonism of innate immune responses. Microbiol Mol Biol Rev 2025; 89:e0001623. [PMID: 39699237 PMCID: PMC11948496 DOI: 10.1128/mmbr.00016-23] [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] [Indexed: 12/20/2024] Open
Abstract
SUMMARYHuman coronaviruses cause a range of respiratory diseases, from the common cold (HCoV-229E, HCoV-NL63, HCoV-OC43, and SARS-CoV-2) to lethal pneumonia (SARS-CoV, SARS-CoV-2, and MERS-CoV). Coronavirus interactions with host innate immune antiviral responses are an important determinant of disease outcome. This review compares the host's innate response to different human coronaviruses. Host antiviral defenses discussed in this review include frontline defenses against respiratory viruses in the nasal epithelium, early sensing of viral infection by innate immune effectors, double-stranded RNA and stress-induced antiviral pathways, and viral antagonism of innate immune responses conferred by conserved coronavirus nonstructural proteins and genus-specific accessory proteins. The common cold coronaviruses HCoV-229E and -NL63 induce robust interferon signaling and related innate immune pathways, SARS-CoV and SARS-CoV-2 induce intermediate levels of activation, and MERS-CoV shuts down these pathways almost completely.
Collapse
Affiliation(s)
- Nikhila S. Tanneti
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helen A. Stillwell
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Khan A, Zakirullah, Wahab S, Hong ST. Advances in antiviral strategies targeting mosquito-borne viruses: cellular, viral, and immune-related approaches. Virol J 2025; 22:26. [PMID: 39905499 PMCID: PMC11792744 DOI: 10.1186/s12985-025-02622-z] [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: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025] Open
Abstract
Mosquito-borne viruses (MBVs) are a major global health threat, causing significant morbidity and mortality. MBVs belong to several distinct viral families, each with unique characteristics. The primary families include Flaviviridae (e.g., Dengue, Zika, West Nile, Yellow Fever, Japanese Encephalitis), transmitted predominantly by Aedes and Culex mosquitoes; Togaviridae, which consists of the genus Alphavirus (e.g., Chikungunya, Eastern and Western Equine Encephalitis viruses), also transmitted by Aedes and Culex; Bunyaviridae (recently reorganized), containing viruses like Rift Valley Fever and Oropouche virus, transmitted by mosquitoes and sometimes sandflies; and Reoviridae, which includes the genus Orbivirus (e.g., West Nile and Bluetongue viruses), primarily affecting animals and transmitted by mosquitoes and sandflies. Despite extensive research, effective antiviral treatments for MBVs remain scarce, and current therapies mainly provide symptomatic relief and supportive care. This review examines the viral components and cellular and immune factors involved in the life cycle of MBVs. It also highlights recent advances in antiviral strategies targeting host factors such as lipid metabolism, ion channels, and proteasomes, as well as viral targets like NS2B-NS3 proteases and nonstructural proteins. Additionally, it explores immunomodulatory therapies to enhance antiviral responses and emphasizes the potential of drug repurposing, bioinformatics, artificial intelligence, and deep learning in identifying novel antiviral candidates. Continued research is crucial in mitigating MBVs' impact and preventing future outbreaks.
Collapse
Affiliation(s)
- Ayyaz Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shahid Wahab
- Department of Agriculture, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea.
| |
Collapse
|
4
|
Keshri AK, Rawat SS, Chaudhary A, Sharma S, Kapoor A, Mehra P, Kaur R, Mishra A, Prasad A. LL-37, the master antimicrobial peptide, its multifaceted role from combating infections to cancer immunity. Int J Antimicrob Agents 2025; 65:107398. [PMID: 39643165 DOI: 10.1016/j.ijantimicag.2024.107398] [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: 08/07/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Antimicrobial peptides (AMPs) represent a unique group of naturally occurring molecules having diverse biological activities, including potent antimicrobial properties. Among them, LL-37 has emerged as a significant player, demonstrating its multifaceted roles during bacterial, fungal, and viral infections, as well as exhibiting intriguing implications in cancer. This review delves into the versatile functions of LL-37, elucidating its mechanisms of action against microbial pathogens and its potential to modulate immune responses. We explored the efficacy of LL-37 in disrupting bacterial membranes, inhibiting fungal growth, and interfering with viral replication, highlighting its potential as a therapeutic agent against a wide array of infectious diseases. Furthermore, we discussed the emerging role of LL-37 in cancer immunity, where its immunomodulatory effects and direct cytotoxicity towards cancer cells offer novel avenues for cancer therapy in the near future. We provided a comprehensive overview of the activities of LL-37 across various diseases and underscored the importance of further research into harnessing the therapeutic potential of this potential antimicrobial peptide along with other suitable candidates.
Collapse
Affiliation(s)
- Anand K Keshri
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Suraj S Rawat
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Anubha Chaudhary
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Swati Sharma
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Ananya Kapoor
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Parul Mehra
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Rimanpreet Kaur
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Amit Prasad
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India; Indian Knowledge System and Mental Health Application Centre, Indian Institute of Technology Mandi, Himachal Pradesh, India.
| |
Collapse
|
5
|
Paludan SR, Pradeu T, Pichlmair A, Wray KB, Mikkelsen JG, Olagnier D, Mogensen TH. Early host defense against virus infections. Cell Rep 2024; 43:115070. [PMID: 39675007 DOI: 10.1016/j.celrep.2024.115070] [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: 06/19/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024] Open
Abstract
Early host defense eliminates many viruses before infections are established while clearing others so they remain subclinical or cause only mild disease. The field of immunology has been shaped by broad concepts, including the pattern recognition theory that currently dominates innate immunology. Focusing on early host responses to virus infections, we analyze the literature to build a working hypothesis for the principles that govern the early line of cellular antiviral defense. Aiming to ultimately arrive at a criteria-based theory with strong explanatory power, we propose that both controlling infection and limiting inflammation are key drivers for the early cellular antiviral response. This response, which we suggest is exerted by a set of "microbe- and inflammation-restricting mechanisms," directly restrict viral replication while also counteracting inflammation. Exploring the mechanisms and physiological importance of the early layer of cellular antiviral defense may open further lines of research in immunology.
Collapse
Affiliation(s)
- Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Thomas Pradeu
- CNRS UMR 5164 ImmunoConcept, University of Bordeaux, Bordeaux, France; Department of Biological and Medical Sciences, University of Bordeaux, Bordeaux, France; Chapman University, Orange, CA, USA
| | - Andreas Pichlmair
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - K Brad Wray
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Centre for Science Studies, Aarhus University, Aarhus, Denmark; Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - David Olagnier
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Trine H Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
6
|
Pashaie F, Hoornweg TE, Bikker FJ, Veenendaal T, Broere F, Veldhuizen EJA. Antiviral activity of cathelicidins against porcine epidemic diarrhea virus (PEDV): Mechanisms, and efficacy. Virus Res 2024; 350:199496. [PMID: 39528011 PMCID: PMC11607671 DOI: 10.1016/j.virusres.2024.199496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a harmful coronavirus infecting pigs, which is resulting in substantial financial losses in the global pig industry. The lack of effective vaccines or treatments underscores the pressing need for new antiviral strategies. Antimicrobial peptides (AMPs), specifically cathelicidins such as LL-37, have demonstrated promising activity against a range of viruses. This study aims to elucidate the antiviral mechanisms of cathelicidins by examining their inhibitory capabilities against PEDV in vitro. Four pig-derived antimicrobial peptides (PMAP-36, PMAP-23, PR-39, and PG-1), together with chicken-derived CATH-B1 and human-derived LL-37 were analyzed for their anti-PEDV activity. Flow cytometry and fluorescent microscopy confirmed that LL-37 and CATH-B1 had strong inhibitory effects at non-toxic concentrations of 5 and 10 µM, significantly reducing GFP-PEDV infection of Vero cells both in co- and pre-incubation setups. In contrast, none of the porcine peptides exhibited any inhibitory effects, even at higher doses. Fluorogenic LL-37 was shown to enter VERO cells, indicative of a possible immunomodulatory antiviral mode of action. However, transmission electron microscopy clearly indicated that both LL-37 and CATH-B1 affected virus morphology and caused aggregation of viral particles, showing that peptide-virus interaction caused reduced virus infectivity. In conclusion, this analysis highlights the potential of LL-37 and CATH-B1 as inhibitors against PEDV, suggesting promising directions for innovative therapeutic antiviral strategies.
Collapse
Affiliation(s)
- Fatemeh Pashaie
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, the Netherlands
| | - Tabitha E Hoornweg
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, the Netherlands
| | - Floris J Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, Amsterdam 1081 LA, the Netherlands
| | - Tineke Veenendaal
- Cell Microscopy Core, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CX, the Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, the Netherlands
| | - Edwin J A Veldhuizen
- Department of Biomolecular Health Sciences, Division Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, the Netherlands.
| |
Collapse
|
7
|
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
|
8
|
Chronopoulos J, Pernet E, Tran KA, McGovern TK, Morozan A, Wang S, Tsai O, Makita K, Divangahi M, Martin JG. Pregnancy enhances antiviral immunity independent of type I IFN but dependent on IL-17-producing γδ + T cells in the nasal mucosa. SCIENCE ADVANCES 2024; 10:eado7087. [PMID: 39331716 PMCID: PMC11430450 DOI: 10.1126/sciadv.ado7087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024]
Abstract
Pregnancy is associated with profound changes in immunity. However, pregnancy-related respiratory immune adaptations in response to influenza infection and their impact on disease severity remain unclear. Here, we describe, in a preclinical model of mid-gestation pregnancy, a mechanism of enhanced host defense against influenza A virus (IAV) localized to the nasal cavity that limits viral replication and reduces the magnitude of intrapulmonary immune responses. Consequently, the pregnant mice show reduced pulmonary pathology and preserved airway function after IAV infection. The early restriction of viral replication is independent of type I interferon (IFN) but dependent on increased antimicrobial peptides (AMPs) driven by interleukin-17+ (IL-17+) γδ+ T cells within the nasal passages. This pathway of host defense against IAV infection in the upper airways during pregnancy restricts early viral infection and prevents virus dissemination into the lung supporting maternal fitness.
