1
|
Engin MMN, Özdemir Ö. Role of vitamin D in COVID-19 and other viral infections. World J Virol 2024; 13:95349. [PMID: 39323448 PMCID: PMC11401007 DOI: 10.5501/wjv.v13.i3.95349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
Vitamin D is a steroid hormone that is naturally produced in the body or obtained through dietary sources, primarily under the influence of UVB radiation. This essential nutrient has a vital role in numerous physiological processes, encompassing immune function, cell growth, differentiation, insulin regulation, and cardiovascular well-being, along with its pivotal role in sustaining the delicate equilibrium of calcium and phosphate concentrations in the body. Moreover, vitamin D reinforces mucosal defense and bolsters the immune system through immunomodulation, making it a critical component of overall health. Numerous studies have unveiled the profound connection between vitamin D and the predisposition to respiratory tract infections, including well-known viruses such as influenza and the novel severe acute respiratory syndrome coronavirus 2. Vitamin D deficiency has been consistently linked to increased severity of coronavirus disease 2019 (COVID-19) and a heightened risk of mortality among afflicted individuals. Retrospective observational studies have further substantiated these findings, indicating that levels of vitamin D are linked with both the occurrence and severity of COVID-19 cases. Vitamin D has its influence on viral infections through a multitude of mechanisms, such as promoting the release of antimicrobial peptides and fine-tuning the responses of the immune system. Additionally, vitamin D is intertwined with the intricate network of the renin-angiotensin system, suggesting a potential impact on the development of complications related to COVID-19. While further clinical trials and extensive research are warranted, the existing body of evidence strongly hints at the possible use of vitamin D as a valuable tool in the prophylaxis and management of COVID-19 and other viral infectious diseases.
Collapse
Affiliation(s)
| | - Öner Özdemir
- Division of Allergy and Immunology, Department of Pediatrics, Sakarya Research and Training Hospital, Sakarya University, Faculty of Medicine, Sakarya 54100, Türkiye
| |
Collapse
|
2
|
Moosazadeh Moghaddam M, Fazel P, Fallah A, Sedighian H, Kachuei R, Behzadi E, Imani Fooladi AA. Host and Pathogen-Directed Therapies against Microbial Infections Using Exosome- and Antimicrobial Peptide-derived Stem Cells with a Special look at Pulmonary Infections and Sepsis. Stem Cell Rev Rep 2023; 19:2166-2191. [PMID: 37495772 DOI: 10.1007/s12015-023-10594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Microbial diseases are a great threat to global health and cause considerable mortality and extensive economic losses each year. The medications for treating this group of diseases (antibiotics, antiviral, antifungal drugs, etc.) directly attack the pathogenic agents by recognizing the target molecules. However, it is necessary to note that excessive use of any of these drugs can lead to an increase in microbial resistance and infectious diseases. New therapeutic methods have been studied recently using emerging drugs such as mesenchymal stem cell-derived exosomes (MSC-Exos) and antimicrobial peptides (AMPs), which act based on two completely different strategies against pathogens including Host-Directed Therapy (HDT) and Pathogen-Directed Therapy (PDT), respectively. In the PDT approach, AMPs interact directly with pathogens to interrupt their intrusion, survival, and proliferation. These drugs interact directly with the cell membrane or intracellular components of pathogens and cause the death of pathogens or inhibit their replication. The mechanism of action of MSC-Exos in HDT is based on immunomodulation and regulation, promotion of tissue regeneration, and reduced host toxicity. This review studies the potential of mesenchymal stem cell-derived exosomes/ATPs therapeutic properties against microbial infectious diseases especially pulmonary infections and sepsis.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Parvindokht Fazel
- Department of Microbiology, Fars Science and Research Branch, Islamic Azad University, Shiraz, Iran
| | - Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- Academy of Medical Sciences of the I.R. of Iran, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Kayongo A, Nyiro B, Siddharthan T, Kirenga B, Checkley W, Lutaakome Joloba M, Ellner J, Salgame P. Mechanisms of lung damage in tuberculosis: implications for chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2023; 13:1146571. [PMID: 37415827 PMCID: PMC10320222 DOI: 10.3389/fcimb.2023.1146571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Pulmonary tuberculosis is increasingly recognized as a risk factor for COPD. Severe lung function impairment has been reported in post-TB patients. Despite increasing evidence to support the association between TB and COPD, only a few studies describe the immunological basis of COPD among TB patients following successful treatment completion. In this review, we draw on well-elaborated Mycobacterium tuberculosis-induced immune mechanisms in the lungs to highlight shared mechanisms for COPD pathogenesis in the setting of tuberculosis disease. We further examine how such mechanisms could be exploited to guide COPD therapeutics.
Collapse
Affiliation(s)
- Alex Kayongo
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Brian Nyiro
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, United States
| | - Bruce Kirenga
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
- Center for Global Non-Communicable Disease Research and Training, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Moses Lutaakome Joloba
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Jerrold Ellner
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
4
|
Patyra E, Kwiatek K. Insect Meals and Insect Antimicrobial Peptides as an Alternative for Antibiotics and Growth Promoters in Livestock Production. Pathogens 2023; 12:854. [PMID: 37375544 DOI: 10.3390/pathogens12060854] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The extensive use of antibiotics in animal production has led to the development of antibiotic-resistant microorganisms and the search for alternative antimicrobial agents in animal production. One such compound may be antimicrobial peptides (AMPs), which are characterized by, among others, a wide range of biocidal activity. According to scientific data, insects produce the largest number of antimicrobial peptides, and the changing EU legislation has allowed processed animal protein derived from insects to be used in feed for farm animals, which, in addition to a protein supplement, may prove to be an alternative to antibiotics and antibiotic growth promoters due to their documented beneficial impact on livestock health. In animals that were fed feeds with the addition of insect meals, changes in their intestinal microbiota, strengthened immunity, and increased antibacterial activity were confirmed to be positive effects obtained thanks to the insect diet. This paper reviews the literature on sources of antibacterial peptides and the mechanism of action of these compounds, with particular emphasis on insect antibacterial peptides and their potential impact on animal health, and legal regulations related to the use of insect meals in animal nutrition.
Collapse
Affiliation(s)
- Ewelina Patyra
- Department of Hygiene of Animal Feedingstuffs, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland
| | - Krzysztof Kwiatek
- Department of Hygiene of Animal Feedingstuffs, National Veterinary Research Institute, Partyzantów 57 Avenue, 24-100 Puławy, Poland
| |
Collapse
|
5
|
Madruga D, Garcia MM, Martino L, Hassan H, Elayat G, Ghali L, Ceballos L. Positive correlational shift between crevicular antimicrobial peptide LL-37, pain and periodontal status following non-surgical periodontal therapy. A pilot study. BMC Oral Health 2023; 23:335. [PMID: 37246231 DOI: 10.1186/s12903-023-03023-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 05/06/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Periodontitis has a high prevalence and uncertain recurrence. Unlike the pro-inflammatory cytokine profile, little is known about the anti-inflammatory cytokine and antimicrobial peptide overview following treatment. The present study aimed to evaluate if any of the antimicrobial peptide LL-37, interleukin (IL) 4, 10 and 6 together with the volume of gingival crevicular fluid (GCF) and total protein concentration in GCF could be used as correlative biomarkers for the severity in periodontitis as well as prognostic factors in the management of the disease. METHODS Forty-five participants were recruited and allocated to the healthy (15), Stage I-II (15) or Stage III-IV periodontitis (15) group. Along with periodontal examination, GCF samples were obtained at baseline and 4-6 weeks following scaling and root planing (SRP) for the periodontitis groups. GCF samples were analyzed by ELISA kits to quantify LL-37 and IL-4, -6 and - 10. One-way ANOVA followed by Dunnett's test was used to determine differences among the three groups at baseline. Two-way ANOVA followed by Sidak's post-hoc test was used to compare between pre- and post-SRP in the two periodontitis groups. RESULTS The amount of GCF volume was significantly correlated to the severity of periodontitis and decreased following SRP, particularly in the Stage III-IV group (p < 0.01). The levels of LL-37, IL-6, and pain and periodontal clinical parameters were significantly correlated to the severity of periodontitis. IL-4 and IL-10 in the periodontitis groups were significantly lower than the healthy group (p < 0.0001) and barely improved following SRP up to the level of the healthy group. CONCLUSIONS With the limitations of this study, crevicular LL-37 may be a candidate for a biomarker of periodontitis and the associated pain upon probing. TRIAL REGISTRATION The study was registered in clinical trials.gov, with number NCT04404335, dated 27/05/2020.
Collapse
Affiliation(s)
- David Madruga
- Area of Stomatology, Department of Nursing and Stomatology, Faculty of Health Sciences, Universidad Rey Juan Carlos (URJC), Avda. de Atenas s/n, Alcorcón, E-28922, Spain
| | - Miguel M Garcia
- Area of Pharmacology, Nutrition and Bromatology, Department of Basic Health Sciences, Faculty of Health Sciences, Universidad Rey Juan Carlos (URJC), Unidad Asociada I+D+i Instituto de Química Médica (IQM) CSIC-URJC, Avda. de Atenas s/n, Alcorcón, E-28922, Spain.
- High Performance Experimental Pharmacology Research Group, Universidad Rey Juan Carlos (PHARMAKOM), Alcorcón, Spain.
- Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Universidad Rey Juan Carlos (URJC), Alcorcón, Spain.
| | - Luca Martino
- Area of Signal Theory and Communications, Department of Signal Theory and Communications and Telematics Systems and Computing, Higher Technical School of Telecommunications Engineering, Universidad Rey Juan Carlos (URJC), Cam. del Molino, 5, Fuenlabrada, E-28942, Spain
- High Performance Data Science and Signal Processing for Networks and Society research group, Universidad Rey Juan Carlos (DSSP), Fuenlabrada, Spain
| | - Haidar Hassan
- Academic Plastic Surgery, School of Medicine and Dentistry, Blizard Institute, Barts and The London, Queen Mary University of London, London, E1 2AD, UK
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, NW4 4BT, UK
| | - Ghada Elayat
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, NW4 4BT, UK
- Department of Pathology, Faculty of Medicine, Tanta University, El Bahr St, Tanta, 31111, Egypt
| | - Lucy Ghali
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, NW4 4BT, UK
| | - Laura Ceballos
- Area of Stomatology, Department of Nursing and Stomatology, Faculty of Health Sciences, Universidad Rey Juan Carlos (URJC), Avda. de Atenas s/n, Alcorcón, E-28922, Spain
- High Performance Development and Innovation in Dental Biomaterials Research Group, Universidad Rey Juan Carlos (IDIBO), Alcorcón, Spain
| |
Collapse
|
6
|
RM VR, Singh N, Murmu S, Abhishek, Raina S, Singh S. Salivary physicochemical characteristics and antimicrobial human peptide among Indian children with dental caries. Bioinformation 2023; 19:428-432. [PMID: 37822819 PMCID: PMC10563558 DOI: 10.6026/97320630019428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/30/2023] [Accepted: 04/30/2023] [Indexed: 10/13/2023] Open
Abstract
Salivary innate defenses encompass mechanical cleaning, calcium phosphate salts and fluoride ion reduction of enamel dissolution rate, buffering capacity and neutralizing capacity, and antibacterial properties employing antimicrobial substances like antimicrobial peptides,agglutins,lactoferrin, lactoperoxidase, lysozyme and immuno globulins. Antimicrobial peptides play a key role in the initial defensive responses that make up innate immunity. The aim of this study was to assess the relationship between salivary physicochemical characteristics like buffering capacity, pH, flow rate, and concentrations antimicrobial human peptide like HBD-3, HNP1-3 and LL-37 and caries activity in young children. Before to collecting the samples, informed permission papers were gathered and completed by the children's legal guardians or parents. There was significant correlation showing reduced caries activity on increased concentrations of HNP1-3. When there was analysis of correlation of CAS with LL-37 concentrations then the p value vas 0.002 showing the correlation was significant. There is significant relationship between salivary physicochemical characteristics like buffering capacity, pH, flow rate, and concentrations antimicrobial human peptide like HBD-3, HNP1-3 and LL-37 and caries activity in young children.
