1
|
Shamim A, Abdul Aziz M, Saeed F, Kumari R, Mary Joseph A, Ponnachan P, Kishore U, Masmoudi K. Revisiting surfactant protein D: an immune surveillance molecule bridging innate and adaptive immunity. Front Immunol 2024; 15:1491175. [PMID: 39742280 PMCID: PMC11685232 DOI: 10.3389/fimmu.2024.1491175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/12/2024] [Indexed: 01/03/2025] Open
Abstract
Surfactant protein D (SP-D) is a C-type lectin that was originally discovered as a lung surfactant associated phospholipid recognising protein. It was originally shown to be of great importance in surfactant turnover and homeostasis in conjunction with another hydrophilic surfactant protein i.e. SP-A. In addition, it was found to agglutinate bacteria in suspension and likely a key defence molecule in the lungs. Since its early days of characterization in 1990s, SP-D has turned out to be a central player in the mucosal immunity as pulmonary as well as extrapulmonary innate immune molecule. The most exciting development has been characterization of its C-type lectin or carbohydrate recognition domain (CRDs) that exists in a homotrimeric form in native as well as recombinant versions. SP-D has a range of strategies to recognise pathogen-associated molecular patterns (PAMPs) and thus act as a soluble PAMP-recognizing receptor (PRR), and subsequent destruction of the pathogens directly, or indirectly via phagocytic cells. SP-D also recognizes a range of allergens, competes out with specific IgE antibodies, and downregulates histamine release by basophils and mast cells. These anti-microbial and anti-allergic properties of SP-D have been validated by in vivo murine models of infection and allergy. The SP-D gene deficient mice exhibit remarkable phenotypes where lungs are leaky, showing features of fibrosis and emphysema. One of the seminal discoveries in the field has been the observation that activated eosinophils (and other immune cells) can be induced into apoptotic pathways by SP-D. This raised the possibility that SP-D can be an innate immune surveillance molecule. Studies have revealed the ability of a recombinant fragment of human SP-D containing homotrimeric neck and CRD region to induce apoptosis via intrinsic as well as extrinsic pathways; in addition, it also seems capable of interfering with epithelial-to-mesenchymal transition. These studies have opened up enormous possibilities for setting up pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Azra Shamim
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mughair Abdul Aziz
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Faryal Saeed
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rekha Kumari
- Department of Zoology, A.N College, Patliputra University, Patna, Bihar, India
| | - Ann Mary Joseph
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Pretty Ponnachan
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Khaled Masmoudi
- Department Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
2
|
Akhlaghipour I, Moghbeli M. Matrix metalloproteinases as the critical regulators of cisplatin response and tumor cell invasion. Eur J Pharmacol 2024; 982:176966. [PMID: 39216742 DOI: 10.1016/j.ejphar.2024.176966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Cisplatin (CDDP) as one of the most common first-line chemotherapy drugs plays a vital role in the treatment of a wide range of malignant tumors. Nevertheless, CDDP resistance is observed as a therapeutic challenge in a large number of cancer patients. Considering the CDDP side effects in normal tissues, predicting the CDDP response of cancer patients can significantly help to choose the appropriate therapeutic strategy. In this regard, investigating the molecular mechanisms involved in CDDP resistance can lead to the introduction of prognostic markers in cancer patients. Matrix metalloproteinases (MMPs) have critical roles in tissue remodeling and cell migration through extracellular matrix degradation. Therefore, defects in MMPs functions can be associated with tumor metastasis and chemo resistance. In the present review, we discussed the role of MMPs in CDDP response and tumor cell invasion. PubMed, Scopus, Google Scholar, and Web of Science were searched using "MMP", "cisplatin", and "cancer" keywords for data retrieval that was limited to Apr 20, 2024. It has been reported that MMPs can increase CDDP resistance in tumor cells as the effectors of PI3K/AKT, MAPK, and NF-κB signaling pathways or independently through the regulation of structural proteins, autophagy, and epithelial-to-mesenchymal transition (EMT) process. This review has an effective role in introducing MMPs as the prognostic markers and therapeutic targets in CDDP-resistant cancer patients.