Collapse
Affiliation(s)
- Julia Chronopoulos
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Erwan Pernet
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medical Biology, Université du Québec à Trois-Rivières, Quebec, Canada
| | - Kim A. Tran
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Toby K. McGovern
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Arina Morozan
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Sadie Wang
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Oscar Tsai
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Kosuke Makita
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - James G. Martin
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Urmi UL, Vijay AK, Willcox MDP, Attard S, Enninful G, Kumar N, Islam S, Kuppusamy R. Exploring the Efficacy of Peptides and Mimics against Influenza A Virus, Adenovirus, and Murine Norovirus. Int J Mol Sci 2024; 25:7030. [PMID: 39000138 PMCID: PMC11240954 DOI: 10.3390/ijms25137030] [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: 05/14/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The ongoing battle against viral pandemics continues, with the possibility of future outbreaks. The search for effective antiviral compounds that can combat a diverse range of viruses continues to be a focal point of research. This study investigated the efficacy of two natural antimicrobial peptides (AMPs) (lactoferricin and LL-37), two synthetic AMPs (melimine and Mel4), and nine AMP mimics (758, 1091, 1096, 1083, 610, NAPL, 3-BIPL, 4-BIPL, and Sau-22) against influenza A virus strains H1N1 and H3N2, human adenovirus 5 (HAdV-5), and murine norovirus 1 (MNV-1). These compounds were tested using virus pre-treatment, cell pre-treatment, or post-cell entry treatment assays, electron microscopy, and circular dichroism (CD), alongside evaluations of cytotoxicity against the host cells. After virus pre-treatment, the AMP mimics 610 and Sau-22 had relatively low IC50 values for influenza strains H1N1 (2.35 and 6.93 µM, respectively) and H3N2 (3.7 and 5.34 µM, respectively). Conversely, natural and synthetic AMPs were not active against these strains. For the non-enveloped viruses, the AMP Mel4 and mimic 1083 had moderate activity against HAdV-5 (Mel4 IC50 = 47.4 µM; 1083 IC50 = 47.2 µM), whereas all AMPs, but none of the mimics, were active against norovirus (LL-37 IC50 = 4.2 µM; lactoferricin IC50 = 23.18 µM; melimine IC50 = 4.8 µM; Mel4 IC50 = 8.6 µM). Transmission electron microscopy demonstrated that the mimics targeted the outer envelope of influenza viruses, while the AMPs targeted the capsid of non-enveloped viruses. CD showed that Mel4 adopted an α-helical structure in a membrane mimetic environment, but mimic 758 remained unstructured. The diverse activity against different virus groups is probably influenced by charge, hydrophobicity, size, and, in the case of natural and synthetic AMPs, their secondary structure. These findings underscore the potential of peptides and mimics as promising candidates for antiviral therapeutics against both enveloped and non-enveloped viruses.
Collapse
Affiliation(s)
- Umme Laila Urmi
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ajay Kumar Vijay
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark D P Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Samuel Attard
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - George Enninful
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Salequl Islam
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh
| | - Rajesh Kuppusamy
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
- The Drug Discovery Initiative, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
10
|
Li J, He S, Yang H, Zhang L, Xiao J, Liang C, Liu S. The Main Mechanisms of Mesenchymal Stem Cell-Based Treatments against COVID-19. Tissue Eng Regen Med 2024; 21:545-556. [PMID: 38573476 PMCID: PMC11087407 DOI: 10.1007/s13770-024-00633-5] [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: 04/11/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has a clinical manifestation of hypoxic respiratory failure and acute respiratory distress syndrome. However, COVID-19 still lacks of effective clinical treatments so far. As a promising potential treatment against COVID-19, stem cell therapy raised recently and had attracted much attention. Here we review the mechanisms of mesenchymal stem cell-based treatments against COVID-19, and provide potential cues for the effective control of COVID-19 in the future. METHODS Literature is obtained from databases PubMed and Web of Science. Key words were chosen for COVID- 19, acute respiratory syndrome coronavirus 2, mesenchymal stem cells, stem cell therapy, and therapeutic mechanism. Then we summarize and critically analyze the relevant articles retrieved. RESULTS Mesenchymal stem cell therapy is a potential effective treatment against COVID-19. Its therapeutic efficacy is mainly reflected in reducing severe pulmonary inflammation, reducing lung injury, improving pulmonary function, protecting and repairing lung tissue of the patients. Possible therapeutic mechanisms might include immunoregulation, anti-inflammatory effect, tissue regeneration, anti-apoptosis effect, antiviral, and antibacterial effect, MSC - EVs, and so on. CONCLUSION Mesenchymal stem cells can effectively treat COVID-19 through immunoregulation, anti-inflammatory, tissue regeneration, anti-apoptosis, anti-virus and antibacterial, MSC - EVs, and other ways. Systematically elucidating the mechanisms of mesenchymal stem cell-based treatments for COVID-19 will provide novel insights into the follow-up research and development of new therapeutic strategies in next step.
Collapse
Affiliation(s)
- Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Laboratory of Basic Medicine Center, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Shipei He
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Hang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Lizeai Zhang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jie Xiao
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Chaoyi Liang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
11
|
Bhattacharjya S, Zhang Z, Ramamoorthy A. LL-37: Structures, Antimicrobial Activity, and Influence on Amyloid-Related Diseases. Biomolecules 2024; 14:320. [PMID: 38540740 PMCID: PMC10968335 DOI: 10.3390/biom14030320] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 11/11/2024] Open
Abstract
Antimicrobial peptides (AMPs), as well as host defense peptides (HDPs), constitute the first line of defense as part of the innate immune system. Humans are known to express antimicrobial precursor proteins, which are further processed to generate AMPs, including several types of α/β defensins, histatins, and cathelicidin-derived AMPs like LL37. The broad-spectrum activity of AMPs is crucial to defend against infections caused by pathogenic bacteria, viruses, fungi, and parasites. The emergence of multi-drug resistant pathogenic bacteria is of global concern for public health. The prospects of targeting antibiotic-resistant strains of bacteria with AMPs are of high significance for developing new generations of antimicrobial agents. The 37-residue long LL37, the only cathelicidin family of AMP in humans, has been the major focus for the past few decades of research. The host defense activity of LL37 is likely underscored by its expression throughout the body, spanning from the epithelial cells of various organs-testis, skin, respiratory tract, and gastrointestinal tract-to immune cells. Remarkably, apart from canonical direct killing of pathogenic organisms, LL37 exerts several other host defense activities, including inflammatory response modulation, chemo-attraction, and wound healing and closure at the infected sites. In addition, LL37 and its derived peptides are bestowed with anti-cancer and anti-amyloidogenic properties. In this review article, we aim to develop integrative, mechanistic insight into LL37 and its derived peptides, based on the known biophysical, structural, and functional studies in recent years. We believe that this review will pave the way for future research on the structures, biochemical and biophysical properties, and design of novel LL37-based molecules.
Collapse
Affiliation(s)
- Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Zhizhuo Zhang
- Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, The University of Michigan, Ann Arbor, MI 48109, USA;
| | - Ayyalusamy Ramamoorthy
- Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, The University of Michigan, Ann Arbor, MI 48109, USA;
- National High Magnetic Field Laboratory, Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
12
|
Murayama A, Igarashi H, Yamada N, Aly HH, Molchanova N, Lin JS, Nishitsuji H, Shimotohno K, Muramatsu M, Barron AE, Kato T. Antiviral effect of peptoids on hepatitis B virus infection in cell culture. Antiviral Res 2024; 223:105821. [PMID: 38272318 PMCID: PMC10939774 DOI: 10.1016/j.antiviral.2024.105821] [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: 11/08/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Although antimicrobial peptides have been shown to inactivate viruses through disruption of their viral envelopes, clinical use of such peptides has been hampered by a number of factors, especially their enzymatically unstable structures. To overcome the shortcomings of antimicrobial peptides, peptoids (sequence-specific N-substituted glycine oligomers) mimicking antimicrobial peptides have been developed. We aimed to demonstrate the antiviral effects of antimicrobial peptoids against hepatitis B virus (HBV) in cell culture. The anti-HBV activity of antimicrobial peptoids was screened and evaluated in an infection system involving the HBV reporter virus and HepG2.2.15-derived HBV. By screening with the HBV reporter virus infection system, three (TM1, TM4, and TM19) of 12 peptoids were identified as reducing the infectivity of HBV, though they did not alter the production levels of HBs antigen in cell culture. These peptoids were not cytotoxic at the evaluated concentrations. Among these peptoids, TM19 was confirmed to reduce HBV infection most potently in a HepG2.2.15-derived HBV infection system that closely demonstrates authentic HBV infection. In cell culture, the most effective administration of TM19 was virus treatment at the infection step, but the reduction in HBV infectivity by pre-treatment or post-treatment of cells with TM19 was minimal. The disrupting effect of TM19 targeting infectious viral particles was clarified in iodixanol density gradient analysis. In conclusion, the peptoid TM19 was identified as a potent inhibitor of HBV. This peptoid prevents HBV infection by disrupting viral particles and is a candidate for a new class of anti-HBV reagents.
Collapse
Affiliation(s)
- Asako Murayama
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hitomi Igarashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Norie Yamada
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hussein Hassan Aly
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Natalia Molchanova
- Department of Bioengineering, Stanford University School of Medicine & School of Engineering, Stanford, CA, 94305, USA
| | - Jennifer S Lin
- Department of Bioengineering, Stanford University School of Medicine & School of Engineering, Stanford, CA, 94305, USA
| | - Hironori Nishitsuji
- Department of Virology, Fujita Health University School of Medicine, Aichi, Japan
| | - Kunitada Shimotohno
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Chiba, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan; Department of Infectious Disease Research, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Annelise E Barron
- Department of Bioengineering, Stanford University School of Medicine & School of Engineering, Stanford, CA, 94305, USA
| | - Takanobu Kato
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
13
|
Watts S, Hänni E, Smith GN, Mahmoudi N, Freire RVM, Lim S, Salentinig S. Human antimicrobial peptide inactivation mechanism of enveloped viruses. J Colloid Interface Sci 2024; 657:971-981. [PMID: 38096780 DOI: 10.1016/j.jcis.2023.11.055] [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: 09/24/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 01/02/2024]
Abstract
HYPOTHESIS Enveloped viruses are pivotal in causing various illnesses, including influenza and COVID-19. The antimicrobial peptide LL-37, a critical part of the human innate immune system, exhibits potential as an antiviral agent capable of thwarting these viral threats. Its mode of action involves versatile and non-specific interactions that culminate in dismantling the viral envelope, ultimately rendering the viruses inert. However, the exact mechanism of action is not yet understood. EXPERIMENTS Here, the mechanism of LL-37 triggered changes in the structure and function of an enveloped virus is investigated. The bacteriophage "Phi6" is used as a surrogate for pathogenic enveloped viruses. Small angle X-ray and neutron scattering combined with light scattering techniques demonstrate that LL-37 actively integrates into the virus's lipid envelope. FINDINGS LL-37 addition to Phi6 leads to curvature modification in the lipid bilayer, ultimately separating the envelope from the nucleocapsid. Additional biological assays confirm the loss of virus infectivity in the presence of LL-37, which coincides with the structural transformations. The results provide a fundamental understanding of the structure-activity relationship related to enveloped viruses. The knowledge of peptide-virus interactions can guide the design of future peptide-based antiviral drugs and therapies.