Collapse
Affiliation(s)
- Vatchala Rani RM
- Department of Oral Pathology and Microbiology Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India
| | - Neha Singh
- Tutor, Department of Periodontology, Dental College, Rims, Ranchi, Jharkhand, India
| | - Swati Murmu
- Post Graduate Student, Department of Paediatric and Preventive Dentistry, Hazaribag College of Dental Sciences and Hospital, Hazaribagh, Jharkhand, India
| | - Abhishek
- Private Practioner, Oral and Maxillofacial Surgeon, Oro Care Facial Trauma Centre, Patna, Bihar, India
| | - Sakshi Raina
- Department of Orthodontics, Dental Officer, Echs Polyclinic (Ministry of Defence) Samastipur, Bihar, India
| | - Shalini Singh
- Senior Resident, Department of Pedodontist, Safdarjung Hospital, New Delhi, India
| |
Collapse
|
7
|
Lappin MJ, Dellett M, Mills KI, Lundy FT, Irwin CR. The neutralising and stimulatory effects of antimicrobial peptide LL-37 in human gingival fibroblasts. Arch Oral Biol 2023; 148:105634. [PMID: 36773560 DOI: 10.1016/j.archoralbio.2023.105634] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To investigate the effects of LL-37, a broad spectrum antimicrobial peptide expressed in periodontal tissues, on human gingival fibroblast responsiveness to microbial challenge and to explore the direct effects of LL-37 on human gingival fibroblasts. DESIGN The effect of LL-37 on bacterial lipopolysaccharide-induced expression of Interleukin (IL-6) and chemokine C-X-C motif ligand (CXCL) 8 was determined by enzyme linked immunosorbent assay (ELISA). LL-37's influence on bacterial lipopolysaccharide-induced IκBα degradation was investigated by western blot. DNA microarray analysis initially determined the direct effects of LL-37 on gene expression, these findings were subsequently confirmed by quantitative polymerase chain reaction and ELISA analysis of selected genes. RESULTS Bacterial lipopolysaccharide-induced IL-6 and CXCL8 production by human gingival fibroblasts was significantly reduced in the presence of LL-37 at concentrations in the range of 1-10 µg/ml. LL-37 led to a reduction in lipopolysaccharide-induced IκBα degradation by Escherichia coli lipopolysaccharide and Porphyromonas gingivalis lipopolysaccharide (10 µg/ml). LL-37 (50 µg/ml) significantly altered the gene expression of 367 genes in human gingival fibroblasts by at least 2-fold. CXCL1, CXCL2, CXCL3, Interleukin-24 (IL-24), CXCL8, Chemokine (C-C motif) Ligand 2, and Suppressor of Cytokine Signalling 3 mRNA were significantly upregulated by LL-37. LL-37 also significantly stimulated expression of CXCL8, hepatocyte growth factor and CXCL1 at the protein level. CONCLUSION LL-37 plays an important regulatory role in the immunomodulatory activity of gingival fibroblasts by inhibiting lipopolysaccharide -induced expression of inflammatory cytokines and directly stimulating the expression of an array of bioactive molecules involved in inflammation and repair.
Collapse
Affiliation(s)
| | - M Dellett
- Patrick G Johnston Centre for Cancer Research, UK
| | - K I Mills
- Patrick G Johnston Centre for Cancer Research, UK
| | - F T Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | | |
Collapse
|
8
|
Molecular Mechanisms of Hyperoxia-Induced Neonatal Intestinal Injury. Int J Mol Sci 2023; 24:ijms24054366. [PMID: 36901800 PMCID: PMC10002283 DOI: 10.3390/ijms24054366] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Oxygen therapy is important for newborns. However, hyperoxia can cause intestinal inflammation and injury. Hyperoxia-induced oxidative stress is mediated by multiple molecular factors and leads to intestinal damage. Histological changes include ileal mucosal thickness, intestinal barrier damage, and fewer Paneth cells, goblet cells, and villi, effects which decrease the protection from pathogens and increase the risk of necrotizing enterocolitis (NEC). It also causes vascular changes with microbiota influence. Hyperoxia-induced intestinal injuries are influenced by several molecular factors, including excessive nitric oxide, the nuclear factor-κB (NF-κB) pathway, reactive oxygen species, toll-like receptor-4, CXC motif ligand-1, and interleukin-6. Nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and some antioxidant cytokines or molecules including interleukin-17D, n-acetylcysteine, arginyl-glutamine, deoxyribonucleic acid, cathelicidin, and health microbiota play a role in preventing cell apoptosis and tissue inflammation from oxidative stress. NF-κB and Nrf2 pathways are essential to maintain the balance of oxidative stress and antioxidants and prevent cell apoptosis and tissue inflammation. Intestinal inflammation can lead to intestinal damage and death of the intestinal tissue, such as in NEC. This review focuses on histologic changes and molecular pathways of hyperoxia-induced intestinal injuries to establish a framework for potential interventions.
Collapse
|
9
|
Kato H, Ohta K, Akagi M, Fukada S, Sakuma M, Naruse T, Nishi H, Shigeishi H, Takechi M, Aikawa T. LL-37-dsRNA Complexes Modulate Immune Response via RIG-I in Oral Keratinocytes. Inflammation 2023; 46:808-823. [PMID: 36763254 DOI: 10.1007/s10753-023-01787-5] [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: 12/19/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Recognition of nucleic acids as pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) promotes an inflammatory response. On the other hand, LL-37, an antimicrobial peptide, is a multifunctional modulator of immune response, though whether it modulates inflammatory responses induced by nucleic acids in oral keratinocytes is unknown. In this study, we firstly investigated the effect of LL-37 on CXCL10 induced by DAMPs and PAMPs in immortalized oral keratinocytes, RT7. Furthermore, the effects of LL-37 on translocation of exogenous nucleic acids into cytoplasm as well as cytosolic receptor, RIG-I on immune responses mediated by LL-37-nucleic acid complexes were examined. From these results, LL-37 enhanced necrotic cell supernatant (NCS)-induced CXCL10 expression in RT7, while the response was decreased by RNase. Complexes of LL-37 and double-stranded (ds) RNA, Poly(I:C) enhanced CXCL10 expression in comparison with each alone, which were associated with NF-κB activation. Furthermore, LL-37 was shown to bind with ds nucleotides and translocate into cytoplasm. Knockdown of RIG-I decreased expression of CXCL10 induced by LL-37-Poly(I:C) complexes, and RIG-I were co-localized with Poly(I:C) entered by LL-37 in cytoplasm. LL-37 modulates dsRNA-mediated inflammatory response via RIG-I in oral keratinocytes, which may play an important role in the pathogenesis of oral inflammatory diseases.
Collapse
Affiliation(s)
- Hiroki Kato
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
- Department of Dentistry, Oral and Maxillofacial Surgery, National Hospital Organization Kure Medical Centerand, Chugoku Cancer Center , 3-1 Aoyama-Cho, Kure, 737-0023, Japan
| | - Kouji Ohta
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan.
| | - Misaki Akagi
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Shohei Fukada
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Miyuki Sakuma
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Takako Naruse
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Hiromi Nishi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Hideo Shigeishi
- Department of Public Oral Health, Program of Oral Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Masaaki Takechi
- Department of Dentistry, Oral and Maxillofacial Surgery, National Hospital Organization Kure Medical Centerand, Chugoku Cancer Center , 3-1 Aoyama-Cho, Kure, 737-0023, Japan
| | - Tomonao Aikawa
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| |
Collapse
|
10
|
Othumpangat S, Noti JD. β-Defensin-1 Regulates Influenza Virus Infection in Human Bronchial Epithelial Cells through the STAT3 Signaling Pathway. Pathogens 2023; 12:pathogens12010123. [PMID: 36678471 PMCID: PMC9865356 DOI: 10.3390/pathogens12010123] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Understanding the host response to influenza A virus (IAV) infection is vital for developing intervention strategies. The primary barriers for invading respiratory pathogens are the respiratory tract epithelial cells and antimicrobial proteins generated by these cells. The antimicrobial peptide, β-defensin-1, has antiviral activity against both enveloped and non-enveloped viruses. Significant downregulation of β-defensin1 gene (DEFB1) expression was observed when human bronchial epithelial cells (HBEpCs) were exposed to IAV. HBEpCs overexpressing DEFB1 caused a significant reduction in IAV, that was confirmed by IAV matrix gene analysis, plaque assay, and confocal microscopy. DEFB1 expression after transfection with two micro RNAs (miRNAs), hsa-miR-186-5p and hsa-miR-340-5p, provided evidence that DEFB1 expression could be modulated by these miRNAs and hsa-miR-186-5p had a higher binding efficiency with DEFB1. Overexpression of DEFB1 in IAV-infected HBEpCs led to increased NF-κB expression. In a PCR array analysis of 84 transcription factors, either overexpressing DEFB1 or siRNA silencing of DEFB1 expression significantly modulated the expression of signal transducer and activator of transcription 3 (STAT3). In addition, Ingenuity Pathway Analysis (IPA) integrated with PCR array data showed that the JAK1/STAT3 pathway was significantly altered in cells overexpressing DEFB1, suggesting this to be one of the pathways by which defensin regulates IAV replication in HBEpCs. In conclusion, the reduction in IAV copy number in DEFB1 overexpressing cells suggests that β-defensin-1 plays a key role in regulating IAV survival through STAT3 and is a potential target for antiviral drug development.