Collapse
Affiliation(s)
- Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Pérez-Polo S, Mena AR, Barros L, Borrajo P, Pazos M, Carrera M, Gestal C. Decoding Octopus Skin Mucus: Impact of Aquarium-Maintenance and Senescence on the Proteome Profile of the Common Octopus ( Octopus vulgaris). Int J Mol Sci 2024; 25:9953. [PMID: 39337441 PMCID: PMC11431876 DOI: 10.3390/ijms25189953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The common octopus (Octopus vulgaris) is an excellent candidate for aquaculture diversification, due to its biological traits and high market demand. To ensure a high-quality product while maintaining welfare in captive environments, it is crucial to develop non-invasive methods for testing health biomarkers. Proteins found in skin mucus offer a non-invasive approach to monitoring octopus welfare. This study compares the protein profiles in the skin mucus of wild, aquarium-maintained, and senescent specimens to identify welfare biomarkers. A tandem mass tag (TMT) coupled with an Orbitrap Eclipse Tribrid mass spectrometer was used to create a reference dataset from octopus skin mucus, identifying 1496 non-redundant protein groups. Although similar profiles were observed, differences in relative abundances led to the identification of potential biomarkers, including caspase-3-like, protocadherin 4, deleted in malignant brain tumors, thioredoxin, papilin, annexin, cofilin and mucin-4 proteins. Some of these proteins also revealed potential as bioactive peptides. This investigation provides the most extensive analysis of the skin mucus proteome in the common octopus and is the first to explore how aquarium maintenance and senescence alter the mucus proteome. This research highlights the potential of skin mucus protein/peptides as non-invasive monitoring biomarkers in cultured animals.
Collapse
Affiliation(s)
| | | | | | | | | | - Mónica Carrera
- Instituto de Investigaciones Marinas (IIM-CSIC), Spanish National Research Council (CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (S.P.-P.); (A.R.M.); (L.B.); (P.B.); (M.P.)
| | - Camino Gestal
- Instituto de Investigaciones Marinas (IIM-CSIC), Spanish National Research Council (CSIC), Eduardo Cabello 6, 36208 Vigo, Spain; (S.P.-P.); (A.R.M.); (L.B.); (P.B.); (M.P.)
| |
Collapse
|
4
|
da Silva MI, Ott T. Effects of conceptus proteins on endometrium and blood leukocytes of dairy cattle using transcriptome and meta-analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591148. [PMID: 38712302 PMCID: PMC11071483 DOI: 10.1101/2024.04.25.591148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
This study investigates the short and long-term effects of IFNT and PAG on the transcriptome of endometrium and blood leukocytes. Holstein heifers received intrauterine infusions of one of the following treatments: 20 mL of a 200 μg/mL bovine serum albumin solution (BSA; vehicle) from day 14 to 16 of the estrous cycle (BSA), vehicle + 10 μg/mL of IFNT from day 14 to 16 (IFNT3), vehicle + 10 μg/mL of IFNT from day 14 to 19 (IFNT6), and vehicle + 10 μg/mL of IFNT from day 14 to 16 followed by vehicle + 10 μg/mL of IFNT + 5 μg/mL of PAG from day 17 to 19 (IFNT+PAG). RNA-seq analysis was performed in endometrial biopsies and blood leukocytes collected after treatments. Acute IFNT signaling in the endometrium (IFNT3 vs BSA), induced differentially expressed genes (DEG) associated with interferon activation, immune response, inflammation, cell death, and inhibited vesicle transport and extracellular matrix remodeling. Prolonged IFNT signaling (IFNT6 vs IFNT3) altered gene expression related to cell invasion, retinoic acid signaling, and embryo implantation. In contrast, PAG induced numerous DEG in blood leukocytes but only 4 DEG in the endometrium. In blood leukocytes, PAG stimulated genes involved in development and TGFB signaling while inhibiting interferon signaling and cell migration. Overall, IFNT is a primary regulator of endometrial gene expression, while PAG predominantly affected the transcriptome of circulating immune cells during early pregnancy. Further research is essential to fully grasp the roles of identified DEG in both the endometrium and blood leukocytes.