Collapse
Affiliation(s)
- Samuel Watts
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland; School of Chemistry, Chemical Engineering and Biotechnology, 70 Nanyang Drive, Block N1.3, Nanyang Technological University, Singapore 637457, Singapore
| | - Eliane Hänni
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Gregory N Smith
- ISIS Neutron and Muon Souce, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Didcot OX11 0QX, United Kingdom
| | - Najet Mahmoudi
- ISIS Neutron and Muon Souce, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Didcot OX11 0QX, United Kingdom
| | - Rafael V M Freire
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Sierin Lim
- School of Chemistry, Chemical Engineering and Biotechnology, 70 Nanyang Drive, Block N1.3, Nanyang Technological University, Singapore 637457, Singapore
| | - Stefan Salentinig
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland.
| |
Collapse
|
14
|
Thathapudi NC, Callai-Silva N, Malhotra K, Basu S, Aghajanzadeh-Kiyaseh M, Zamani-Roudbaraki M, Groleau M, Lombard-Vadnais F, Lesage S, Griffith M. Modified host defence peptide GF19 slows TNT-mediated spread of corneal herpes simplex virus serotype I infection. Sci Rep 2024; 14:4096. [PMID: 38374240 PMCID: PMC10876564 DOI: 10.1038/s41598-024-53662-4] [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: 12/11/2023] [Accepted: 02/03/2024] [Indexed: 02/21/2024] Open
Abstract
Corneal HSV-1 infections are a leading cause of infectious blindness globally by triggering tissue damage due to the intense inflammation. HSV-1 infections are treated mainly with antiviral drugs that clear the infections but are inefficient as prophylactics. The body produces innate cationic host defence peptides (cHDP), such as the cathelicidin LL37. Various epithelia, including the corneal epithelium, express LL37. cHDPs can cause disintegration of pathogen membranes, stimulate chemokine production, and attract immune cells. Here, we selected GF17, a peptide containing the LL37 fragment with bioactivity but with minimal cytotoxicity, and added two cell-penetrating amino acids to enhance its activity. The resulting GF19 was relatively cell-friendly, inducing only partial activation of antigen presenting immune cells in vitro. We showed that HSV-1 spreads by tunneling nanotubes in cultured human corneal epithelial cells. GF19 given before infection was able to block infection, most likely by blocking viral entry. When cells were sequentially exposed to viruses and GF19, the infection was attenuated but not arrested, supporting the contention that the GF19 mode of action was to block viral entry. Encapsulation into silica nanoparticles allowed a more sustained release of GF19, enhancing its activity. GF19 is most likely suitable as a prevention rather than a virucidal treatment.
Collapse
Affiliation(s)
- Neethi C Thathapudi
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Natalia Callai-Silva
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Kamal Malhotra
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, University of Ottawa, Ottawa, K1Y 4W7, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Sankar Basu
- Department of Microbiology, Asutosh College, (Affiliated With University of Calcutta), Kolkata, 700026, India
| | - Mozhgan Aghajanzadeh-Kiyaseh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Mostafa Zamani-Roudbaraki
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Marc Groleau
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | | | - Sylvie Lesage
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada.
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
15
|
Cutuli SL, Ferrando ES, Cammarota F, Franchini E, Caroli A, Lombardi G, Tanzarella ES, Grieco DL, Antonelli M, De Pascale G. Update on vitamin D role in severe infections and sepsis. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2024; 4:4. [PMID: 38263252 PMCID: PMC10804708 DOI: 10.1186/s44158-024-00139-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
Severe infections frequently require admission to the intensive care unit and cause life-threatening complications in critically ill patients. In this setting, severe infections are acknowledged as prerequisites for the development of sepsis, whose pathophysiology implies a dysregulated host response to pathogens, leading to disability and mortality worldwide.Vitamin D is a secosteroid hormone that plays a pivotal role to maintain immune system homeostasis, which is of paramount importance to resolve infection and modulate the burden of sepsis. Specifically, vitamin D deficiency has been widely reported in critically ill patients and represents a risk factor for the development of severe infections, sepsis and worse clinical outcomes. Several studies have demonstrated the feasibility, safety and effectiveness of vitamin D supplementation strategies to improve vitamin D body content, but conflictual results support its benefit in general populations of critically ill patients. In contrast, small randomised clinical trials reported that vitamin D supplementation may improve host-defence to pathogen invasion via the production of cathelicidin and specific cytokines. Nonetheless, no large scale investigations have been designed to specifically assess the impact of vitamin D supplementation on the outcome of critically ill septic patients admitted to the intensive care unit.
Collapse
Affiliation(s)
- Salvatore Lucio Cutuli
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Fabiola Cammarota
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emanuele Franchini
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Caroli
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gianmarco Lombardi
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eloisa Sofia Tanzarella
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico Luca Grieco
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimo Antonelli
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gennaro De Pascale
- Department of Emergency, Intensive Care Medicine and Anesthesia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Istituto Di Anestesiologia E Rianimazione, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
16
|
Kalia V, Sarkar S. Vitamin D and antiviral immunity. FELDMAN AND PIKE'S VITAMIN D 2024:1011-1034. [DOI: 10.1016/b978-0-323-91338-6.00045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Bhatt T, Dam B, Khedkar SU, Lall S, Pandey S, Kataria S, Ajnabi J, Gulzar SEJ, Dias PM, Waskar M, Raut J, Sundaramurthy V, Vemula PK, Ghatlia N, Majumdar A, Jamora C. Niacinamide enhances cathelicidin mediated SARS-CoV-2 membrane disruption. Front Immunol 2023; 14:1255478. [PMID: 38022563 PMCID: PMC10663372 DOI: 10.3389/fimmu.2023.1255478] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
The continual emergence of SARS-CoV-2 variants threatens to compromise the effectiveness of worldwide vaccination programs, and highlights the need for complementary strategies for a sustainable containment plan. An effective approach is to mobilize the body's own antimicrobial peptides (AMPs), to combat SARS-CoV-2 infection and propagation. We have found that human cathelicidin (LL37), an AMP found at epithelial barriers as well as in various bodily fluids, has the capacity to neutralise multiple strains of SARS-CoV-2. Biophysical and computational studies indicate that LL37's mechanism of action is through the disruption of the viral membrane. This antiviral activity of LL37 is enhanced by the hydrotropic action of niacinamide, which may increase the bioavailability of the AMP. Interestingly, we observed an inverse correlation between LL37 levels and disease severity of COVID-19 positive patients, suggesting enhancement of AMP response as a potential therapeutic avenue to mitigate disease severity. The combination of niacinamide and LL37 is a potent antiviral formulation that targets viral membranes of various variants and can be an effective strategy to overcome vaccine escape.
Collapse
Affiliation(s)
- Tanay Bhatt
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Binita Dam
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Department of Biological Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Sneha Uday Khedkar
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Sahil Lall
- National Centre for Biological Sciences (TIFR), Bangalore, Karnataka, India
| | - Subhashini Pandey
- Integrative Chemical Biology, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Sunny Kataria
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Johan Ajnabi
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Department of Biological Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | | | | | | | | | | | - Praveen Kumar Vemula
- Integrative Chemical Biology, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | | | | | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| |
Collapse
|
18
|
Ali F, Narges N, Amin J. Restraint of VP1 Protein of Foot and Mouth Disease Virus using Specific Antiviral Peptides: an in Silico Investigation. ARCHIVES OF RAZI INSTITUTE 2023; 78:1483-1495. [PMID: 38590669 PMCID: PMC10998950 DOI: 10.22092/ari.2023.78.5.1483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 04/10/2024]
Abstract
Foot and mouth diseases are among the important threats in the animal husbandry industry which lead to huge economic losses. In this regard, the current project aimed to inhibit the VP1 protein of foot and mouth disease viruses using specific peptides. For this purpose, a wide range of potential antiviral peptides were collected from the database. Physicochemical properties, hydrophobicity/hydrophilicity, and solubility properties of potential antiviral peptides were investigated using reliable servers. Afterward, the tertiary structures of the selected peptides along with the VP1 protein were modeled by the I-TASSER server. Moreover, interactions between VP1 protein and selected antiviral peptides were investigated using the ClusPro 2.0 server. Finally, the outputs of molecular docking were assessed by LigPlot+ and visualized by PyMol software. The results revealed that Dermaseptin-3, Ginkbilobin, Circulin-F, Maximin1, Cycloviolin-A, Cycloviolin-D, Circulin-C, Cycloviolin-C, and Antihypertensive protein BDS-1 peptides with a hydrophobicity value of > 30 were soluble with positive instability index and positive net charge. Moreover, the results of the molecular docking process demonstrated that Dermaseptin-3 and Ginkbilobin peptides could strongly inhibit the VP1 protein using 10 hydrogen bonds. Therefore, these two peptides, which had the most hydrogen bonds, were introduced as the best anti-foot and mouth disease virus peptides to apply.
Collapse
Affiliation(s)
- Forouharmehr Ali
- Department of Animal Science, Faculty of Agriculture, Lorestan University, Khorramabad, Iran
| | - Nazifi Narges
- Department of Basic Sciences, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Jaydari Amin
- Department of Microbiology, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| |
Collapse
|
19
|
von Beck T, Navarrete K, Arce NA, Gao M, Dale GA, Davis-Gardner ME, Floyd K, Mena Hernandez L, Mullick N, Vanderheiden A, Skountzou I, Kuchipudi SV, Saravanan R, Li R, Skolnick J, Suthar MS, Jacob J. A wild boar cathelicidin peptide derivative inhibits severe acute respiratory syndrome coronavirus-2 and its drifted variants. Sci Rep 2023; 13:14650. [PMID: 37670110 PMCID: PMC10480232 DOI: 10.1038/s41598-023-41850-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/31/2023] [Indexed: 09/07/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a clear threat to humanity. It has infected over 200 million and killed 4 million people worldwide, and infections continue with no end in sight. To control the pandemic, multiple effective vaccines have been developed, and global vaccinations are in progress. However, the virus continues to mutate. Even when full vaccine coverage is achieved, vaccine-resistant mutants will likely emerge, thus requiring new annual vaccines against drifted variants analogous to influenza. A complimentary solution to this problem could be developing antiviral drugs that inhibit SARS CoV-2 and its drifted variants. Host defense peptides represent a potential source for such an antiviral as they possess broad antimicrobial activity and significant diversity across species. We screened the cathelicidin family of peptides from 16 different species for antiviral activity and identified a wild boar peptide derivative that inhibits SARS CoV-2. This peptide, which we named Yongshi and means warrior in Mandarin, acts as a viral entry inhibitor. Following the binding of SARS-CoV-2 to its receptor, the spike protein is cleaved, and heptad repeats 1 and 2 multimerize to form the fusion complex that enables the virion to enter the cell. A deep learning-based protein sequence comparison algorithm and molecular modeling suggest that Yongshi acts as a mimetic to the heptad repeats of the virus, thereby disrupting the fusion process. Experimental data confirm the binding of Yongshi to the heptad repeat 1 with a fourfold higher affinity than heptad repeat 2 of SARS-CoV-2. Yongshi also binds to the heptad repeat 1 of SARS-CoV-1 and MERS-CoV. Interestingly, it inhibits all drifted variants of SARS CoV-2 that we tested, including the alpha, beta, gamma, delta, kappa and omicron variants.