Collapse
|
11
|
Fang X, Nong K, Wang Z, Jin Y, Gao F, Zeng Q, Wang X, Zhang H. Human cathelicidin LL-37 exerts amelioration effects against EHEC O157:H7 infection regarding inflammation, enteric dysbacteriosis, and impairment of gut barrier function. Peptides 2023; 159:170903. [PMID: 36370932 DOI: 10.1016/j.peptides.2022.170903] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) O157:H7 infection impairs intestinal barrier function, causing intestinal inflammation and enteric dysbacteriosis. The human cathelicidin LL-37 can regulate excessive inflammatory responses, barrier function, and balance the intestinal microbial community; however, little is known about its effects on inflammation, intestinal barrier function, and microbiota disorders in EHEC O157:H7-infected mice. In this study, we investigated the protective effect of LL-37 against EHEC O157:H7 infection and elucidated the underlying mechanism using a mouse model. LL-37 treatment was found to inhibit body weight loss, restore edema and destruction of the intestinal villi, and significantly reduce epithelial apoptosis (P < 0.05) in EHEC O157:H7-infected mice. Furthermore, inflammatory infiltration of macrophages and neutrophils into the jejunum and colon was significantly decreased (P < 0.05). LL-37 significantly downregulated the production of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) (P < 0.05) and upregulated the anti-inflammatory cytokine (IL-10) during EHEC O157:H7 infection. LL-37 increased the expression of tight junction proteins (ZO-1, ZO-2, claudin-1, and occludin), which are associated with intestinal barrier function, and had a positive effect on EHEC O157:H7-induced microbial disorders, particularly in terms of the inflammation-related microbiota. LL-37 also significantly decreased the E. coli load in the liver and spleen (P < 0.01) and restored the structure of the liver and kidney. Taken together, LL-37 conferred protection in a EHEC O157:H7-induced mouse model by reducing intestinal inflammation, enhancing intestinal barrier function, and restoring the balance of the intestinal microbiota, which indicates the therapeutic potential of LL-37 against pathogen infection.
Collapse
Affiliation(s)
- Xin Fang
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Keyi Nong
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Zihan Wang
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Yuanli Jin
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Feng Gao
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Qiuyu Zeng
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Xuemei Wang
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China
| | - Haiwen Zhang
- College of Animal Science and Technology of Hainan University, Haikou, Hainan 570228, China.
| |
Collapse
|
12
|
Wang F, Cui Y, He D, Gong L, Liang H. Natural killer cells in sepsis: Friends or foes? Front Immunol 2023; 14:1101918. [PMID: 36776839 PMCID: PMC9909201 DOI: 10.3389/fimmu.2023.1101918] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Sepsis is one of the major causes of death in the hospital worldwide. The pathology of sepsis is tightly associated with dysregulation of innate immune responses. The contribution of macrophages, neutrophils, and dendritic cells to sepsis is well documented, whereas the role of natural killer (NK) cells, which are critical innate lymphoid lineage cells, remains unclear. In some studies, the activation of NK cells has been reported as a risk factor leading to severe organ damage or death. In sharp contrast, some other studies revealed that triggering NK cell activity contributes to alleviating sepsis. In all, although there are several reports on NK cells in sepsis, whether they exert detrimental or protective effects remains unclear. Here, we will review the available experimental and clinical studies about the opposing roles of NK cells in sepsis, and we will discuss the prospects for NK cell-based immunotherapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yiqin Cui
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dongmei He
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lisha Gong
- School of Laboratory Medicine and Technology, Harbin Medical University, Daqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
13
|
Chou HC, Chen CM. Cathelicidin Attenuates Hyperoxia-Induced Lung Injury by Inhibiting Ferroptosis in Newborn Rats. Antioxidants (Basel) 2022; 11:antiox11122405. [PMID: 36552613 PMCID: PMC9774284 DOI: 10.3390/antiox11122405] [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: 10/07/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022] Open
Abstract
High oxygen concentrations are often required to treat newborn infants with respiratory distress but have adverse effects, such as increased oxidative stress and ferroptosis and impaired alveolarization. Cathelicidins are a family of antimicrobial peptides that exhibit antioxidant activity, and they can reduce hyperoxia-induced oxidative stress. This study evaluated the effects of cathelicidin treatment on lung ferroptosis and alveolarization in hyperoxia-exposed newborn rats. Sprague Dawley rat pups were either reared in room air (RA) or hyperoxia (85% O2) and then randomly given cathelicidin (8 mg/kg) in 0.05 mL of normal saline (NS), or NS was administered intraperitoneally on postnatal days from 1-6. The four groups obtained were as follows: RA + NS, RA + cathelicidin, O2 + NS, and O2 + cathelicidin. On postnatal day 7, lungs were harvested for histological, biochemical, and Western blot analyses. The rats nurtured in hyperoxia and treated with NS exhibited significantly lower body weight and cathelicidin expression, higher Fe2+, malondialdehyde, iron deposition, mitochondrial damage (TOMM20), and interleukin-1β (IL-1β), and significantly lower glutathione, glutathione peroxidase 4, and radial alveolar count (RAC) compared to the rats kept in RA and treated with NS or cathelicidin. Cathelicidin treatment mitigated hyperoxia-induced lung injury, as demonstrated by higher RAC and lower TOMM20 and IL-1β levels. The attenuation of lung injury was accompanied by decreased ferroptosis. These findings indicated that cathelicidin mitigated hyperoxia-induced lung injury in the rats, most likely by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence:
| |
Collapse
|
14
|
Pezzanite LM, Chow L, Strumpf A, Johnson V, Dow SW. Immune Activated Cellular Therapy for Drug Resistant Infections: Rationale, Mechanisms, and Implications for Veterinary Medicine. Vet Sci 2022; 9:610. [PMID: 36356087 PMCID: PMC9695672 DOI: 10.3390/vetsci9110610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/18/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Antimicrobial resistance and biofilm formation both present challenges to treatment of bacterial infections with conventional antibiotic therapy and serve as the impetus for development of improved therapeutic approaches. Mesenchymal stromal cell (MSC) therapy exerts an antimicrobial effect as demonstrated in multiple acute bacterial infection models. This effect can be enhanced by pre-conditioning the MSC with Toll or Nod-like receptor stimulation, termed activated cellular therapy (ACT). The purpose of this review is to summarize the current literature on mechanisms of antimicrobial activity of MSC with emphasis on enhanced effects through receptor agonism, and data supporting use of ACT in treatment of bacterial infections in veterinary species including dogs, cats, and horses with implications for further treatment applications. This review will advance the field's understanding of the use of activated antimicrobial cellular therapy to treat infection, including mechanisms of action and potential therapeutic applications.
Collapse
Affiliation(s)
- Lynn M. Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Alyssa Strumpf
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Valerie Johnson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Michigan State University, Lansing, MI 48824, USA
| | - Steven W. Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
15
|
Wu E, Zhu J, Ma Z, Tuo B, Terai S, Mizuno K, Li T, Liu X. Gastric alarmin release: A warning signal in the development of gastric mucosal diseases. Front Immunol 2022; 13:1008047. [PMID: 36275647 PMCID: PMC9583272 DOI: 10.3389/fimmu.2022.1008047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Alarmins exist outside cells and are early warning signals to the immune system; as such, alarmin receptors are widely distributed on various immune cells. Alarmins, proinflammatory molecular patterns associated with tissue damage, are usually released into the extracellular space, where they induce immune responses and participate in the damage and repair processes of mucosal diseases.In the stomach, gastric alarmin release has been shown to be involved in gastric mucosal inflammation, antibacterial defense, adaptive immunity, and wound healing; moreover, this release causes damage and results in the development of gastric mucosal diseases, including various types of gastritis, ulcers, and gastric cancer. Therefore, it is necessary to understand the role of alarmins in gastric mucosal diseases. This review focuses on the contribution of alarmins, including IL33, HMGB1, defensins and cathelicidins, to the gastric mucosal barrier and their role in gastric mucosal diseases. Here, we offer a new perspective on the prevention and treatment of gastric mucosal diseases.
Collapse
Affiliation(s)
- Enqin Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shuji Terai
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenichi Mizuno
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Xuemei Liu, ; Taolang Li,
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Xuemei Liu, ; Taolang Li,
| |
Collapse
|
16
|
Antimicrobial Peptides in Early-Life Host Defense, Perinatal Infections, and Necrotizing Enterocolitis—An Update. J Clin Med 2022; 11:jcm11175074. [PMID: 36079001 PMCID: PMC9457252 DOI: 10.3390/jcm11175074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
Host defense against early-life infections such as chorioamnionitis, neonatal sepsis, or necrotizing enterocolitis (NEC) relies primarily on innate immunity, in which antimicrobial peptides (AMPs) play a major role. AMPs that are important for the fetus and neonate include α and β defensins, cathelicidin LL-37, antiproteases (elafin, SLPI), and hepcidin. They can be produced by the fetus or neonate, the placenta, chorioamniotic membranes, recruited neutrophils, and milk-protein ingestion or proteolysis. They possess antimicrobial, immunomodulating, inflammation-regulating, and tissue-repairing properties. AMPs are expressed as early as the 13th week and increase progressively through gestation. Limited studies are available on AMP expression and levels in the fetus and neonate. Nevertheless, existing evidence supports the role of AMPs in pathogenesis of chorioamnionitis, neonatal sepsis, and NEC, and their association with disease severity. This suggests a potential role of AMPs in diagnosis, prevention, prognosis, and treatment of sepsis and NEC. Herein, we present an overview of the antimicrobial and immunomodulating properties of human AMPs, their sources in the intrauterine environment, fetus, and neonate, and their changes during pre- and post-natal infections and NEC. We also discuss emerging data regarding the potential utility of AMPs in early-life infections, as diagnostic or predictive biomarkers and as therapeutic alternatives or adjuncts to antibiotic therapy considering the increase of antibiotic resistance in neonatal intensive care units.
Collapse
|
17
|
Diagnostic and Therapeutic Potential for HNP-1, HBD-1 and HBD-4 in Pregnant Women with COVID-19. Int J Mol Sci 2022; 23:ijms23073450. [PMID: 35408809 PMCID: PMC8998699 DOI: 10.3390/ijms23073450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 01/27/2023] Open
Abstract
Pregnancy is characterized by significant immunological changes and a cytokine profile, as well as vitamin deficiencies that can cause problems for the correct development of a fetus. Defensins are small antimicrobial peptides that are part of the innate immune system and are involved in several biological activities. Following that, this study aims to compare the levels of various cytokines and to investigate the role of defensins between pregnant women with confirmed COVID-19 infection and pregnant women without any defined risk factor. TNF-α, TGF-β, IL-2 and IL-10, β-defensins, have been evaluated by gene expression in our population. At the same time, by ELISA assay IL-6, IL-8, defensin alpha 1, defensin beta 1 and defensin beta 4 have been measured. The data obtained show that mothers affected by COVID-19 have an increase in pro-inflammatory factors (TNF-α, TGF-β, IL-2, IL-6, IL-8) compared to controls; this increase could generate a sort of “protection of the fetus” from virus attacks. Contemporarily, we have an increase in the anti-inflammatory cytokine IL-10 and an increase in AMPs, which highlights how the mother’s body is responding to the viral attack. These results allow us to hypothesize a mechanism of “trafficking” of antimicrobial peptides from the mother to the fetus that would help the fetus to protect itself from the infection in progress.