Collapse
Affiliation(s)
- Maria Isabel da Silva
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Troy Ott
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
5
|
Abbondio M, Tanca A, De Diego L, Sau R, Bibbò S, Pes GM, Dore MP, Uzzau S. Metaproteomic assessment of gut microbial and host functional perturbations in Helicobacter pylori-infected patients subjected to an antimicrobial protocol. Gut Microbes 2023; 15:2291170. [PMID: 38063474 PMCID: PMC10730194 DOI: 10.1080/19490976.2023.2291170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
The impact of therapeutic interventions on the human gut microbiota (GM) is a clinical issue of paramount interest given the strong interconnection between microbial dynamics and human health. Orally administered antibiotics are known to reduce GM biomass and modify GM taxonomic profile. However, the impact of antimicrobial therapies on GM functions and biochemical pathways has scarcely been studied. Here, we characterized the fecal metaproteome of 10 Helicobacter pylori-infected patients before (T0) and after 10 days (T1) of a successful quadruple therapy (bismuth, tetracycline, metronidazole, and rabeprazole) and 30 days after therapy cessation (T2), to investigate how GM and host functions change during the eradication and healing processes. At T1, the abundance ratio between microbial and host proteins was reversed compared with that at T0 and T2. Several pathobionts (including Klebsiella, Proteus, Enterococcus, Muribaculum, and Enterocloster) were increased at T1. Therapy reshaped the relative contributions of the functions required to produce acetate, propionate, and butyrate. Proteins related to the uptake and processing of complex glycans were increased. Microbial cross-feeding with sialic acid, fucose, and rhamnose was enhanced, whereas hydrogen sulfide production was reduced. Finally, microbial proteins involved in antibiotic resistance and inflammation were more abundant after therapy. Moreover, a reduction in host proteins with known roles in inflammation and H. pylori-mediated carcinogenesis was observed. In conclusion, our results support the use of metaproteomics to monitor drug-induced remodeling of GM and host functions, opening the way for investigating new antimicrobial therapies aimed at preserving gut environmental homeostasis.
Collapse
Affiliation(s)
- Marcello Abbondio
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandro Tanca
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Laura De Diego
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Rosangela Sau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Stefano Bibbò
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Giovanni Mario Pes
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Maria Pina Dore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Sergio Uzzau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
6
|
Medeiros MCD, The S, Bellile E, Russo N, Schmitd L, Danella E, Singh P, Banerjee R, Bassis C, Murphy GR, Sartor MA, Lombaert I, Schmidt TM, Eisbruch A, Murdoch-Kinch CA, Rozek L, Wolf GT, Li G, Chen GY, D'Silva NJ. Salivary microbiome changes distinguish response to chemoradiotherapy in patients with oral cancer. MICROBIOME 2023; 11:268. [PMID: 38037123 PMCID: PMC10687843 DOI: 10.1186/s40168-023-01677-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/26/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Oral squamous cell carcinoma (SCC) is associated with oral microbial dysbiosis. In this unique study, we compared pre- to post-treatment salivary microbiome in patients with SCC by 16S rRNA gene sequencing and examined how microbiome changes correlated with the expression of an anti-microbial protein. RESULTS Treatment of SCC was associated with a reduction in overall bacterial richness and diversity. There were significant changes in the microbial community structure, including a decrease in the abundance of Porphyromonaceae and Prevotellaceae and an increase in Lactobacillaceae. There were also significant changes in the microbial community structure before and after treatment with chemoradiotherapy, but not with surgery alone. In patients treated with chemoradiotherapy alone, several bacterial populations were differentially abundant between responders and non-responders before and after therapy. Microbiome changes were associated with a change in the expression of DMBT1, an anti-microbial protein in human saliva. Additionally, we found that salivary DMBT1, which increases after treatment, could serve as a post-treatment salivary biomarker that links to microbial changes. Specifically, post-treatment increases in human salivary DMBT1 correlated with increased abundance of Gemella spp., Pasteurellaceae spp., Lactobacillus spp., and Oribacterium spp. This is the first longitudinal study to investigate treatment-associated changes (chemoradiotherapy and surgery) in the oral microbiome in patients with SCC along with changes in expression of an anti-microbial protein in saliva. CONCLUSIONS The composition of the oral microbiota may predict treatment responses; salivary DMBT1 may have a role in modulating the oral microbiome in patients with SCC. After completion of treatment, 6 months after diagnosis, patients had a less diverse and less rich oral microbiome. Leptotrichia was a highly prevalent bacteria genus associated with disease. Expression of DMBT1 was higher after treatment and associated with microbiome changes, the most prominent genus being Gemella Video Abstract.