Collapse
Affiliation(s)
- Troy von Beck
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Karla Navarrete
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Nicholas A Arce
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mu Gao
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, GA, 30332, USA
| | - Gordon A Dale
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Meredith E Davis-Gardner
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Katharine Floyd
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Luis Mena Hernandez
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Nikita Mullick
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Abigail Vanderheiden
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Ioanna Skountzou
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Suresh V Kuchipudi
- Animal Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, 16802, USA
| | - Rathi Saravanan
- Centre of Regulatory Excellence (CoRE), Duke-NUS Medical School, Level 6, 8 College Road, Singapore, 169857, Singapore
| | - Renhao Li
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, GA, 30332, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Joshy Jacob
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA.
| |
Collapse
|
20
|
Samanipour R, Tabatabaee S, delyanee M, Tavakoli A. The promising approach of MSCs therapy for COVID-19 treatment. Cell Tissue Bank 2023; 24:597-612. [PMID: 36526819 PMCID: PMC9757632 DOI: 10.1007/s10561-022-10060-2] [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: 10/30/2021] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Several ongoing investigations have been founded on the development of an optimized therapeutic strategy for the COVID-19 virus as an undeniable acute challenge for human life. Cell-based therapy and particularly, mesenchymal stem cells (MSCs) therapy has obtained desired outcomes in decreasing the mortality rate of severe acute respiratory syndrome coronavirus 2 (SARS-COV-2), mainly owing to its immunoregulatory impact that prevents the overactivation of the immune system. Also, these cells with their multipotent nature, are capable of repairing the damaged tissue of the lung which leads to reducing the probability of acute respiratory distress syndrome (ARDS). Although this cell-based method is not quite cost-effective for developing countries, regarding its promising results in order to treat SARS-COV-2, more economical evaluation as well as clinical trials should be performed for improving this therapeutic approach. Here in this article, the functional mechanism of MSCs therapy for the treatment of COVID-19 and the clinical trials based on this method will be reviewed. Moreover, its economic efficiency will be discussed.
Collapse
Affiliation(s)
- Reza Samanipour
- Department of Tissue Engineering and Applied Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Tabatabaee
- Department of Bio-Computing, Faculty of Interdisciplinary Sciences and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Mahsa delyanee
- Biomedical Engineering Department, Amirkabir University of Technology, Tehran, Iran
| | - Amirhossein Tavakoli
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Tomkinson S, Triscott C, Schenk E, Foey A. The Potential of Probiotics as Ingestible Adjuvants and Immune Modulators for Antiviral Immunity and Management of SARS-CoV-2 Infection and COVID-19. Pathogens 2023; 12:928. [PMID: 37513775 PMCID: PMC10384479 DOI: 10.3390/pathogens12070928] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Probiotic bacteria are able to modulate general antiviral responsiveness, including barrier functionality and innate and adaptive immune responses. The COVID-19 pandemic, resulting from SARS-CoV-2 infection, has created a need to control and treat this viral infection and its ensuing immunopathology with a variety of approaches; one such approach may involve the administration of probiotic bacteria. As with most viral infections, its pathological responses are not fully driven by the virus, but are significantly contributed to by the host's immune response to viral infection. The potential adoption of probiotics in the treatment of COVID-19 will have to appreciate the fine line between inducing antiviral immunity without over-provoking immune inflammatory responses resulting in host-derived immunopathological tissue damage. Additionally, the effect exerted on the immune system by SARS-CoV-2 evasion strategies will also have to be considered when developing a robust response to this virus. This review will introduce the immunopathology of COVID-19 and the immunomodulatory effects of probiotic strains, and through their effects on a range of respiratory pathogens (IAV, SARS-CoV, RSV), as well as SARS-CoV-2, will culminate in a focus on how these bacteria can potentially manipulate both infectivity and immune responsiveness via barrier functionality and both innate and adaptive immunity. In conclusion, the harnessing of induction and augmentation of antiviral immunity via probiotics may not only act as an ingestible adjuvant, boosting immune responsiveness to SARS-CoV-2 infection at the level of barrier integrity and innate and adaptive immunity, but also act prophylactically to prevent infection and enhance protection afforded by current vaccine regimens.
Collapse
Affiliation(s)
- Sophie Tomkinson
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Cloe Triscott
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Emily Schenk
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
- Peninsula Medical School, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Andrew Foey
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| |
Collapse
|
22
|
Arora A, Singh A. Exploring the role of neutrophils in infectious and noninfectious pulmonary disorders. Int Rev Immunol 2023; 43:41-61. [PMID: 37353973 DOI: 10.1080/08830185.2023.2222769] [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: 03/25/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
With the change in global environment, respiratory disorders are becoming more threatening to the health of people all over the world. These diseases are closely linked to performance of immune system. Within the innate arm of immune system, Neutrophils are an important moiety to serve as an immune defense barrier. They are one of the first cells recruited to the site of infection and plays a critical role in pathogenesis of various pulmonary diseases. It is established that the migration and activation of neutrophils can lead to inflammation either directly or indirectly and this inflammation caused is very crucial for the clearance of pathogens and resolution of infection. However, the immunopathological mechanisms involved to carry out the same is very complex and not well understood. Despite there being studies concentrating on the role of neutrophils in multiple respiratory diseases, there is still a long way to go in order to completely understand the complexity of the participation of neutrophils and mechanisms involved in the development of these respiratory diseases. In the present article, we have reviewed the literature to comprehensively provide an insight in the current development and advancements about the role of neutrophils in infectious respiratory disorders including viral respiratory disorders such as Coronavirus disease (COVID-19) and bacterial pulmonary disorders with a focused review on pulmonary tuberculosis as well as in noninfectious disorders like Chronic obstructive pulmonary disease (COPD) and asthma. Also, future directions into research and therapeutic targets have been discussed for further exploration.
Collapse
Affiliation(s)
- Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
23
|
van Dijk A, Guabiraba R, Bailleul G, Schouler C, Haagsman HP, Lalmanach AC. Evolutionary diversification of defensins and cathelicidins in birds and primates. Mol Immunol 2023; 157:53-69. [PMID: 36996595 DOI: 10.1016/j.molimm.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Divergent evolution for more than 310 million years has resulted in an avian immune system that is complex and more compact than that of primates, sharing much of its structure and functions. Not surprisingly, well conserved ancient host defense molecules, such as defensins and cathelicidins, have diversified over time. In this review, we describe how evolution influenced the host defense peptides repertoire, its distribution, and the relationship between structure and biological functions. Marked features of primate and avian HDPs are linked to species-specific characteristics, biological requirements, and environmental challenge.
Collapse
|
24
|
Sowers A, Wang G, Xing M, Li B. Advances in Antimicrobial Peptide Discovery via Machine Learning and Delivery via Nanotechnology. Microorganisms 2023; 11:1129. [PMID: 37317103 PMCID: PMC10223199 DOI: 10.3390/microorganisms11051129] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
Antimicrobial peptides (AMPs) have been investigated for their potential use as an alternative to antibiotics due to the increased demand for new antimicrobial agents. AMPs, widely found in nature and obtained from microorganisms, have a broad range of antimicrobial protection, allowing them to be applied in the treatment of infections caused by various pathogenic microorganisms. Since these peptides are primarily cationic, they prefer anionic bacterial membranes due to electrostatic interactions. However, the applications of AMPs are currently limited owing to their hemolytic activity, poor bioavailability, degradation from proteolytic enzymes, and high-cost production. To overcome these limitations, nanotechnology has been used to improve AMP bioavailability, permeation across barriers, and/or protection against degradation. In addition, machine learning has been investigated due to its time-saving and cost-effective algorithms to predict AMPs. There are numerous databases available to train machine learning models. In this review, we focus on nanotechnology approaches for AMP delivery and advances in AMP design via machine learning. The AMP sources, classification, structures, antimicrobial mechanisms, their role in diseases, peptide engineering technologies, currently available databases, and machine learning techniques used to predict AMPs with minimal toxicity are discussed in detail.
Collapse
Affiliation(s)
- Alexa Sowers
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
- School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
25
|
Yang Y, Huang C, Hui L, Song Y, Fu Y, Li M, Yang H, Wu J, Sun J, Xu W, Wei L. Cathelicidins Target HSP60 To Restrict CVB3 Transmission via Disrupting the Exosome and Reducing Cardiomyocyte Apoptosis. J Virol 2023; 97:e0143322. [PMID: 36916989 PMCID: PMC10062171 DOI: 10.1128/jvi.01433-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
Cathelicidin antimicrobial peptides (mouse, CRAMP; human, LL-37) have broad-spectrum antiviral activities against enveloped viruses, but their mechanisms of action against nonenveloped viruses remain to be elucidated. Coxsackievirus B3 (CVB3), a member of nonenveloped virus belonging to the Enterovirus genus of Picornaviridae, is an important pathogen of viral myocarditis and dilated cardiomyopathy. Here, we observed that cardiac CRAMP expression was significantly upregulated in mice after CVB3 infection. The administration of CRAMP or LL-37 markedly suppressed CVB3 infection in mice, and CRAMP deficiency increased the susceptibility of mice to CVB3. CRAMP and LL-37 inhibited CVB3 replication in primary cardiomyocytes. However, they did not inactivate CVB3 particles and did not regulate the response of cardiomyocytes against CVB3 infection. Intriguingly, they inhibited CVB3 transmission through the exosome, but not virus receptor. In detail, CRAMP and LL-37 directly induced the lysis of exosomes by interfering with exosomal heat shock protein 60 (HSP60) and then blocked the diffusion of exosomes to recipient cells and inhibited the establishment of productive infection by exosomes. In addition, the interaction of CRAMP and LL-37 with HSP60 simultaneously inhibited HSP60-induced apoptosis in cardiomyocytes and reduced HSP60-enhanced CVB3 replication. Our findings reveal a novel mechanism of cathelicidins against viral infection and provide a new therapeutic strategy for CVB3-induced viral myocarditis. IMPORTANCE The relative mechanisms that cathelicidin antimicrobial peptides use to influence nonenveloped virus infection are unclear. We show here that cathelicidin antimicrobial peptides (CRAMP and LL-37) directly target exosomal HSP60 to destroy exosomes, which in turn block the diffusion of exosomes to recipient cardiomyocytes and reduced HSP60-induced apoptosis, thus restricting coxsackievirus B3 infection. Our results provide new insights into the mechanisms cathelicidin antimicrobial peptides use against viral infection.