Collapse
|
18
|
Rather IA, Sabir JSM, Asseri AH, Ali S. Antifungal Activity of Human Cathelicidin LL-37, a Membrane Disrupting Peptide, by Triggering Oxidative Stress and Cell Cycle Arrest in Candida auris. J Fungi (Basel) 2022; 8:204. [PMID: 35205958 PMCID: PMC8875705 DOI: 10.3390/jof8020204] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/06/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023] Open
Abstract
Candida auris, an evolving multidrug-resistant pathogenic yeast, is known for causing severe invasive infections associated with high mortality rates in hospitalized individuals. Distinct from other Candida species, C. auris can persist for longer periods on different surfaces and is resistant to all of the major classes of antifungal drugs. Therefore, there is an urgent need for new antimycotic drugs with improved efficacy and reduced toxicity. The development of new antifungals based on antimicrobial peptides from various sources is considered a promising alternative. In this study, we examined the in vitro anti-yeast activity of the human cathelicidin peptides LL-37 against clinical strains of C. auris alone and in combination with different antifungal drugs by broth microdilution assay. To understand the antifungal mechanism of action, cell envelopes, cell cycle arrest, and effect on oxidative stress enzymes were studied using standard protocols. The minimum inhibitory and fungicidal concentrations of cathelicidin LL-37 ranged from 25-100 and 50-200 µg/mL, respectively. A combination interaction in a 1:1 ratio (cathelicidin LL-37: antifungal drug) resulted in 70% synergy with fluconazole and 100% synergy with amphotericin B and caspofungin. Assessment of the C. auris membrane by using propidium iodide assay after exposure to cathelicidin LL-37 linked membrane permeabilization with inhibition of C. auris cell growth and viability. These results were backed up by scanning electron microscopy studies demonstrating that exposure with cathelicidin LL-37 caused C. auris cells to undergo extensive surface changes. Spectrophotometric analysis revealed that cathelicidin LL-37 caused oxidative stress in C. auris, as is evident from the significant increase in the activity of primary antioxidant enzymes. In addition, cathelicidin LL-37 inhibited the cell cycle and accumulated cells in the S phase. Therefore, these results specify the potential of cathelicidin LL-37 for developing a new and effective anti-Candida agent.
Collapse
Affiliation(s)
- Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia;
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Jamal S. M. Sabir
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia;
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Amer H. Asseri
- Biochemistry Department, Faculty of Science, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia;
| | - Sajad Ali
- Department of Biotechnology, Yeungnam University, Gyeongsan 385541, Korea
| |
Collapse
|
19
|
Kim J, Yang YL, Jeong Y, Jang YS. Application of Antimicrobial Peptide LL-37 as an Adjuvant for Middle East Respiratory Syndrome-Coronavirus Antigen Induces an Efficient Protective Immune Response Against Viral Infection After Intranasal Immunization. Immune Netw 2022; 22:e41. [DOI: 10.4110/in.2022.22.e41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Ju Kim
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Korea
| | - Ye Lin Yang
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| | - Yongsu Jeong
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea
| | - Yong-Suk Jang
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Korea
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
20
|
Elemam NM, Ramakrishnan RK, Hundt JE, Halwani R, Maghazachi AA, Hamid Q. Innate Lymphoid Cells and Natural Killer Cells in Bacterial Infections: Function, Dysregulation, and Therapeutic Targets. Front Cell Infect Microbiol 2021; 11:733564. [PMID: 34804991 PMCID: PMC8602108 DOI: 10.3389/fcimb.2021.733564] [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: 06/30/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Infectious diseases represent one of the largest medical challenges worldwide. Bacterial infections, in particular, remain a pertinent health challenge and burden. Moreover, such infections increase over time due to the continuous use of various antibiotics without medical need, thus leading to several side effects and bacterial resistance. Our innate immune system represents our first line of defense against any foreign pathogens. This system comprises the innate lymphoid cells (ILCs), including natural killer (NK) cells that are critical players in establishing homeostasis and immunity against infections. ILCs are a group of functionally heterogenous but potent innate immune effector cells that constitute tissue-resident sentinels against intracellular and extracellular bacterial infections. Being a nascent subset of innate lymphocytes, their role in bacterial infections is not clearly understood. Furthermore, these pathogens have developed methods to evade the host immune system, and hence permit infection spread and tissue damage. In this review, we highlight the role of the different ILC populations in various bacterial infections and the possible ways of immune evasion. Additionally, potential immunotherapies to manipulate ILC responses will be briefly discussed.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rakhee K Ramakrishnan
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jennifer E Hundt
- Lübeck Institute for Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Rabih Halwani
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Prince Abdullah Ben Khaled Celiac Disease Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Azzam A Maghazachi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| |
Collapse
|
21
|
Lohova E, Vitenberga-Verza Z, Kazoka D, Pilmane M. Local Defence System in Healthy Lungs. Clin Pract 2021; 11:728-746. [PMID: 34698129 PMCID: PMC8544484 DOI: 10.3390/clinpract11040088] [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: 06/29/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 11/22/2022] Open
Abstract
Background: The respiratory system is one of the main entrance gates for infection. The aim of this work was to compare the appearance of specific mucosal pro-inflammatory and common anti-microbial defence factors in healthy lung tissue, from an ontogenetic point of view. Materials and methods: Healthy lung tissues were collected from 15 patients (three females and 12 males) in the age range from 18 to 86. Immunohistochemistry to human β defensin 2 (HBD-2), human β defensin 3 (HBD-3), human β defensin 4 (HBD-4), cathelicidine (LL-37) and interleukine 17A (IL-17A) were performed. Results: The lung tissue material contained bronchial and lung parenchyma material in which no histological changes, connected with the inflammatory process, were detected. During the study, various statistically significant differences were detected in immunoreactive expression between different factors in all lung tissue structures. Conclusion: All healthy lung structures, but especially the cartilage, alveolar epithelium and the alveolar macrophages, are the main locations for the baseline synthesis of antimicrobial proteins and IL-17A. Cartilage shows high functional plasticity of this structure, including significant antimicrobial activity and participation in local lung protection response. Interrelated changes between antimicrobial proteins in different tissue confirm baseline synergistical cooperation of all these factors in healthy lung host defence.
Collapse
|
22
|
Yang T, Li J, Jia Q, Zhan S, Zhang Q, Wang Y, Wang X. Antimicrobial peptide 17BIPHE2 inhibits the proliferation of lung cancer cells in vitro and in vivo by regulating the ERK signaling pathway. Oncol Lett 2021; 22:501. [PMID: 33981363 PMCID: PMC8108245 DOI: 10.3892/ol.2021.12762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/24/2021] [Indexed: 12/19/2022] Open
Abstract
In 2018, there were 18.1 million new cancer cases and 9.6 million cancer-related deaths worldwide, among which the incidence rate of lung cancer (11.6%) and fatality rate (18.4%) both ranked first. The antimicrobial peptide LL-37 is an important component of the natural immune system and possesses several biological properties, including antibacterial, antiviral and anticancer effects. The antimicrobial peptide 17BIPHE2, the shortest synthetic peptide derivative of LL-37, exhibits biological activities similar to those of LL-37. The objective of the present study was to investigate the mechanism of action of exogenous 17BIPHE2 against lung cancer cells. The human lung adenocarcinoma cell line A549 was treated with 17BIPHE2. Changes in cell proliferation, migration, invasion, mitochondrial membrane potential (ΔΨm), and the levels of reactive oxygen species (ROS), Ca2+ and apoptosis-related proteins, including BAX, BCL-2 and ERK, were detected using flow cytometry, transmission electron microscopy and western blotting. The results showed that 17BIPHE2 significantly increased the apoptosis rate of A549 cells and elevated BAX expression, ERK phosphorylation, and ROS and Ca2+ levels, but decreased the expression of BCL-2, ERK and Ki67. In addition, the peptide reduced ΔΨm and the cell migration ability of A549 cells and inhibited tumor growth. ERK inhibition significantly attenuated the anticancer effect of 17BIPHE2. The present observations suggested that 17BIPHE2 can effectively inhibit cancer cells by regulating the ERK signaling pathway.
Collapse
Affiliation(s)
- Tingting Yang
- Department of Clinical Laboratory, Yinchuan Maternal and Child Health Care Hospital, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Jun Li
- Department of Clinical Laboratory, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Qinqin Jia
- Department of Laboratory Medicine, Health Center, Chun Rong, Gansu 745211, P.R. China
| | - Shisheng Zhan
- Department of Clinical Laboratory, Hebei Yanda Lu Daopei Hospital, Langfang, Hebei 065200, P.R. China
| | - Qiannan Zhang
- Department of Laboratory Medicine, College of Clinical Medicine, Shuangyi Campus, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yarong Wang
- Department of Pathology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, P.R. China
| | - Xiuqing Wang
- Department of Laboratory Medicine, College of Clinical Medicine, Shuangyi Campus, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
23
|
Laneri S, Brancaccio M, Mennitti C, De Biasi MG, Pero ME, Pisanelli G, Scudiero O, Pero R. Antimicrobial Peptides and Physical Activity: A Great Hope against COVID 19. Microorganisms 2021; 9:1415. [PMID: 34209064 PMCID: PMC8304224 DOI: 10.3390/microorganisms9071415] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/07/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial peptides (AMPs), α- and β-defensins, possess antiviral properties. These AMPs achieve viral inhibition through different mechanisms of action. For example, they can: (i) bind directly to virions; (ii) bind to and modulate host cell-surface receptors, disrupting intracellular signaling; (iii) function as chemokines to augment and alter adaptive immune responses. Given their antiviral properties and the fact that the development of an effective coronavirus disease 2019 (COVID-19) treatment is an urgent public health priority, they and their derivatives are being explored as potential therapies against COVID-19. These explorations using various strategies, range from their direct interaction with the virus to using them as vaccine adjuvants. However, AMPs do not work in isolation, specifically in their role as potent immune modulators, where they interact with toll-like receptors (TLRs) and chemokine receptors. Both of these receptors have been shown to play roles in COVID-19 pathogenesis. In addition, it is known that a healthy lifestyle accompanied by controlled physical activity can represent a natural weapon against COVID-19. In competitive athletes, an increase in serum defensins has been shown to function as self-protection from the attack of microorganisms, consequently a controlled physical activity could act as a support to any therapies in fighting COVID-19. Therefore, including information on all these players' interactions would produce a complete picture of AMP-based therapies' response.
Collapse
Affiliation(s)
- Sonia Laneri
- Department of Pharmacy, University of Naples Federico II, Via Montesano, 80138 Naples, Italy; (S.L.); (M.G.D.B.)
| | - Mariarita Brancaccio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.B.); (C.M.)
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.B.); (C.M.)
| | - Margherita G. De Biasi
- Department of Pharmacy, University of Naples Federico II, Via Montesano, 80138 Naples, Italy; (S.L.); (M.G.D.B.)
| | - Maria Elena Pero
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy; (M.E.P.); (G.P.)
| | - Giuseppe Pisanelli
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy; (M.E.P.); (G.P.)
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.B.); (C.M.)
- Ceinge Biotecnologie Avanzate S.C.aR.L., 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Raffaela Pero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.B.); (C.M.)