Collapse
Affiliation(s)
- Marcell Costa de Medeiros
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Stephanie The
- Cancer Data Science Shared Resource, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Emily Bellile
- Cancer Data Science Shared Resource, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Nickole Russo
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Ligia Schmitd
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Erika Danella
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Priyanka Singh
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Rajat Banerjee
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA
| | - Christine Bassis
- Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI, 331248109, USA
| | - George R Murphy
- Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA
- Biointerfaces Institute, Ann Arbor, MI, USA
| | - Maureen A Sartor
- Computational Medicine and Bioinformatics, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Isabelle Lombaert
- Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA
- Biointerfaces Institute, Ann Arbor, MI, USA
| | - Thomas M Schmidt
- Microbiology and Immunology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Avi Eisbruch
- Radiation Oncology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Carol Anne Murdoch-Kinch
- Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, 1011 North Michigan St, Indianapolis, IN, USA
| | - Laura Rozek
- Environmental Health Sciences, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Gregory T Wolf
- Otolaryngology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Gen Li
- Biostatistics, University of Michigan School of Public Health, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA
| | - Grace Y Chen
- Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI, 331248109, USA.
| | - Nisha J D'Silva
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Ave, Room G018, Ann Arbor, MI, 48109-1078, USA.
- Pathology, University of Michigan Medical School, 1500 E. Medical Center Dr, Ann Arbor, MI, USA.
- Rogel Cancer Center, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Delcroix V, Mauduit O, Lee HS, Ivanova A, Umazume T, Knox SM, de Paiva CS, Dartt DA, Makarenkova HP. The First Transcriptomic Atlas of the Adult Lacrimal Gland Reveals Epithelial Complexity and Identifies Novel Progenitor Cells in Mice. Cells 2023; 12:1435. [PMID: 37408269 PMCID: PMC10216974 DOI: 10.3390/cells12101435] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/07/2023] Open
Abstract
The lacrimal gland (LG) secretes aqueous tears. Previous studies have provided insights into the cell lineage relationships during tissue morphogenesis. However, little is known about the cell types composing the adult LG and their progenitors. Using scRNAseq, we established the first comprehensive cell atlas of the adult mouse LG to investigate the cell hierarchy, its secretory repertoire, and the sex differences. Our analysis uncovered the complexity of the stromal landscape. Epithelium subclustering revealed myoepithelial cells, acinar subsets, and two novel acinar subpopulations: Tfrchi and Car6hi cells. The ductal compartment contained Wfdc2+ multilayered ducts and an Ltf+ cluster formed by luminal and intercalated duct cells. Kit+ progenitors were identified as: Krt14+ basal ductal cells, Aldh1a1+ cells of Ltf+ ducts, and Sox10+ cells of the Car6hi acinar and Ltf+ epithelial clusters. Lineage tracing experiments revealed that the Sox10+ adult populations contribute to the myoepithelial, acinar, and ductal lineages. Using scRNAseq data, we found that the postnatally developing LG epithelium harbored key features of putative adult progenitors. Finally, we showed that acinar cells produce most of the sex-biased lipocalins and secretoglobins detected in mouse tears. Our study provides a wealth of new data on LG maintenance and identifies the cellular origin of sex-biased tear components.