Collapse
Affiliation(s)
- Yang Yang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Chunjing Huang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Li Hui
- The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yahui Song
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yuxuan Fu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
26
|
Hamza FN, Daher S, Fakhoury HMA, Grant WB, Kvietys PR, Al-Kattan K. Immunomodulatory Properties of Vitamin D in the Intestinal and Respiratory Systems. Nutrients 2023; 15:nu15071696. [PMID: 37049536 PMCID: PMC10097244 DOI: 10.3390/nu15071696] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Vitamin D plays a crucial role in modulating the innate immune response by interacting with its intracellular receptor, VDR. In this review, we address vitamin D/VDR signaling and how it contributes to the regulation of intestinal and respiratory microbiota. We additionally review some components of the innate immune system, such as the barrier function of the pulmonary and intestinal epithelial membranes and secretion of mucus, with their respective modulation by vitamin D. We also explore the mechanisms by which this vitamin D/VDR signaling mounts an antimicrobial response through the transduction of microbial signals and the production of antimicrobial peptides that constitute one of the body’s first lines of defense against pathogens. Additionally, we highlight the role of vitamin D in clinical diseases, namely inflammatory bowel disease and acute respiratory distress syndrome, where excessive inflammatory responses and dysbiosis are hallmarks. Increasing evidence suggests that vitamin D supplementation may have potentially beneficial effects on those diseases.
Collapse
Affiliation(s)
- Fatheia N. Hamza
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Sarah Daher
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Hana M. A. Fakhoury
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
- Correspondence:
| | - William B. Grant
- Sunlight, Nutrition, and Health Research Center, P.O. Box 641603, San Francisco, CA 94164-1603, USA
| | - Peter R. Kvietys
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Khaled Al-Kattan
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| |
Collapse
|
27
|
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
|
28
|
Honardoost M, Ghavideldarestani M, Khamseh ME. Role of vitamin D in pathogenesis and severity of COVID-19 infection. Arch Physiol Biochem 2023; 129:26-32. [PMID: 33125298 DOI: 10.1080/13813455.2020.1792505] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coronavirus disease (COVID-19) is an infectious disease caused by a new virus that causes respiratory illness. Older adults and individuals who have pre-existing chronic medical conditions are at higher risk for more serious complications from COVID-19. Hypovitaminosis D is attributed to the increased risk of lung injury and acute respiratory distress syndrome (ARDS) as well as diabetes, cardiovascular events and associated comorbidities, which are the main causes of severe clinical complications in COVID-19 patients. Considering the defensive role of vitamin D, mediated through modulation of the innate and adaptive immune system as well as inhibition of the Renin Angiotensin System (RAS), vitamin D supplementation might boost the immune system of COVID-19 patients and reduce severity of the disease in vitamin D deficient individuals.
Collapse
Affiliation(s)
- Maryam Honardoost
- Endocrine Research Center, Institute of Endocrinology and Metabolism, University of Medical Sciences, Tehran, Iran
- Cardio-Oncology Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Ghavideldarestani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, University of Medical Sciences, Tehran, Iran
| | - Mohammad E Khamseh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Charoenngam N, Jaroenlapnopparat A, Mettler SK, Grover A. Genetic Variations of the Vitamin D Metabolic Pathway and COVID-19 Susceptibility and Severity: Current Understanding and Existing Evidence. Biomedicines 2023; 11:400. [PMID: 36830936 PMCID: PMC9953304 DOI: 10.3390/biomedicines11020400] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The immunomodulatory and metabolic effects of vitamin D receptor (VDR) activation have been considered beneficial in mitigating the susceptibility and severity of COVID-19 infection. Furthermore, vitamin D-binding protein (DBP) has pleiotropic effects on the immune system that may influence inflammation associated with COVID-19. Multiple observational studies have demonstrated an association between low levels of serum 25-hydroxyvitamin D and risk and the severity of COVID-19 infection. However, the impact of vitamin D supplementation as an adjunctive treatment for COVID-19 based on evidence from randomized clinical trials is unclear. Equally important is that certain variations of the genes involved in the vitamin D metabolic pathway have been shown to affect immune function and linked with various clinical outcomes, including cardio-metabolic disorders, autoimmune diseases, infections, and cancers. This indicates inter-individual difference in body response to vitamin D. There is also emerging evidence that common polymorphisms of these genes may influence the susceptibility and severity of COVID-19, although the confidence of these findings is limited by a small number of studies and participants. Further studies are needed to address the potential role of VDR activation and DBP in the pathophysiology of COVID-19 which take into account the genetic variations of vitamin D metabolic pathway.
Collapse
Affiliation(s)
- Nipith Charoenngam
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA 02138, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 73170, Thailand
| | | | - Sofia K. Mettler
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA 02138, USA
| | - Ashna Grover
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA 02138, USA
| |
Collapse
|
30
|
Song Y, Wang Y, Yan S, Nakamura K, Kikukawa T, Ayabe T, Aizawa T. Efficient recombinant production of mouse-derived cryptdin family peptides by a novel facilitation strategy for inclusion body formation. Microb Cell Fact 2023; 22:9. [PMID: 36635697 PMCID: PMC9838031 DOI: 10.1186/s12934-023-02016-2] [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: 10/31/2022] [Accepted: 01/01/2023] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND A number of antimicrobial peptides (AMPs) hold promise as new drugs owing to their potent bactericidal activity and because they are often refractory to the development of drug resistance. Cryptdins (Crps) are a family of antimicrobial peptides found in the small intestine of mice, comprising six isoforms containing three sets of disulfide bonds. Although Crp4 is actively being investigated, there have been few studies to date on the other Crp isoforms. A prerequisite for detailed characterization of the other Crp isoforms is establishment of efficient sample preparation methods. RESULTS To avoid degradation during recombinant expression of Crps in E. coli, co-expression of Crps with the aggregation-prone protein human α-lactalbumin (HLA) was used to promote the formation of stable inclusion bodies. Using this method, the production of Crp4 and Crp6 by the BL21 strain was effective, but the expression of other Crp isoforms was not as efficient. The results of a cell-free system study suggested that Crps were degraded, even though a substantial amounts of Crps were synthesized. Therefore, using the Origami™ B strain, we were able to significantly increase the expression efficiency of Crps by promoting the formation of erroneous intermolecular disulfide bonds between HLA and Crps, thereby promoting protein aggregation and inclusion body formation, which prevented degradation. The various Crp isoforms were successfully refolded in vitro and purified using reversed-phase HPLC. In addition, the yield was further improved by deformylation of formyl-Crps. We measured the antibacterial activity of Crps against both Gram-positive and Gram-negative bacteria. Each Crp isoform exhibited a completely different trend in antimicrobial activity, although conformational analysis by circular dichroism did not reveal any significant steric differences. CONCLUSION In this study, we established a novel and efficient method for the production of the cryptdin family of cysteine-containing antimicrobial peptides. Additionally, we found that there were notable differences in the antibacterial activities of the various Crp family members. The expression system established in this study is expected to provide new insights regarding the mechanisms underlying the different antibacterial activities of the Crp family of peptides.
Collapse
Affiliation(s)
- Yuchi Song
- grid.39158.360000 0001 2173 7691Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| | - Yi Wang
- grid.39158.360000 0001 2173 7691Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| | - Shaonan Yan
- grid.39158.360000 0001 2173 7691Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| | - Kiminori Nakamura
- grid.39158.360000 0001 2173 7691Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| | - Takashi Kikukawa
- grid.39158.360000 0001 2173 7691Laboratory of Biological Information Analysis Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| | - Tokiyoshi Ayabe
- grid.39158.360000 0001 2173 7691Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| | - Tomoyasu Aizawa
- grid.39158.360000 0001 2173 7691Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido Japan
| |
Collapse
|
31
|
Park S, Cho NJ. Lipid Membrane Interface Viewpoint: From Viral Entry to Antiviral and Vaccine Development. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1-11. [PMID: 36576966 DOI: 10.1021/acs.langmuir.2c02501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Membrane-enveloped viruses are responsible for most viral pandemics in history, and more effort is needed to advance broadly applicable countermeasures to mitigate the impact of future outbreaks. In this Perspective, we discuss how biosensing techniques associated with lipid model membrane platforms are contributing to improving our mechanistic knowledge of membrane fusion and destabilization that is closely linked to viral entry as well as vaccine and antiviral drug development. A key benefit of these platforms is the simplicity of interpreting the results which can be complemented by other techniques to decipher more complicated biological observations and evaluate the biophysical functionalities that can be correlated to biological activities. Then, we introduce exciting application examples of membrane-targeting antivirals that have been refined over time and will continue to improve based on biophysical insights. Two ways to abrogate the function of viral membranes are introduced here: (1) selective disruption of the viral membrane structure and (2) alteration of the membrane component. While both methods are suitable for broadly useful antivirals, the latter also has the potential to produce an inactivated vaccine. Collectively, we emphasize how biosensing tools based on membrane interfacial science can provide valuable information that could be translated into biomedicines and improve their selectivity and performance.
Collapse
Affiliation(s)
- Soohyun Park
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
32
|
Agamennone M, Fantacuzzi M, Vivenzio G, Scala MC, Campiglia P, Superti F, Sala M. Antiviral Peptides as Anti-Influenza Agents. Int J Mol Sci 2022; 23:11433. [PMID: 36232735 PMCID: PMC9569631 DOI: 10.3390/ijms231911433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Influenza viruses represent a leading cause of high morbidity and mortality worldwide. Approaches for fighting flu are seasonal vaccines and some antiviral drugs. The development of the seasonal flu vaccine requires a great deal of effort, as careful studies are needed to select the strains to be included in each year's vaccine. Antiviral drugs available against Influenza virus infections have certain limitations due to the increased resistance rate and negative side effects. The highly mutative nature of these viruses leads to the emergence of new antigenic variants, against which the urgent development of new approaches for antiviral therapy is needed. Among these approaches, one of the emerging new fields of "peptide-based therapies" against Influenza viruses is being explored and looks promising. This review describes the recent findings on the antiviral activity, mechanism of action and therapeutic capability of antiviral peptides that bind HA, NA, PB1, and M2 as a means of countering Influenza virus infection.
Collapse
Affiliation(s)
- Mariangela Agamennone
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Marialuigia Fantacuzzi
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Giovanni Vivenzio
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Maria Carmina Scala
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Fabiana Superti
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marina Sala
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| |
Collapse
|
33
|
Sîrbe C, Rednic S, Grama A, Pop TL. An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases. Int J Mol Sci 2022; 23:9784. [PMID: 36077185 PMCID: PMC9456003 DOI: 10.3390/ijms23179784] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 12/16/2022] Open
Abstract
Vitamin D intervenes in calcium and phosphate metabolism and bone homeostasis. Experimental studies have shown that 1,25-dihydroxyvitamin D (calcitriol) generates immunologic activities on the innate and adaptive immune system and endothelial membrane stability. Low levels of serum 25-hydroxyvitamin D (25(OH)D) are associated with an increased risk of developing immune-related diseases such as psoriasis, type 1 diabetes, multiple sclerosis, and autoimmune diseases. Various clinical trials describe the efficacy of supplementation of vitamin D and its metabolites for treating these diseases that result in variable outcomes. Different disease outcomes are observed in treatment with vitamin D as high inter-individual difference is present with complex gene expression in human peripheral blood mononuclear cells. However, it is still not fully known what level of serum 25(OH)D is needed. The current recommendation is to increase vitamin D intake and have enough sunlight exposure to have serum 25(OH)D at a level of 30 ng/mL (75 nmol/L) and better at 40-60 ng/mL (100-150 nmol/L) to obtain the optimal health benefits of vitamin D.