- Ceinge Biotecnologie Avanzate S.C.aR.L., 80131 Naples, Italy
| |
Collapse
|
24
|
Ghosh SK, Weinberg A. Ramping Up Antimicrobial Peptides Against Severe Acute Respiratory Syndrome Coronavirus-2. Front Mol Biosci 2021; 8:620806. [PMID: 34235176 PMCID: PMC8255374 DOI: 10.3389/fmolb.2021.620806] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 05/27/2021] [Indexed: 12/17/2022] Open
Abstract
Human-derived antimicrobial peptides (AMPs), such as defensins and cathelicidin LL-37, are members of the innate immune system and play a crucial role in early pulmonary defense against viruses. These AMPs achieve viral inhibition through a variety of mechanisms including, but not limited to, direct binding to virions, binding to and modulating host cell-surface receptors, blocking viral replication, and aggregation of viral particles and indirectly by functioning as chemokines to enhance or curb adaptive immune responses. Given the fact that we are in a pandemic of unprecedented severity and the urgent need for therapeutic options to combat severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), naturally expressed AMPs and their derivatives have the potential to combat coronavirus disease 2019 (COVID-19) and impede viral infectivity in various ways. Provided the fact that development of effective treatments is an urgent public health priority, AMPs and their derivatives are being explored as potential prophylactic and therapeutic candidates. Additionally, cell-based platforms such as human mesenchymal stem cell (hMSC) therapy are showing success in saving the lives of severely ill patients infected with SARS-CoV-2. This could be partially due to AMPs released from hMSCs that also act as immunological rheostats to modulate the host inflammatory response. This review highlights the utilization of AMPs in strategies that could be implemented as novel therapeutics, either alone or in combination with other platforms, to treat CoV-2-infected individuals.
Collapse
Affiliation(s)
| | - Aaron Weinberg
- Department of Biological Sciences, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
25
|
Aslanhan U, Cakir E, Pur Ozyigit L, Kucuksezer UC, Gelmez YM, Yuksel M, Deniz G, Cetin Aktas E. Pseudomonas aeruginosa colonization in cystic fibrosis: Impact on neutrophil functions and cytokine secretion capacity. Pediatr Pulmonol 2021; 56:1504-1513. [PMID: 33512090 DOI: 10.1002/ppul.25294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 01/03/2021] [Accepted: 01/22/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Chronic colonization with Pseudomonas (P.) aeruginosa worsens the prognosis of cystic fibrosis (CF) patients. This study aims to analyze the functional properties of neutrophils in CF patients with P. aeruginosa colonization. METHODS Patients with CF (n = 16) were grouped by positivity of P. aeruginosa in sputum culture, as positive (P.+) or negative (P.-), then compared with age and sex matched healthy controls (n = 8). Adhesion molecules, apoptotic index, intracellular CAP-18, interleukin 8 (IL-8), and tumor necrosis factor α (TNF-α) levels of neutrophils, following P. aeruginosa and lipopolysaccharides (LPS) stimulation, were analyzed by flow cytometry. IL-1β, IL-6, TNF-α, and IL-17 plasma levels were determined by Luminex. RESULTS Patients with CF had increased phagocytosis of Escherichia coli and P. aeruginosa, upregulated oxidative burst and chemotaxis. Increased neutrophil apoptosis was noted in CF patients. In unstimulated conditions, higher levels of CD16+ TNF-α+ and CD16+ IL-8+ neutrophils were determined, whereas bacteria and LPS stimulation significantly decreased secretion of CAP-18 from CD16+ neutrophils of CF patients. Plasma levels of IL-1β, TNF-α and IL-17 in P.+ patients were higher than in P.- group. CONCLUSION Our findings confirm inadequate neutrophil defense towards pathogens in CF. A significant difference in migration, phagocytosis, oxidative burst, percentage of IL-8 producing neutrophils, IL-1β, TNF-α, and IL-17 secretions were noted among CF patients according to their colonization status, which might induce a further destructive effect on airways, resulting in an unfavorable prognosis for children with CF who also have colonization.
Collapse
Affiliation(s)
- Umit Aslanhan
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.,Department of Immunology, Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Erkan Cakir
- Department of Pediatric Pulmonology, Bezmialem Vakif University Medical Faculty, Istanbul, Turkey
| | - Leyla Pur Ozyigit
- Department of Allergy and Immunology, University Hospitals of Leicester, Leicester, UK
| | - Umut Can Kucuksezer
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Yusuf Metin Gelmez
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Mine Yuksel
- Department of Pediatric Pulmonology, Bezmialem Vakif University Medical Faculty, Istanbul, Turkey
| | - Gunnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Esin Cetin Aktas
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
26
|
Ramezani J, Khaligh MR, Ansari G, Yazdani Y, Mohammadi S. Association of salivary physicochemical characteristics and peptide levels with dental caries in children. J Indian Soc Pedod Prev Dent 2021; 39:189-195. [PMID: 34341240 DOI: 10.4103/jisppd.jisppd_251_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIM The purpose of this investigation was to evaluate the association of physicochemical properties and antimicrobial peptide levels of saliva with caries activity in children. MATERIALS AND METHODS The required volume of unstimulated saliva was collected from 41 children aged 3-12 years with no systemic diseases. Caries activity was calculated using DMFS and dmfs records for each participating child. Collected saliva samples were then examined for their flow rate, pH, and buffering capacity. The concentration of three peptides was assessed including LL-37, human neutrophil peptide (HNP) 1-3, and human beta-defensin (HBD)-3 through an enzyme-linked immunosorbent assay. The correlation between caries activity score (CAS) and salivary variables was looked using the linear regression and Spearman's correlation method. The comparison of CAS means between high- and low-value groups of salivary items was performed using independent sample t-test while the association of CAS and salivary parameters in categorical scale was tested by Chi-square test. RESULTS No statistically significant differences were found between the CAS means at low and high categories of each salivary physicochemical parameter and those of antimicrobial peptides. There was a negative correlation between HNP1-3 and CAS and also between HBD-3 and CAS, but these results were not statistically meaningful. High HNP1-3 concentration was noted in 67% of the low caries rate group and 29% of the high caries rate group, with a statistically significant difference between the low and high caries rate groups (P = 0.019). CONCLUSION Salivary inherent factors are not dominant determinants in caries activity. The current results may suggest that α-defensins (HNP1-3) have a protective role against dental caries.
Collapse
Affiliation(s)
- Jamileh Ramezani
- Department of Pediatric Dentistry, Faculty of Dentistry, Golestan University of Medical Sciences; Dental Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Rezaei Khaligh
- Department of Pediatric Dentistry, Faculty of Dentistry, Golestan University of Medical Sciences; Dental Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ghassem Ansari
- Department of Pediatric Dentistry, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yaghoub Yazdani
- Department of Medical Immunology, Faculty of Advanced Medical Science Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeed Mohammadi
- Department of Molecular Medicine, Faculty of Advanced Medical Science Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
27
|
The Role of Bacterial and Fungal Human Respiratory Microbiota in COVID-19 Patients. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6670798. [PMID: 33681368 PMCID: PMC7907751 DOI: 10.1155/2021/6670798] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/04/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023]
Abstract
Recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), has led to a worldwide pandemic with millions of infected patients. Alteration in humans' microbiota was also reported in COVID-19 patients. The alteration in human microbiota may contribute to bacterial or viral infections and affect the immune system. Moreover, human's microbiota can be altered due to SARS-CoV-2 infection, and these microbiota changes can indicate the progression of COVID-19. While current studies focus on the gut microbiota, it seems necessary to pay attention to the lung microbiota in COVID-19. This study is aimed at reviewing respiratory microbiota dysbiosis among COVID-19 patients to encourage further studies on the field for assessment of SARS-CoV-2 and respiratory microbiota interaction.
Collapse
|
28
|
Abstract
The coronavirus disease 2019 (COVID-19) pandemic was declared a public health emergency of international concern by the World Health Organization. COVID-19 has high transmissibility and could result in acute lung injury in a fraction of patients. By counterbalancing the activity of the renin-angiotensin system, angiotensin-converting enzyme 2, which is the fusion receptor of the virus, plays a protective role against the development of complications of this viral infection. Vitamin D can induce the expression of angiotensin-converting enzyme 2 and regulate the immune system through different mechanisms. Epidemiologic studies of the relationship between vitamin D and various respiratory infections were reviewed and, here, the postulated mechanisms and clinical data supporting the protective role of vitamin D against COVID-19-mediated complications are discussed.
Collapse
Affiliation(s)
- Fatemeh Hadizadeh
- Department of Bioinformatics, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
29
|
Pedan H, Janosova V, Hajtman A, Calkovsky V. Non-Reflex Defense Mechanisms of Upper Airway Mucosa: Possible Clinical Application. Physiol Res 2021; 69:S55-S67. [PMID: 32228012 DOI: 10.33549/physiolres.934404] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The sinonasal mucosa has an essential role in defense mechanisms of the upper respiratory tract. The innate immune system presents the primary defense against noxious microorganisms followed by induction of the adaptive immune mechanisms as a consequence of the presence of pathogens. This well-known activation of adaptive immune system in response to presence of the antigen on mucosal surfaces is now broadly applicated in vaccinology research. Prevention of infectious diseases belongs to substantial challenges in maintaining the population health. Non-invasive, easily applicable mucosal vaccination purposes various research opportunities that could be usable in daily practice. However, the existence of multiple limitations such as rapid clearance of vaccine from nasal mucosa by means of mucociliary transport represents a great challenge in development of safe and efficient vaccines. Here we give an updated view on nasal functions with focus on nasal mucosal immunity and its potential application in vaccination in nearly future.