Collapse
Affiliation(s)
- Claudia Sîrbe
- 2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Simona Rednic
- Rheumatology Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania
- Rheumatology Discipline, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alina Grama
- 2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Tudor Lucian Pop
- 2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| |
Collapse
|
34
|
de Tapia L, García-Fojeda B, Kronqvist N, Johansson J, Casals C. The collectin SP-A and its trimeric recombinant fragment protect alveolar epithelial cells from the cytotoxic and proinflammatory effects of human cathelicidin in vitro. Front Immunol 2022; 13:994328. [PMID: 36105805 PMCID: PMC9464622 DOI: 10.3389/fimmu.2022.994328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/08/2022] [Indexed: 01/19/2023] Open
Abstract
Human cathelicidin (LL-37) is a defense peptide with antimicrobial activity against various pathogens. However, LL-37 can also trigger tissue injury by binding to host cell membranes. The cytotoxic effects of LL-37 may be especially relevant in chronic respiratory diseases characterized by increased LL-37. The aim of this study was to investigate whether the human collectin SP-A and a trimeric recombinant fragment thereof (rfhSP-A) can regulate the activities of LL-37. To this end, we studied the interaction of LL-37 with SP-A and rfhSP-A by intrinsic fluorescence, dynamic light scattering, and circular dichroism, as well as the effects of these proteins on the antimicrobial and cytotoxic activities of LL-37. Both SP-A and rfhSP-A bound LL-37 with high affinity at physiological ionic strength (KD = 0.45 ± 0.01 nM for SP-A and 1.22 ± 0.7 nM for rfhSP-A). Such interactions result in the reduction of LL-37-induced cell permeability and IL-8 release in human pneumocytes, mediated by P2X7 channels. Binding of LL-37 to SP-A did not modify the properties of SP-A or the antibacterial activity of LL-37 against respiratory pathogens (Klebsiella pneumoniae, Pseudomonas aeruginosa, and nontypeable Haemophilus influenzae). SP-A/LL-37 complexes showed a greater ability to aggregate LPS vesicles than LL-37, which reduces endotoxin bioactivity. These results reveal the protective role of native SP-A in controlling LL-37 activities and suggest a potential therapeutic effect of rfhSP-A in reducing the cytotoxic and inflammatory actions of LL-37, without affecting its microbicidal activity against Gram-negative pathogens.
Collapse
Affiliation(s)
- Lidia de Tapia
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Belén García-Fojeda
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Nina Kronqvist
- Department of Biosciences and Nutrition, Neo, Karolinska Institutet, Huddinge, Sweden
| | - Jan Johansson
- Department of Biosciences and Nutrition, Neo, Karolinska Institutet, Huddinge, Sweden
| | - Cristina Casals
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
35
|
Monroe MK, Wang H, Anderson CF, Jia H, Flexner C, Cui H. Leveraging the therapeutic, biological, and self-assembling potential of peptides for the treatment of viral infections. J Control Release 2022; 348:1028-1049. [PMID: 35752254 PMCID: PMC11022941 DOI: 10.1016/j.jconrel.2022.06.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022]
Abstract
Peptides and peptide-based materials have an increasing role in the treatment of viral infections through their use as active pharmaceutical ingredients, targeting moieties, excipients, carriers, or structural components in drug delivery systems. The discovery of peptide-based therapeutic compounds, coupled with the development of new stabilization and formulation strategies, has led to a resurgence of antiviral peptide therapeutics over the past two decades. The ability of peptides to bind cell receptors and to facilitate membrane penetration and subsequent intracellular trafficking enables their use in various antiviral systems for improved targeting efficiency and treatment efficacy. Importantly, the self-assembly of peptides into well-defined nanostructures provides a vast library of discrete constructs and supramolecular biomaterials for systemic and local delivery of antiviral agents. We review here the recent progress in exploiting the therapeutic, biological, and self-assembling potential of peptides, peptide conjugates, and their supramolecular assemblies in treating human viral infections, with an emphasis on the treatment strategies for Human Immunodeficiency Virus (HIV).
Collapse
Affiliation(s)
- Maya K Monroe
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Caleb F Anderson
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Hongpeng Jia
- Department of Surgery, The Johns Hopkins University School of Medicine, United States of America
| | - Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, The Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD 21205, United States of America.
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Deptartment of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America; Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, United States of America.
| |
Collapse
|
36
|
Arora J, Patel DR, Nicol MJ, Field CJ, Restori KH, Wang J, Froelich NE, Katkere B, Terwilliger JA, Weaver V, Luley E, Kelly K, Kirimanjeswara GS, Sutton TC, Cantorna MT. Vitamin D and the Ability to Produce 1,25(OH) 2D Are Critical for Protection from Viral Infection of the Lungs. Nutrients 2022; 14:3061. [PMID: 35893921 PMCID: PMC9332570 DOI: 10.3390/nu14153061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
Vitamin D supplementation is linked to improved outcomes from respiratory virus infection, and the COVID-19 pandemic renewed interest in understanding the potential role of vitamin D in protecting the lung from viral infections. Therefore, we evaluated the role of vitamin D using animal models of pandemic H1N1 influenza and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. In mice, dietary-induced vitamin D deficiency resulted in lung inflammation that was present prior to infection. Vitamin D sufficient (D+) and deficient (D-) wildtype (WT) and D+ and D- Cyp27B1 (Cyp) knockout (KO, cannot produce 1,25(OH)2D) mice were infected with pandemic H1N1. D- WT, D+ Cyp KO, and D- Cyp KO mice all exhibited significantly reduced survival compared to D+ WT mice. Importantly, survival was not the result of reduced viral replication, as influenza M gene expression in the lungs was similar for all animals. Based on these findings, additional experiments were performed using the mouse and hamster models of SARS-CoV-2 infection. In these studies, high dose vitamin D supplementation reduced lung inflammation in mice but not hamsters. A trend to faster weight recovery was observed in 1,25(OH)2D treated mice that survived SARS-CoV-2 infection. There was no effect of vitamin D on SARS-CoV-2 N gene expression in the lung of either mice or hamsters. Therefore, vitamin D deficiency enhanced disease severity, while vitamin D sufficiency/supplementation reduced inflammation following infections with H1N1 influenza and SARS-CoV-2.
Collapse
Affiliation(s)
- Juhi Arora
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Devanshi R. Patel
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - McKayla J. Nicol
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Cassandra J. Field
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Katherine H. Restori
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Jinpeng Wang
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Nicole E. Froelich
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Bhuvana Katkere
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Josey A. Terwilliger
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Veronika Weaver
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Erin Luley
- Animal Diagnostic Laboratory, The Pennsylvania State University, University Park, PA 16802, USA; (E.L.); (K.K.)
| | - Kathleen Kelly
- Animal Diagnostic Laboratory, The Pennsylvania State University, University Park, PA 16802, USA; (E.L.); (K.K.)
| | - Girish S. Kirimanjeswara
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Troy C. Sutton
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| | - Margherita T. Cantorna
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (J.A.); (D.R.P.); (M.J.N.); (C.J.F.); (K.H.R.); (J.W.); (N.E.F.); (B.K.); (J.A.T.); (V.W.); (G.S.K.)
| |
Collapse
|
37
|
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
|
38
|
Duan Z, Zhang J, Chen X, Liu M, Zhao H, Jin L, Zhang Z, Luan N, Meng P, Wang J, Tan Z, Li Y, Deng G, Lai R. Role of LL-37 in thrombotic complications in patients with COVID-19. Cell Mol Life Sci 2022; 79:309. [PMID: 35596804 PMCID: PMC9123294 DOI: 10.1007/s00018-022-04309-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/28/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Blood clot formation induced by dysfunctional coagulation is a frequent complication of coronavirus disease 2019 (COVID-19) and a high-risk factor for severe illness and death. Neutrophil extracellular traps (NETs) are implicated in COVID-19-induced immunothrombosis. Furthermore, human cathelicidin, a NET component, can perturb the interaction between the SARS-CoV-2 spike protein and its ACE2 receptor, which mediates viral entry into cells. At present, however, the levels of cathelicidin antimicrobial peptides after SARS-CoV-2 infection and their role in COVID-19 thrombosis formation remain unclear. In the current study, we analyzed coagulation function and found a decrease in thrombin time but an increase in fibrinogen level, prothrombin time, and activated partial thromboplastin time in COVID-19 patients. In addition, the cathelicidin antimicrobial peptide LL-37 was upregulated by the spike protein and significantly elevated in the plasma of patients. Furthermore, LL-37 levels were negatively correlated with thrombin time but positively correlated with fibrinogen level. In addition to platelet activation, cathelicidin peptides enhanced the activity of coagulation factors, such as factor Xa (FXa) and thrombin, which may induce hypercoagulation in diseases with high cathelicidin peptide levels. Injection of cathelicidin peptides promoted the formation of thrombosis, whereas deletion of cathelicidin inhibited thrombosis in vivo. These results suggest that cathelicidin antimicrobial peptide LL-37 is elevated during SARS-CoV-2 infection, which may induce hypercoagulation in COVID-19 patients by activating coagulation factors.
Collapse
Affiliation(s)
- Zilei Duan
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Juan Zhang
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Xue Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Ming Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Hongwen Zhao
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Lin Jin
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Zhiye Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Ning Luan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Ping Meng
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650041, Yunnan, China
| | - Jing Wang
- Department of Laboratory Diagnosis, Chongqing Public Health Medical Center, Public Health Hospital of Southwest University, 109 Baoyu Rd. Shapingba, Chongqing, 400038, China
| | - Zhaoxia Tan
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Yaxiong Li
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650041, Yunnan, China.
| | - Guohong Deng
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China.
| | - Ren Lai
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China.
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China.
| |
Collapse
|
39
|
Lee YCJ, Shirkey JD, Park J, Bisht K, Cowan AJ. An Overview of Antiviral Peptides and Rational Biodesign Considerations. BIODESIGN RESEARCH 2022; 2022:9898241. [PMID: 37850133 PMCID: PMC10521750 DOI: 10.34133/2022/9898241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 10/19/2023] Open
Abstract
Viral diseases have contributed significantly to worldwide morbidity and mortality throughout history. Despite the existence of therapeutic treatments for many viral infections, antiviral resistance and the threat posed by novel viruses highlight the need for an increased number of effective therapeutics. In addition to small molecule drugs and biologics, antimicrobial peptides (AMPs) represent an emerging class of potential antiviral therapeutics. While AMPs have traditionally been regarded in the context of their antibacterial activities, many AMPs are now known to be antiviral. These antiviral peptides (AVPs) have been shown to target and perturb viral membrane envelopes and inhibit various stages of the viral life cycle, from preattachment inhibition through viral release from infected host cells. Rational design of AMPs has also proven effective in identifying highly active and specific peptides and can aid in the discovery of lead peptides with high therapeutic selectivity. In this review, we highlight AVPs with strong antiviral activity largely curated from a publicly available AMP database. We then compile the sequences present in our AVP database to generate structural predictions of generic AVP motifs. Finally, we cover the rational design approaches available for AVPs taking into account approaches currently used for the rational design of AMPs.
Collapse
Affiliation(s)
- Ying-Chiang J. Lee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jaden D. Shirkey
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jongbeom Park
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Karishma Bisht
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Alexis J. Cowan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
40
|
Aloul KM, Nielsen JE, Defensor EB, Lin JS, Fortkort JA, Shamloo M, Cirillo JD, Gombart AF, Barron AE. Upregulating Human Cathelicidin Antimicrobial Peptide LL-37 Expression May Prevent Severe COVID-19 Inflammatory Responses and Reduce Microthrombosis. Front Immunol 2022; 13:880961. [PMID: 35634307 PMCID: PMC9134243 DOI: 10.3389/fimmu.2022.880961] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/11/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is characterized by hyperactivation by inflammatory cytokines and recruitment of macrophages, neutrophils, and other immune cells, all hallmarks of a strong inflammatory response that can lead to severe complications and multi-organ damage. Mortality in COVID-19 patients is associated with a high prevalence of neutrophil extracellular trap (NET) formation and microthrombosis that are exacerbated by hyperglycemia, diabetes, and old age. SARS-CoV-2 infection in humans and non-human primates have revealed long-term neurological consequences of COVID-19, possibly concomitant with the formation of Lewy bodies in the brain and invasion of the nervous system via the olfactory bulb. In this paper, we review the relevance of the human cathelicidin LL-37 in SARS-CoV-2 infections. LL-37 is an immunomodulatory, host defense peptide with direct anti-SARS-CoV-2 activity, and pleiotropic effects on the inflammatory response, neovascularization, Lewy body formation, and pancreatic islet cell function. The bioactive form of vitamin D and a number of other compounds induce LL-37 expression and one might predict its upregulation, could reduce the prevalence of severe COVID-19. We hypothesize upregulation of LL-37 will act therapeutically, facilitating efficient NET clearance by macrophages, speeding endothelial repair after inflammatory tissue damage, preventing α-synuclein aggregation, and supporting blood-glucose level stabilization by facilitating insulin release and islet β-cell neogenesis. In addition, it has been postulated that LL-37 can directly bind the S1 domain of SARS-CoV-2, mask angiotensin converting enzyme 2 (ACE2) receptors, and limit SARS-CoV-2 infection. Purposeful upregulation of LL-37 could also serve as a preventative and therapeutic strategy for SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Karim M. Aloul
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| | - Josefine Eilsø Nielsen
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Erwin B. Defensor
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jennifer S. Lin
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| | - John A. Fortkort
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| | - Mehrdad Shamloo
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M College of Medicine, Bryan, TX, United States
| | - Adrian F. Gombart
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, United States
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Annelise E. Barron
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
41
|
Castillo JA, Giraldo DM, Smit JM, Rodenhuis-Zybert IA, Urcuqui-Inchima S. Vitamin D-induced LL-37 modulates innate immune responses of human primary macrophages during DENV-2 infection. Pathog Dis 2022; 80:6581314. [PMID: 35512569 DOI: 10.1093/femspd/ftac014] [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: 01/17/2022] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemics of dengue, an acute and potentially severe disease caused by mosquito-borne dengue virus (DENV), pose a major challenge to clinicians and health care services across the sub(tropics). Severe disease onset is associated with a dysregulated inflammatory response to the virus and there are currently no drugs to alleviate disease symptoms. LL-37 is a potent antimicrobial peptide with a wide range of immunoregulatory properties. In this study, we assessed the effect of LL-37 on DENV-2-induced responses in human monocyte-derived macrophages (MDMs). We show that simultaneous exposure of exogenous LL-37 and DENV-2 resulted in reduced replication of the virus in MDMs, while the addition of LL-37 post-exposure to DENV-2 did not. Interestingly, the latter condition reduced the production of IL-6 and increased the expression of genes involved in virus sensing and antiviral response. Finally, we demonstrate that low endogenous levels and limited production of LL-37 in MDMs in response to DENV-2 infection can be increased by differentiating MDMs in the presence of Vitamin D (VitD3). Taken together, this study demonstrates that in addition to its antimicrobial properties, LL-37 has immunomodulatory properties in the curse of DENV infection and its production can be increased by VitD3.
Collapse
Affiliation(s)
- Jorge Andrés Castillo
- Grupo Inmunovirología. Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia.,Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Diana Marcela Giraldo
- Grupo Inmunovirología. Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia
| | - Jolanda M Smit
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Izabela A Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología. Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia
| |
Collapse
|
42
|
Jeong H, Shin H, Hong S, Kim Y. Physiological Roles of Monomeric Amyloid-β and Implications for Alzheimer's Disease Therapeutics. Exp Neurobiol 2022; 31:65-88. [PMID: 35673997 PMCID: PMC9194638 DOI: 10.5607/en22004] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) progressively inflicts impairment of synaptic functions with notable deposition of amyloid-β (Aβ) as senile plaques within the extracellular space of the brain. Accordingly, therapeutic directions for AD have focused on clearing Aβ plaques or preventing amyloidogenesis based on the amyloid cascade hypothesis. However, the emerging evidence suggests that Aβ serves biological roles, which include suppressing microbial infections, regulating synaptic plasticity, promoting recovery after brain injury, sealing leaks in the blood-brain barrier, and possibly inhibiting the proliferation of cancer cells. More importantly, these functions were found in in vitro and in vivo investigations in a hormetic manner, that is to be neuroprotective at low concentrations and pathological at high concentrations. We herein summarize the physiological roles of monomeric Aβ and current Aβ-directed therapies in clinical trials. Based on the evidence, we propose that novel therapeutics targeting Aβ should selectively target Aβ in neurotoxic forms such as oligomers while retaining monomeric Aβ in order to preserve the physiological functions of Aβ monomers.
Collapse
Affiliation(s)
- Hyomin Jeong
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Heewon Shin
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Seungpyo Hong
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - YoungSoo Kim
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
43
|
IL-26 inhibits hepatitis C virus replication in hepatocytes. J Hepatol 2022; 76:822-831. [PMID: 34952035 DOI: 10.1016/j.jhep.2021.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 11/15/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Interleukin-26 (IL-26) is a proinflammatory cytokine that has properties atypical for a cytokine, such as direct antibacterial activity and DNA-binding capacity. We previously observed an accumulation of IL-26 in fibrotic and inflammatory lesions in the livers of patients with chronic HCV infection and showed that infiltrating CD3+ lymphocytes were the principal source of IL-26. Surprisingly, IL-26 was also detected in the cytoplasm of hepatocytes from HCV-infected patients, even though these cells do not produce IL-26, even when infected with HCV. Based on this observation and possible interactions between IL-26 and nucleic acids, we investigated the possibility that IL-26 controlled HCV infection independently of the immune system. METHODS We evaluated the ability of IL-26 to interfere with HCV replication in hepatocytes and investigated the mechanisms by which IL-26 exerts its antiviral activity. RESULTS We showed that IL-26 penetrated HCV-infected hepatocytes, where it interacted directly with HCV double-stranded RNA replication intermediates, thereby inhibiting viral replication. IL-26 interfered with viral RNA-dependent RNA polymerase activity, preventing the de novo synthesis of viral genomic single-stranded RNA. CONCLUSIONS These findings reveal a new role for IL-26 in direct protection against HCV infection, independently of the immune system, and increase our understanding of the antiviral defense mechanisms controlling HCV infection. Future studies should evaluate the possible use of IL-26 for treating other chronic disorders caused by RNA viruses, for which few treatments are currently available, or emerging RNA viruses. LAY SUMMARY This study sheds new light on the body's arsenal for controlling hepatitis C virus (HCV) infection and identifies interleukin-26 (IL-26) as an antiviral molecule capable of blocking HCV replication. IL-26, which has unique biochemical and structural characteristics, penetrates infected hepatocytes and interacts directly with viral RNA, thereby blocking viral replication. IL-26 is, therefore, a new player in antiviral defenses, operating independently of the immune system. It is of considerable potential interest for treating HCV infection and other chronic disorders caused by RNA viruses for which few treatments are currently available, and for combating emerging RNA viruses.
Collapse
|
44
|
Abstract
Vitamin D, best known for its role in skeletal health, has emerged as a key regulator of innate immune responses to microbial threat. In immune cells such as macrophages, expression of CYP27B1, the 25-hydroxyvitamin D 1α-hydroxylase, is induced by immune-specific inputs, leading to local production of hormonal 1,25-dihydroxyvitamin D (1,25D) at sites of infection, which in turn directly induces the expression of genes encoding antimicrobial peptides. Vitamin D signaling is active upstream and downstream of pattern recognition receptors, which promote front-line innate immune responses. Moreover, 1,25D stimulates autophagy, which has emerged as a mechanism critical for control of intracellular pathogens such as M. tuberculosis. Strong laboratory and epidemiological evidence links vitamin D deficiency to increased rates of conditions such as dental caries, as well as inflammatory bowel diseases arising from dysregulation of innate immune handling intestinal flora. 1,25D is also active in signaling cascades that promote antiviral innate immunity; 1,25D-induced expression of the antimicrobial peptide CAMP/LL37, originally characterized for its antibacterial properties, is a key component of antiviral responses. Poor vitamin D status is associated with greater susceptibility to viral infections, including those of the respiratory tract. Although the severity of the COVID-19 pandemic has been alleviated in some areas by the arrival of vaccines, it remains important to identify therapeutic interventions that reduce disease severity and mortality, and accelerate recovery. This review outlines of our current knowledge of the mechanisms of action of vitamin D signaling in the innate immune system. It also provides an assessment of the therapeutic potential of vitamin D supplementation in infectious diseases, including an up-to-date analysis of the putative benefits of vitamin D supplementation in the ongoing COVID-19 crisis.
Collapse
Affiliation(s)
- Aiten Ismailova
- Departments of Physiology, McGill University, Montreal, Qc, Canada
| | - John H White
- Departments of Physiology, McGill University, Montreal, Qc, Canada.