Collapse
Affiliation(s)
- H Pedan
- Clinic of Otorhinolaryngology and Head and Neck Surgery, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, and Martin University Hospital, Martin, Slovak
| | | | | | | |
Collapse
|
30
|
Zhang L, Ghosh SK, Basavarajappa SC, Muller-Greven J, Penfield J, Brewer A, Ramakrishnan P, Buck M, Weinberg A. Molecular dynamics simulations and functional studies reveal that hBD-2 binds SARS-CoV-2 spike RBD and blocks viral entry into ACE2 expressing cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.07.425621. [PMID: 33442698 PMCID: PMC7805467 DOI: 10.1101/2021.01.07.425621] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
New approaches to complement vaccination are needed to combat the spread of SARS-CoV-2 and stop COVID-19 related deaths and long-term medical complications. Human beta defensin 2 (hBD-2) is a naturally occurring epithelial cell derived host defense peptide that has antiviral properties. Our comprehensive in-silico studies demonstrate that hBD-2 binds the site on the CoV-2-RBD that docks with the ACE2 receptor. Biophysical and biochemical assays confirm that hBD-2 indeed binds to the CoV-2-receptor binding domain (RBD) (KD ~ 300 nM), preventing it from binding to ACE2 expressing cells. Importantly, hBD-2 shows specificity by blocking CoV-2/spike pseudoviral infection, but not VSV-G mediated infection, of ACE2 expressing human cells with an IC50 of 2.4± 0.1 μM. These promising findings offer opportunities to develop hBD-2 and/or its derivatives and mimetics to safely and effectively use as novel agents to prevent SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Liqun Zhang
- Chemical Engineering, Tennessee Technological University, Cookeville, TN 38505
- contributed equally
| | - Santosh K. Ghosh
- Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH 44124
- contributed equally
| | - Shrikanth C. Basavarajappa
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44124
- contributed equally
| | - Jeannine Muller-Greven
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44124
| | - Jackson Penfield
- Chemical Engineering, Tennessee Technological University, Cookeville, TN 38505
| | - Ann Brewer
- Chemical Engineering, Tennessee Technological University, Cookeville, TN 38505
| | | | - Matthias Buck
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44124
| | - Aaron Weinberg
- Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH 44124
- Lead contact
| |
Collapse
|
31
|
Pinkerton JW, Kim RY, Koeninger L, Armbruster NS, Hansbro NG, Brown AC, Jayaraman R, Shen S, Malek N, Cooper MA, Nordkild P, Horvat JC, Jensen BAH, Wehkamp J, Hansbro PM. Human β-defensin-2 suppresses key features of asthma in murine models of allergic airways disease. Clin Exp Allergy 2020; 51:120-131. [PMID: 33098152 DOI: 10.1111/cea.13766] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/23/2020] [Accepted: 10/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Asthma is an airway inflammatory disease and a major health problem worldwide. Anti-inflammatory steroids and bronchodilators are the gold-standard therapy for asthma. However, they do not prevent the development of the disease, and critically, a subset of asthmatics are resistant to steroid therapy. OBJECTIVE To elucidate the therapeutic potential of human β-defensins (hBD), such as hBD2 mild to moderate and severe asthma. METHODS We investigated the role of hBD2 in a steroid-sensitive, house dust mite-induced allergic airways disease (AAD) model and a steroid-insensitive model combining ovalbumin-induced AAD with C muridarum (Cmu) respiratory infection. RESULTS In both models, we demonstrated that therapeutic intranasal application of hBD2 significantly reduced the influx of inflammatory cells into the bronchoalveolar lavage fluid. Furthermore, key type 2 asthma-related cytokines IL-9 and IL-13, as well as additional immunomodulating cytokines, were significantly decreased after administration of hBD2 in the steroid-sensitive model. The suppression of inflammation was associated with improvements in airway physiology and treatment also suppressed airway hyper-responsiveness (AHR) in terms of airway resistance and compliance to methacholine challenge. CONCLUSIONS AND CLINICAL RELEVANCE These data indicate that hBD2 reduces the hallmark features and has potential as a new therapeutic agent in allergic and especially steroid-resistant asthma.
Collapse
Affiliation(s)
- James W Pinkerton
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,National Heart & Lung Institute, Imperial College London, London, UK
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Louis Koeninger
- Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | | | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ranjith Jayaraman
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Sijie Shen
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Nisar Malek
- Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Matthew A Cooper
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Qld, Australia
| | - Peter Nordkild
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Benjamin A H Jensen
- Section for Human Genomics and Metagenomics in Metabolism, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Wehkamp
- Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, University of Newcastle, & Hunter Medical Research Institute, Newcastle, NSW, Australia.,Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
32
|
Alfonso García SL, Parada-Sanchez MT, Arboleda Toro D. The phenotype of gingival fibroblasts and their potential use in advanced therapies. Eur J Cell Biol 2020; 99:151123. [PMID: 33070040 DOI: 10.1016/j.ejcb.2020.151123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023] Open
Abstract
Advanced therapies in medicine use stem cells, gene editing, and tissues to treat a wide range of conditions. One of their goals is to stimulate endogenous repair of tissues and organs by manipulating stem cells and their niche, as well as to optimize the intrinsic characteristics and plasticity of differentiated cells in adult tissues. In this context, fibroblasts emerge as an alternative source to stem cells because they share phenotypic and regenerative characteristics. Specifically, fibroblasts of the oral mucosae have been shown to have improved regenerative capacity compared to other fibroblast populations. Additionally, their easy access by means of minimally invasive procedures without generating aesthetic problems, with easy and rapid in vitro expansion and with great capacity to respond to extrinsic factors, make oral fibroblasts an attractive and interesting resource for regenerative medicine. This review summarizes current concepts regarding the phenotypic and functional aspects of human Gingival Fibroblasts and their niche, differentiating them from other fibroblast populations of oral-lining mucosa and skin fibroblasts. Furthermore, some applications are presented in regenerative medicine, emphasizing on the biological potential of human Gingival Fibroblasts.
Collapse
Affiliation(s)
- Sandra Liliana Alfonso García
- Department of Integrated Basic Studies, Faculty of Dentistry, Universidad de Antioquia, Medellín, 050010, Colombia; Department of Oral Health, Faculty of Dentistry, Universidad Nacional de Colombia, Bogotá, 111311, Colombia.
| | | | - David Arboleda Toro
- Department of Integrated Basic Studies, Faculty of Dentistry, Universidad de Antioquia, Medellín, 050010, Colombia
| |
Collapse
|
33
|
Rosca AS, Castro J, Sousa LGV, Cerca N. Gardnerella and vaginal health: the truth is out there. FEMS Microbiol Rev 2020; 44:73-105. [PMID: 31697363 DOI: 10.1093/femsre/fuz027] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
The human vagina is a dynamic ecosystem in which homeostasis depends on mutually beneficial interactions between the host and their microorganisms. However, the vaginal ecosystem can be thrown off balance by a wide variety of factors. Bacterial vaginosis (BV) is the most common vaginal infection in women of childbearing age but its etiology is not yet fully understood, with different controversial theories being raised over the years. What is generally accepted is that BV is often characterized by a shift in the composition of the normal vaginal microbiota, from a Lactobacillus species dominated microbiota to a mixture of anaerobic and facultative anaerobic bacteria. During BV, a polymicrobial biofilm develops in the vaginal microenvironment, being mainly composed of Gardnerella species. The interactions between vaginal microorganisms are thought to play a pivotal role in the shift from health to disease and might also increase the risk of sexually transmitted infections acquisition. Here, we review the current knowledge regarding the specific interactions that occur in the vaginal niche and discuss mechanisms by which these interactions might be mediated. Furthermore, we discuss the importance of novel strategies to fight chronic vaginal infections.
Collapse
Affiliation(s)
- Aliona S Rosca
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Joana Castro
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Lúcia G V Sousa
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Nuno Cerca
- Centre of Biological Engineering (CEB), Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
34
|
Berry SB, Haack AJ, Theberge AB, Brighenti S, Svensson M. Host and Pathogen Communication in the Respiratory Tract: Mechanisms and Models of a Complex Signaling Microenvironment. Front Med (Lausanne) 2020; 7:537. [PMID: 33015094 PMCID: PMC7511576 DOI: 10.3389/fmed.2020.00537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/29/2020] [Indexed: 01/15/2023] Open
Abstract
Chronic lung diseases are a leading cause of morbidity and mortality across the globe, encompassing a diverse range of conditions from infections with pathogenic microorganisms to underlying genetic disorders. The respiratory tract represents an active interface with the external environment having the primary immune function of resisting pathogen intrusion and maintaining homeostasis in response to the myriad of stimuli encountered within its microenvironment. To perform these vital functions and prevent lung disorders, a chemical and biological cross-talk occurs in the complex milieu of the lung that mediates and regulates the numerous cellular processes contributing to lung health. In this review, we will focus on the role of cross-talk in chronic lung infections, and discuss how different cell types and signaling pathways contribute to the chronicity of infection(s) and prevent effective immune clearance of pathogens. In the lung microenvironment, pathogens have developed the capacity to evade mucosal immunity using different mechanisms or virulence factors, leading to colonization and infection of the host; such mechanisms include the release of soluble and volatile factors, as well as contact dependent (juxtracrine) interactions. We explore the diverse modes of communication between the host and pathogen in the lung tissue milieu in the context of chronic lung infections. Lastly, we review current methods and approaches used to model and study these host-pathogen interactions in vitro, and the role of these technological platforms in advancing our knowledge about chronic lung diseases.
Collapse
Affiliation(s)
- Samuel B. Berry
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Amanda J. Haack
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | | | - Susanna Brighenti
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Hosseiniyan Khatibi SM, Kheyrolahzadeh K, Barzegari A, Rahbar Saadat Y, Zununi Vahed S. Medicinal signaling cells: A potential antimicrobial drug store. J Cell Physiol 2020; 235:7731-7746. [PMID: 32352173 DOI: 10.1002/jcp.29728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/06/2020] [Accepted: 04/12/2020] [Indexed: 12/11/2022]
Abstract
Medicinal signaling cells (MSCs) are multipotent cells derived from mammalian bone marrow and periosteum that can be extended in culture. They can keep their ability in vitro to form a variety of mesodermal phenotypes and tissues. Over recent years, there has been great attention over MSCs since they can impact the organ transplantation as well as autoimmune and bacterial diseases. MSCs can secrete different bioactive factors such as growth factors, antimicrobial peptides/proteins and cytokines that can suppress the immune system and prevent infection via direct and indirect mechanisms. Moreover, MSCs are able to increase bacterial clearance in sepsis models by producing antimicrobial peptides such as defensins, cathelicidins, lipocalin and hepcidin. It is the aim of the present review to focus on the antibacterial effector functions of MSCs and their mechanisms of action against the pathogenic microbes.
Collapse
Affiliation(s)
| | - Keyvan Kheyrolahzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Azad University, Tabriz Branch, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Rahbar Saadat
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
36
|
Zsila F, Beke-Somfai T. Human host-defense peptide LL-37 targets stealth siderophores. Biochem Biophys Res Commun 2020; 526:780-785. [PMID: 32265033 DOI: 10.1016/j.bbrc.2020.03.162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/28/2020] [Indexed: 02/02/2023]
Abstract
A growing number of evidence shows that human-associated microbiota is an important contributor in health and disease. However, much of the complexity of host-microbiota interaction remains to be elucidated both at cellular and molecular levels. Siderophores are chemically diverse, ferric-specific chelators synthesized and secreted by microbes to secure their iron acquisition. The host defense peptide LL-37 is ubiquitously produced at epithelial surfaces modulating microbial communities and suppressing pathogenic strains. The present work demonstrates that LL-37 binds tightly siderocalin-resistant stealth siderophores which are important contributors to the virulence of several pathogens. As indicated by circular dichroism spectroscopic experiments, addition of aerobactin and rhizoferrin increases the membrane active α-helical conformation of the partially folded peptide. The cationic nature of LL-37 (+6 net charge at pH 7.4) and the multiple carboxylate groups present in siderophores refer to the dominant contribution of electrostatic interactions in the stabilization of peptide-chelator adducts. It is proposed that aside siderocalin proteins, LL-37 may be a complementary, less specific component of the siderophore scavenging repertoire of the innate immune system.