- Departments of Medicine, McGill University, Montreal, Qc, Canada.
| |
Collapse
|
45
|
Maleki Dizaj S, Salatin S, Khezri K, Lee JY, Lotfipour F. Targeting Multidrug Resistance With Antimicrobial Peptide-Decorated Nanoparticles and Polymers. Front Microbiol 2022; 13:831655. [PMID: 35432230 PMCID: PMC9009044 DOI: 10.3389/fmicb.2022.831655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/09/2022] [Indexed: 01/21/2023] Open
Abstract
As a category of small peptides frequently found in nature, antimicrobial peptides (AMPs) constitute a major part of the innate immune system of various organisms. Antimicrobial peptides feature various inhibitory effects against fungi, bacteria, viruses, and parasites. Due to the increasing concerns of antibiotic resistance among microorganisms, development of antimicrobial peptides is an emerging tool as a favorable applicability prospect in food, medicine, aquaculture, animal husbandry, and agriculture. This review presents the latest research progress made in the field of antimicrobial peptides, such as their mechanism of action, classification, application status, design techniques, and a review on decoration of nanoparticles and polymers with AMPs that are used in treating multidrug resistance. Lastly, we will highlight recent progress in antiviral peptides to treat emerging viral diseases (e.g., anti-coronavirus peptides) and discuss the outlook of AMP applications.
Collapse
Affiliation(s)
- Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Dental Biomaterials, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Salatin
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Khezri
- Deputy of Food and Drug Administration, Urmia University of Medical Sciences, Urmia, Iran
| | - Jyh-Yeuan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Farzaneh Lotfipour
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Food and Drug Safety Research Center, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
46
|
Albergamo A, Apprato G, Silvagno F. The Role of Vitamin D in Supporting Health in the COVID-19 Era. Int J Mol Sci 2022; 23:3621. [PMID: 35408981 PMCID: PMC8998275 DOI: 10.3390/ijms23073621] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
The genomic activity of vitamin D is associated with metabolic effects, and the hormone has a strong impact on several physiological functions and, therefore, on health. Among its renowned functions, vitamin D is an immunomodulator and a molecule with an anti-inflammatory effect, and, recently, it has been much studied in relation to its response against viral infections, especially against COVID-19. This review aims to take stock of the correlation studies between vitamin D deficiency and increased risks of severe COVID-19 disease and, similarly, between vitamin D deficiency and acute respiratory distress syndrome. Based on this evidence, supplementation with vitamin D has been tested in clinical trials, and the results are discussed. Finally, this study includes a biochemical analysis on the effects of vitamin D in the body's defense mechanisms against viral infection. In particular, the antioxidant and anti-inflammatory functions are considered in relation to energy metabolism, and the potential, beneficial effect of vitamin D in COVID-19 is described, with discussion of its influence on different biochemical pathways. The proposed, broader view of vitamin D activity could support a better-integrated approach in supplementation strategies against severe COVID-19, which could be valuable in a near future of living with an infection becoming endemic.
Collapse
Affiliation(s)
- Alice Albergamo
- Department of Oncology, University of Torino, 10126 Torino, Italy
| | - Giulia Apprato
- Department of Oncology, University of Torino, 10126 Torino, Italy
| | | |
Collapse
|
47
|
Influence of exercise and vitamin D on the immune system against Covid-19: an integrative review of current literature. Mol Cell Biochem 2022; 477:1725-1737. [PMID: 35258807 PMCID: PMC8902492 DOI: 10.1007/s11010-022-04402-7] [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: 08/25/2021] [Accepted: 02/23/2022] [Indexed: 10/26/2022]
Abstract
Respiratory infections of viral origin have become the leading cause of infectious diseases in the world. In 2020, the World Health Organization (WHO) declared a pandemic due to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), Coronavirus Disease 2019 (Covid-19). The pandemic caused by the new coronavirus has challenged the entire global health system, since Covid-19 has a high rate of morbidity and mortality. The immune response to the virus depends on factors such as age, genetics, nutritional status, physical status, as well as environmental factors. Despite scientific advances, so far, there is still no specific therapy for the disease. Thus, this study aims to analyze the contribution of physical exercise and maintenance and/or supplementation of vitamin D to the strengthening of the immune system against viral infections, among them, Covid-19. Regular practice of moderate-intensity physical activity is responsible for promoting a reduction in the concentrations of pro-inflammatory cytokines (IL-6, TNF-α and IL-1β), as well as triggering the increase in the production of anti-inflammatory cytokines (IL-4 and IL-10). In addition, hypovitaminosis D predisposes to the development of chronic diseases and infections. Therefore, in patients affected by Covid-19, the maintenance of vitamin D levels contributes significantly to the 0prevention of the cytokine storm. Thus, the association between maintaining vitamin D levels and performing moderate-intensity physical exercise is responsible for strengthening the immune system and, therefore, triggering a defense mechanism against infections by intracellular microorganisms, in which SARS -CoV-2.
Collapse
|
48
|
Aronen M, Viikari L, Langen H, Kohonen I, Wuorela M, Vuorinen T, Söderlund-Venermo M, Viitanen M, Camargo CA, Vahlberg T, Jartti T. The long-term prognostic value of serum 25(OH)D, albumin, and LL-37 levels in acute respiratory diseases among older adults. BMC Geriatr 2022; 22:146. [PMID: 35189828 PMCID: PMC8860370 DOI: 10.1186/s12877-022-02836-8] [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: 07/01/2021] [Accepted: 02/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background Older adults are more susceptible to respiratory tract infection than healthy working age adults. The increased susceptibility of older adults is thought to be interlinked with vitamin D status, nourishment, and immunological state in general. Data are scarce whether these parameters could serve as prognostic markers. Aim To study whether serum 25(OH)D, albumin, and LL-37 level could give prognostic value of long-term survival in the older adults with multimorbidity and acute respiratory infection. Methods Consecutive episodes of hospital care of patients 65 years and older with respiratory symptoms were prospectively studied as a cohort. Standard clinical questionnaire was filled by the study physician. Laboratory markers included serum levels of 25(OH)D, albumin and LL-37, C-reactive protein (CRP), white blood cell count (WBC) and polymerase chain reaction diagnostics for 14 respiratory viruses. Pneumonia was confirmed by chest radiographs. Respiratory illness severity, death at ward, length of hospital stays, and 5-year survival were used as outcomes. Results In total, 289 older adult patients with mean age of 83 years were included in the study. Serum 25(OH)D deficiency (< 50 nmol/liter) was present in 59% and hypoalbuminemia (< 3.5 g/dL) in 55% of the study patients. Low serum albumin level was associated to one, two- and five-year mortality after hospital stay (all P < .05). In addition, it was associated with pneumonia, dyspnea, over 13-night long stay at ward and death at ward (all P < .05). No associations were seen between serum 25(OH)D and LL-37 levels and disease severity, short-term clinical outcome, or long-term survival. Associations between serum 25(OH)D, albumin, and LL-37 levels and respiratory virus presence were not seen. Conclusions Serum albumin level on admission seems to give valuable information about the patients’ general health and recovery potential in treating older adults with respiratory symptoms. Serum 25(OH)D and LL-37 had no associations with disease severity or long- and short-term prognosis among older adults hospitalized with respiratory symptoms. Supplementary Information The online version contains supplementary material available at 10.1186/s12877-022-02836-8.
Collapse
Affiliation(s)
- Matti Aronen
- Department of Geriatrics, Turku University Hospital, Turku, Finland. .,, Uikunkuja 7, N28100, Pori, Finland.
| | | | - Henriikka Langen
- Department of Geriatrics, Turku University Hospital, Turku, Finland
| | - Ia Kohonen
- Department of Radiology, Turku University Hospital, Turku, Finland
| | | | - Tytti Vuorinen
- Department of Medical Microbiology, Turku University Hospital and Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | | | - Tero Vahlberg
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Tuomas Jartti
- PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu, Finland.,Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland.,Department of Pediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
49
|
Xin Y, Chen S, Tang K, Wu Y, Guo Y. Identification of Nifurtimox and Chrysin as Anti-Influenza Virus Agents by Clinical Transcriptome Signature Reversion. Int J Mol Sci 2022; 23:ijms23042372. [PMID: 35216485 PMCID: PMC8876279 DOI: 10.3390/ijms23042372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022] Open
Abstract
The rapid development in the field of transcriptomics provides remarkable biomedical insights for drug discovery. In this study, a transcriptome signature reversal approach was conducted to identify the agents against influenza A virus (IAV) infection through dissecting gene expression changes in response to disease or compounds’ perturbations. Two compounds, nifurtimox and chrysin, were identified by a modified Kolmogorov–Smirnov test statistic based on the transcriptional signatures from 81 IAV-infected patients and the gene expression profiles of 1309 compounds. Their activities were verified in vitro with half maximal effective concentrations (EC50s) from 9.1 to 19.1 μM against H1N1 or H3N2. It also suggested that the two compounds interfered with multiple sessions in IAV infection by reversing the expression of 28 IAV informative genes. Through network-based analysis of the 28 reversed IAV informative genes, a strong synergistic effect of the two compounds was revealed, which was confirmed in vitro. By using the transcriptome signature reversion (TSR) on clinical datasets, this study provides an efficient scheme for the discovery of drugs targeting multiple host factors regarding clinical signs and symptoms, which may also confer an opportunity for decelerating drug-resistant variant emergence.
Collapse
Affiliation(s)
- Yijing Xin
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shubing Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ke Tang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - You Wu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Guo
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: ; Tel.: +86-010-63161716
| |
Collapse
|
50
|
Mesenchymal stem cell-based treatments for COVID-19: status and future perspectives for clinical applications. Cell Mol Life Sci 2022; 79:142. [PMID: 35187617 PMCID: PMC8858603 DOI: 10.1007/s00018-021-04096-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 01/08/2023]
Abstract
As a result of cross-species transmission in December 2019, the coronavirus disease 2019 (COVID-19) became a serious endangerment to human health and the causal agent of a global pandemic. Although the number of infected people has decreased due to effective management, novel methods to treat critical COVID-19 patients are still urgently required. This review describes the origins, pathogenesis, and clinical features of COVID-19 and the potential uses of mesenchymal stem cells (MSCs) in therapeutic treatments for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients. MSCs have previously been shown to have positive effects in the treatment of lung diseases, such as acute lung injury, idiopathic pulmonary fibrosis, acute respiratory distress syndrome, lung cancer, asthma, and chronic obstructive pulmonary disease. MSC mechanisms of action involve differentiation potentials, immune regulation, secretion of anti-inflammatory factors, migration and homing, anti-apoptotic properties, antiviral effects, and extracellular vesicles. Currently, 74 clinical trials are investigating the use of MSCs (predominately from the umbilical cord, bone marrow, and adipose tissue) to treat COVID-19. Although most of these trials are still in their early stages, the preliminary data are promising. However, long-term safety evaluations are still lacking, and large-scale and controlled trials are required for more conclusive judgments regarding MSC-based therapies. The main challenges and prospective directions for the use of MSCs in clinical applications are discussed herein. In summary, while the clinical use of MSCs to treat COVID-19 is still in the preliminary stages of investigation, promising results indicate that they could potentially be utilized in future treatments.
Collapse
|