Collapse
Affiliation(s)
- Ferenc Zsila
- Biomolecular Self-Assembly Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, P.O. Box 286, H-1519, Budapest, Hungary.
| | - Tamás Beke-Somfai
- Biomolecular Self-Assembly Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, P.O. Box 286, H-1519, Budapest, Hungary
| |
Collapse
|
37
|
Abstract
Aim: Supplemental oxygen is often used to treat neonates with respiratory disorders. Human and animal studies have demonstrated that neonatal hyperoxia increases oxidative stress and induces damage and collagen deposition in kidney during the perinatal period. Cathelicidin LL-37 is one important group of human antimicrobial peptides which exhibits antioxidant activity and its overexpression resists hyperoxia-induced oxidative stress. This study was designed to evaluate the protective effects of cathelicidin in hyperoxia-induced kidney injury in newborn rats. Methods: Sprague-Dawley rat pups were reared in either room air (RA) or hyperoxia (85% O2) and were randomly treated with low-dose (4 mg/kg) and high-dose (8 mg/kg) cathelicidin in normal saline (NS) administered intraperitoneally on postnatal days 1–6. The following six groups were obtained: RA + NS, RA + low-dose cathelicidin, RA + high-dose cathelicidin, O2 + NS, O2 + low-dose cathelicidin, and O2 + high-dose cathelicidin. Kidneys were taken for Western blot and histological analyses on postnatal day 7. Results: The hyperoxia-reared rats exhibited significantly lower body weights and anti-inflammatory M2 macrophages, but the kidney injury scores, oxidative stress marker 8-hydroxy-2'-deoxyguanosine (8-OHdG)-positive cells, pro-inflammatory M1 macrophages, collagen deposition, and NF-κB expression were higher than did the RA-reared rats. Conclusions: Cathelicidin treatment attenuated kidney injury as evidenced by lower kidney injury scores, 8-OHdG-positive cells, collagen deposition, and reversion of hyperoxia-induced M1/M2 macrophage polarization. The role of Cathelicidin in ameliorates kidney injury of the hyperoxia newborn rats was accompanied by decreased NF-κB expression, which probably through the modulating NF-κB activity in the kidney.
Collapse
Affiliation(s)
- Hsiu-Chu Chou
- Department of Anatomy and Cellular Biology, School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
38
|
Liu Q, Liu Q, Meng H, Lv H, Liu Y, Liu J, Wang H, He L, Qin J, Wang Y, Dai Y, Otto M, Li M. Staphylococcus epidermidis Contributes to Healthy Maturation of the Nasal Microbiome by Stimulating Antimicrobial Peptide Production. Cell Host Microbe 2020; 27:68-78.e5. [PMID: 31866425 PMCID: PMC10988655 DOI: 10.1016/j.chom.2019.11.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 08/19/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022]
Abstract
The composition of the human microbiome profoundly impacts human well-being. However, the mechanisms underlying microbiome maturation are poorly understood. The nasal microbiome is of particular importance as a source of many respiratory infections. Here, we performed a large sequencing and culture-based analysis of the human nasal microbiota from different age groups. We observed a significant decline of pathogenic bacteria before adulthood, with an increase of the commensal Staphylococcus epidermidis. In seniors, this effect was partially reversed. In vitro, many S. epidermidis isolates stimulated nasal epithelia to produce antimicrobial peptides, killing pathogenic competitors, while S. epidermidis itself proved highly resistant owing to its exceptional capacity to form biofilms. Furthermore, S. epidermidis isolates with high antimicrobial peptide-inducing and biofilm-forming capacities outcompeted pathogenic bacteria during nasal colonization in vivo. Our study identifies a pivotal role of S. epidermidis in healthy maturation of the nasal microbiome, which is achieved at least in part by symbiotic cooperation with innate host defense.
Collapse
Affiliation(s)
- Qian Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Qingyun Liu
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China
| | - Hongwei Meng
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Huiying Lv
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yao Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Junlan Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Hua Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Lei He
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Juanxiu Qin
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yanan Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yingxin Dai
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD 20814, USA.
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| |
Collapse
|
39
|
Bicker KL, Cobb SL. Recent advances in the development of anti-infective peptoids. Chem Commun (Camb) 2020; 56:11158-11168. [DOI: 10.1039/d0cc04704j] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This feature article highlights the progress that has been made towards the development of novel anti-infective peptoids and the key areas for future development within this field.
Collapse
Affiliation(s)
- Kevin L. Bicker
- Department of Chemistry
- Middle Tennessee State University
- Murfreesboro
- USA
| | - Steven L Cobb
- Deparment of Chemistry
- Biophysical Sciences Institute
- Durham University
- Durham
- UK
| |
Collapse
|
40
|
Tatsuta M, Kan-O K, Ishii Y, Yamamoto N, Ogawa T, Fukuyama S, Ogawa A, Fujita A, Nakanishi Y, Matsumoto K. Effects of cigarette smoke on barrier function and tight junction proteins in the bronchial epithelium: protective role of cathelicidin LL-37. Respir Res 2019; 20:251. [PMID: 31706310 PMCID: PMC6842552 DOI: 10.1186/s12931-019-1226-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/31/2019] [Indexed: 11/10/2022] Open
Abstract
Background Airway epithelial barrier function is maintained by the formation of tight junctions (TJs) and adherens junctions (AJs). Inhalation of cigarette smoke causes airway epithelial barrier dysfunction and may contribute to the pathogenesis of chronic lung diseases such as asthma and chronic obstructive pulmonary disease (COPD). We assessed the effects of cigarette smoke on barrier function and expression of multiple TJ and AJ proteins in the bronchial epithelium. We also examined whether treatment with glucocorticosteroids (GCSs), long-acting β2-agonists (LABAs), and human cathelicidin LL-37 can protect against cigarette smoke extract (CSE)-induced barrier dysfunction. Methods Calu-3 cells cultured at the air-liquid interface were pretreated with or without GCSs, LABAs, GCSs plus LABAs, or LL-37, and subsequently exposed to CSE. Barrier function was assessed by transepithelial electronic resistance (TEER) measurements. Gene and protein expression levels of TJ and AJ proteins were analyzed by quantitative PCR and western blotting, respectively. Immunofluorescence staining of TJ and AJ proteins was performed. Results CSE decreased TEER and increased permeability in a concentration-dependent manner. CSE suppressed gene expression of claudin-1, claudin-3, claudin-4, claudin-7, claudin-15, occludin, E-cadherin, junctional adhesion molecule-A (JAM-A) and zonula occludens-1 (ZO-1) within 12 h post-CSE exposure, while suppressed protein expression levels of occludin at 12 h. CSE-treated cells exhibited discontinuous or attenuated immunostaining for claudin-1, claudin-3, claudin-4, occludin, ZO-1, and E-cadherin compared with untreated cells. GCS treatment partially restored CSE-induced TEER reduction, while LABA treatment had no effect. GCS and LABA combination treatment had no additive effect on CSE-induced TEER reduction and gene suppression of TJ and AJ proteins. Human cathelicidin LL-37 counteracted CSE-induced TEER reduction and prevented disruption of occludin and ZO-1. LL-37 also attenuated CSE-induced decreases in gene and protein expression levels of occludin. Conclusions CSE caused airway epithelial barrier dysfunction and simultaneously downregulated multiple TJ and AJ proteins. GCS and LABA combination treatment had no additive effect on CSE-induced TEER reduction. LL-37 counteracted CSE-induced TEER reduction and prevented disruption of occludin and ZO-1. Use of LL-37 to counteract airway epithelial barrier dysfunction may have significant benefits for respiratory diseases such as asthma and COPD.
Collapse
Affiliation(s)
- Miyoko Tatsuta
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Respiratory Medicine, National Hospital Organization Omuta National Hospital, Fukuoka, 837-0911, Japan
| | - Keiko Kan-O
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Department of Endoscopic Diagnostics and Therapeutics, Kyushu University Hospital, Fukuoka, 812-8582, Japan.
| | - Yumiko Ishii
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Norio Yamamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Ogawa
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoru Fukuyama
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Aimi Ogawa
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akitaka Fujita
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoichi Nakanishi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koichiro Matsumoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
41
|
Esfandiyari R, Halabian R, Behzadi E, Sedighian H, Jafari R, Imani Fooladi AA. Performance evaluation of antimicrobial peptide ll-37 and hepcidin and β-defensin-2 secreted by mesenchymal stem cells. Heliyon 2019; 5:e02652. [PMID: 31687504 PMCID: PMC6820248 DOI: 10.1016/j.heliyon.2019.e02652] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/02/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
Peptides are secreted by different cell types and are trendy therapeutic agents that have attracted attention for the treatment of several diseases such as infections. Antimicrobial peptides exert various mechanisms such as changing cell membrane permeability which leads to inhibition or death of bacterial cells. mesenchymal stem cells (MSCs) are key to produce antimicrobial peptides and to inhibit the growth of pathogens. These cells have been shown to be capable of producing antimicrobial peptides upon exposure to different bacteria. As a result, antimicrobial peptides can be considered as novel agents for the treatment of infectious diseases. The purpose of this review was to investigate the targets and mechanisms of antimicrobial peptides secreted by MSCs.
Collapse
Affiliation(s)
- Reza Esfandiyari
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ramezan Jafari
- Department of Radiology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Casaroto AR, da Silva RA, Salmeron S, Rezende MLRD, Dionísio TJ, Santos CFD, Pinke KH, Klingbeil MFG, Salomão PA, Lopes MMR, Lara VS. Candida albicans-Cell Interactions Activate Innate Immune Defense in Human Palate Epithelial Primary Cells via Nitric Oxide (NO) and β-Defensin 2 (hBD-2). Cells 2019; 8:cells8070707. [PMID: 31336838 PMCID: PMC6678605 DOI: 10.3390/cells8070707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/28/2019] [Accepted: 07/05/2019] [Indexed: 01/29/2023] Open
Abstract
The presence of Candida albicans in the biofilm underlying the dental prosthesis is related to denture stomatitis (DS), an inflammatory reaction of the oral mucosa. The oral epithelium, a component of the innate immune response, has the ability to react to fungal invasion. In this study, we evaluated the in vitro effect of viable C. albicans on the apoptosis, nitric oxide (NO) production, and β-defensin 2 (hBD-2) expression and production of human palate epithelial cells (HPECs). We further determined whether or not these effects were correlated with fungal invasion of epithelial cells. Interaction between HPEC primary culture and C. albicans was obtained through either direct or indirect cell–cell contact with a supernatant from a hyphal fungus. We found that the hyphae supernatants were sufficient to induce slight HPEC apoptosis, which occurred prior to the activation of the specific mechanisms of epithelial defense. The epithelial defense responses were found to occur via NO and antimicrobial peptide hBD-2 production only during direct contact between C. albicans and HPECs and coincided with the fungus’s intraepithelial invasion. However, although the hBD-2 levels remained constant in the HPEC supernatants over time, the NO release and hBD-2 gene expression were reduced at a later time (10 h), indicating that the epithelial defense capacity against the fungal invasion was not maintained in later phases. This aspect of the immune response was associated with increased epithelial invasion and apoptosis maintenance.
Collapse
Affiliation(s)
- Ana Regina Casaroto
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil.
| | - Rafaela Alves da Silva
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Samira Salmeron
- Department of Prosthodontics and Periodontology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Maria Lúcia Rubo de Rezende
- Department of Prosthodontics and Periodontology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Thiago José Dionísio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Carlos Ferreira Dos Santos
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Karen Henriette Pinke
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | | | - Priscila Aranda Salomão
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Marcelo Milanda Ribeiro Lopes
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| | - Vanessa Soares Lara
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, 17012-901 Bauru, SP, Brazil
| |
Collapse
|
43
|
Chou HC, Chen CM. Cathelicidin attenuates hyperoxia-induced intestinal injury through inhibition of NF-κB activity in newborn rats. Exp Mol Pathol 2019; 113:104269. [PMID: 31202738 DOI: 10.1016/j.yexmp.2019.104269] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023]
Abstract
Supplemental oxygen is often used to treat neonates with respiratory disorders. Preclinical studies have demonstrated that neonatal hyperoxia injures the distal small intestine and activates nuclear factor-κB (NF-κB). Cathelicidin inhibits NF-κB activity and ameliorates lipopolysaccharide-induced intestinal barrier disruption in rats. Sprague-Dawley rat pups were reared in either room air (RA) or hyperoxia (85% O2) and were randomly treated with low-dose cathelicidin (4 mg/kg, LDC) and high-dose cathelicidin (HDC, 8 mg/kg) in 0.05 mL of normal saline (NS) administered intraperitoneally on postnatal days 1-6. The following six groups were obtained: RA + NS, RA + LDC, RA + HDC, O2 + NS, O2 + LDC, and O2 + HDC. The animals were sacrificed and the terminal ileum was removed for Western blot and histological analyses on postnatal day 7. The hyperoxia-reared rats exhibited significantly lower body weights, higher intestinal injury scores, lower occludin and ZO-1 expression, higher intestinal permeability and inducible IκB kinase inhibitor (IKKi) and NF-κB expression than the RA-reared rats. Cathelicidin treatment attenuated intestinal injury as evidenced by lower intestinal injury scores and intestinal permeability and higher intestinal barrier protein expression. The decrease in intestinal injury was accompanied by a decrease in IKKi and NF-κB. Cathelicidin attenuated hyperoxia-induced intestinal injury in the newborn rats, likely through NF-κB activity inhibition.
Collapse
Affiliation(s)
- Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan, ROC; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
44
|
Hall JW, Lima BP, Herbomel GG, Gopinath T, McDonald L, Shyne MT, Lee JK, Kreth J, Ross KF, Veglia G, Herzberg MC. An intramembrane sensory circuit monitors sortase A-mediated processing of streptococcal adhesins. Sci Signal 2019; 12:12/580/eaas9941. [PMID: 31064885 DOI: 10.1126/scisignal.aas9941] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial adhesins mediate adhesion to substrates and biofilm formation. Adhesins of the LPXTG family are posttranslationally processed by the cell membrane-localized peptidase sortase A, which cleaves the LPXTG motif. This generates a short C-terminal peptide (C-pep) that remains in the cell membrane, whereas the mature adhesin is incorporated into the cell wall. Genes encoding adhesins of the oral bacterium Streptococcus gordonii were differentially expressed depending on whether the bacteria were isolated from saliva or dental plaque and appeared to be coordinately regulated. Deletion of sspA and sspB (sspAB), both of which encode LPXTG-containing adhesins, unexpectedly enhanced adhesion and biofilm formation. C-peps produced from a model LPXTG-containing adhesin localized to the cell membrane and bound to and inhibited the intramembrane sensor histidine kinase SGO_1180, thus preventing activation of the cognate response regulator SGO_1181. The absence of SspAB C-peps induced the expression of the scaCBA operon encoding the lipoprotein adhesin ScaA, which was sufficient to preserve and even enhance biofilm formation. This C-pep-driven regulatory circuit also exists in pathogenic streptococci and is likely conserved among Gram-positive bacteria. This quality control mechanism ensures that the bacteria can form biofilms under diverse environmental conditions and may play a role in optimizing adhesion and biofilm formation.
Collapse
Affiliation(s)
- Jeffrey W Hall
- Department of Biological and Diagnostic Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bruno P Lima
- Department of Biological and Diagnostic Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Tata Gopinath
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - LeAnna McDonald
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael T Shyne
- Biostatistical Design and Analysis Center (BDAC), Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - John K Lee
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jens Kreth
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, OR 97239, USA
| | - Karen F Ross
- Department of Biological and Diagnostic Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark C Herzberg
- Department of Biological and Diagnostic Sciences, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
45
|
Neopane P, Paudel D, Yoshida K, Raj Adhikari B, Morikawa T, Onishi A, Hiraki D, Uehara O, Sato J, Nishimura M, Chiba I, Abiko Y. Immunohistochemical Localization of RNase 7 in Normal and Inflamed Oral Epithelia and Salivary Glands. Acta Histochem Cytochem 2019; 52:35-43. [PMID: 31341339 PMCID: PMC6643094 DOI: 10.1267/ahc.18027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/31/2019] [Indexed: 11/22/2022] Open
Abstract
RNase 7 is a skin-derived antimicrobial peptide expressed in various epithelial tissues. It is upregulated by stimulation with microbes and pro-inflammatory cytokines. Herein, we examined the expression levels of RNase 7 in tissues from normal and inflamed oral epithelia and salivary glands via immunohistochemistry. RNase 7 was expressed mainly in the surface layers of the parakeratinized and orthokeratinized oral epithelium. In addition, it was strongly and weakly expressed in oral lichen planus and radicular cysts, respectively. RNase 7 was constitutively expressed in salivary glands, particularly in the serous and duct cells. In the case of Sjogren’s syndrome, RNase 7 was strongly expressed in serous, ductal, and mucous cells in areas with lymphocytic infiltration. The localization patterns of RNase 7 were similar to those of other epithelial antimicrobial peptides, including beta-defensins. Thus, epithelial antimicrobial peptides may act against microbial infections in a coordinated manner in oral epithelia and salivary glands.
Collapse
Affiliation(s)
- Puja Neopane
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Durga Paudel
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Koki Yoshida
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Bhoj Raj Adhikari
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Tetsuro Morikawa
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Aya Onishi
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Daichi Hiraki
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Osamu Uehara
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido
| | - Jun Sato
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Michiko Nishimura
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Itsuo Chiba
- Division of Disease Control and Molecular Epidemiology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido
| | - Yoshihiro Abiko
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| |
Collapse
|
46
|
Antimicrobial Host Defence Peptides: Immunomodulatory Functions and Translational Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:149-171. [DOI: 10.1007/978-981-13-3588-4_10] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Pang M, Zhu M, Lei X, Xu P, Cheng B. Microbiome Imbalances: An Overlooked Potential Mechanism in Chronic Nonhealing Wounds. INT J LOW EXTR WOUND 2019; 18:31-41. [PMID: 30836811 DOI: 10.1177/1534734619832754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic nonhealing wounds are a severe burden to health care systems worldwide, causing millions of patients to have lengthy hospital stays, high health care costs, periods of unemployment, and reduced quality of life. Moreover, treating chronic nonhealing wounds effectively and reasonably in countries with limited medical resources can be extremely challenging. With many outstanding questions surrounding chronic nonhealing wounds, in this review, we offer changes to the microbiome as a potentially ignored mechanism important in the formation and treatment of chronic wounds. Our analysis helps bring a whole new understanding to wound formation and healing and provides a potential breakthrough in the treatment of chronic nonhealing wounds in the future.
Collapse
Affiliation(s)
- Mengru Pang
- The Graduate School of Southern Medical University, Guangzhou, China
- General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Meishu Zhu
- The Graduate School of Southern Medical University, Guangzhou, China
- The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaoxuan Lei
- The Graduate School of Southern Medical University, Guangzhou, China
- General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Pengcheng Xu
- General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Biao Cheng
- The Graduate School of Southern Medical University, Guangzhou, China
- General Hospital of Southern Theater Command, PLA, Guangzhou, China
- The Key Laboratory of Trauma Treatment and Tissue Repair of Tropical Area, PLA, Guangzhou, China
| |
Collapse
|
48
|
Carbohydrate-Dependent and Antimicrobial Peptide Defence Mechanisms Against Helicobacter pylori Infections. Curr Top Microbiol Immunol 2019; 421:179-207. [PMID: 31123890 DOI: 10.1007/978-3-030-15138-6_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human stomach is a harsh and fluctuating environment for bacteria with hazards such as gastric acid and flow through of gastric contents into the intestine. H. pylori gains admission to a stable niche with nutrient access from exudates when attached to the epithelial cells under the mucus layer, whereof adherence to glycolipids and other factors provides stable and intimate attachment. To reach this niche, H. pylori must overcome mucosal defence mechanisms including the continuously secreted mucus layer, which provides several layers of defence: (1) mucins in the mucus layer can bind H. pylori and transport it away from the gastric niche with the gastric emptying, (2) mucins can inhibit H. pylori growth, both via glycans that can have antibiotic like function and via an aggregation-dependent mechanism, (3) antimicrobial peptides (AMPs) have antimicrobial activity and are retained in a strategic position in the mucus layer and (4) underneath the mucus layer, the membrane-bound mucins provide a second barrier, and can function as releasable decoys. Many of these functions are dependent on H. pylori interactions with host glycan structures, and both the host glycosylation and concentration of antimicrobial peptides change with infection and inflammation, making these interactions dynamic. Here, we review our current understanding of mucin glycan and antimicrobial peptide-dependent host defence mechanisms against H. pylori infection.
Collapse
|
49
|
Serum concentrations of antimicrobial peptide cathelicidin LL-37 in patients with bacterial lung infections. Cent Eur J Immunol 2018; 43:453-457. [PMID: 30799994 PMCID: PMC6384432 DOI: 10.5114/ceji.2018.81355] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/12/2017] [Indexed: 11/30/2022] Open
Abstract
Nowadays, data indicate that antimicrobial peptides play an important role in immunological defense. Human cathelicidin LL-37 possesses a broad spectrum of antimicrobial properties against Gram-positive and Gram-negative bacteria, and is thereby an important component of defense mechanisms within the respiratory tract. In this study, we determined the LL-37 serum level in patients with pneumonia caused by different bacteria species in comparison with healthy subjects. Twenty-two patients with pneumonia caused by coccal Gram-positive bacteria (I), 16 patients with pneumonia caused by Haemophilus influenzae (II), 29 patients with pneumonia caused by members of the Enterobacteriaceae (III), 13 patients caused by non-fermenting Gram-negative bacteria (IV), and 30 healthy controls were enrolled in the study. Serum LL-37 concentration was measured using an enzyme-linked immunosorbent assay (ELISA). The mean LL-37 concentration in pneumonia patients was significantly higher in group I (p = 0.0032), group II (p = 0.0022), and group III (p = 0.019), and significantly lower in group IV (p = 0.000004) as compared with healthy volunteers. Our data suggest that LL-37 plays an important role in defense mechanisms during pneumonia. The reduced level of this peptide in subjects with pneumonia caused by opportunistic bacteria may reflect weakened immune system reactivity in these patients.
Collapse
|
50
|
Pan WL, Wang Y, Hao Y, Wong JH, Chan WC, Wan DCC, Ng TB. Overexpression of CXCR4 synergizes with LL-37 in the metastasis of breast cancer cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3837-3846. [DOI: 10.1016/j.bbadis.2018.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/20/2018] [Accepted: 09/08/2018] [Indexed: 01/14/2023]
|