Collapse
Affiliation(s)
- Vanessa Delcroix
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Olivier Mauduit
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Hyun Soo Lee
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Anastasiia Ivanova
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Takeshi Umazume
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| | - Sarah M. Knox
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA;
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Cintia S. de Paiva
- The Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Darlene A. Dartt
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA;
| | - Helen P. Makarenkova
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (V.D.); (H.S.L.); (A.I.); (T.U.)
| |
Collapse
|
8
|
Valdes J, Gagné-Sansfaçon J, Reyes V, Armas A, Marrero G, Moyo-Muamba M, Ramanathan S, Perreault N, Ilangumaran S, Rivard N, Fortier LC, Menendez A. Defects in the expression of colonic host defense factors associate with barrier dysfunction induced by a high-fat/high-cholesterol diet. Anat Rec (Hoboken) 2022; 306:1165-1183. [PMID: 36196983 DOI: 10.1002/ar.25083] [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: 04/07/2022] [Revised: 08/25/2022] [Accepted: 09/11/2022] [Indexed: 11/07/2022]
Abstract
The effect of Western diets in the gastrointestinal system is largely mediated by their ability to promote alterations in the immunity and physiology of the intestinal epithelium, and to affect the composition of the commensal microbiota. To investigate the response of the colonic epithelium to high-fat/high-cholesterol diets (HFHCDs), we evaluated the synthesis of host defense factors involved in the maintenance of the colonic homeostasis. C57BL/6 mice were fed an HFHCD for 3 weeks and their colons were evaluated for histopathology, gene expression, and microbiota composition. In addition, intestinal permeability and susceptibility to Citrobacter rodentium were also studied. HFHCD caused colonic hyperplasia, loss of goblet cells, thinning of the mucus layer, moderate changes in the composition of the intestinal microbiota, and an increase in intestinal permeability. Gene expression analyses revealed significant drops in the transcript levels of Muc1, Muc2, Agr2, Atoh1, Spdef, Ang4, Camp, Tff3, Dmbt1, Fcgbp, Saa3, and Retnlb. The goblet cell granules of HFHCD-fed mice were devoid of Relmβ and Tff3, indicating defective production of those two factors critical for intestinal epithelial defense and homeostasis. In correspondence with these defects, colonic bacteria were in close contact with, and invading the epithelium. Fecal shedding of C. rodentium showed an increased bacterial burden in HFHCD-fed animals accompanied by increased epithelial damage. Collectively, our results show that HFHCD perturbs the synthesis of colonic host defense factors, which associate with alterations in the commensal microbiota, the integrity of the intestinal barrier, and the host's susceptibility to enteric infections.
Collapse
Affiliation(s)
- Jennifer Valdes
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jessica Gagné-Sansfaçon
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Vilcy Reyes
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Anny Armas
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gisela Marrero
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mitterrand Moyo-Muamba
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nathalie Perreault
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nathalie Rivard
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Louis-Charles Fortier
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
9
|
Yarmola E, Ishkov IP, di Cologna NM, Menashe M, Whitener RL, Long JR, Abranches J, Hagen SJ, Brady LJ. Amyloid Aggregates Are Localized to the Nonadherent Detached Fraction of Aging Streptococcus mutans Biofilms. Microbiol Spectr 2022; 10:e0166122. [PMID: 35950854 PMCID: PMC9431626 DOI: 10.1128/spectrum.01661-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/21/2022] [Indexed: 11/23/2022] Open
Abstract
The number of bacterial species recognized to utilize purposeful amyloid aggregation within biofilms continues to grow. The oral pathogen Streptococcus mutans produces several amyloidogenic proteins, including adhesins P1 (also known as AgI/II, PAc) and WapA, whose truncation products, namely, AgII and AgA, respectively, represent the amyloidogenic moieties. Amyloids demonstrate common biophysical properties, including recognition by Thioflavin T (ThT) and Congo red (CR) dyes that bind to the cross β-sheet quaternary structure of amyloid aggregates. Previously, we observed amyloid formation to occur only after 60 h or more of S. mutans biofilm growth. Here, we extend those findings to investigate where amyloid is detected within 1- and 5-day-old biofilms, including within tightly adherent compared with those in nonadherent fractions. CR birefringence and ThT uptake demonstrated amyloid within nonadherent material removed from 5-day-old cultures but not within 1-day-old or adherent samples. These experiments were done in conjunction with confocal microscopy and immunofluorescence staining with AgII- and AgA-reactive antibodies, including monoclonal reagents shown to discriminate between monomeric protein and amyloid aggregates. These results also localized amyloid primarily to the nonadherent fraction of biofilms. Lastly, we show that the C-terminal region of P1 loses adhesive function following amyloidogenesis and is no longer able to competitively inhibit binding of S. mutans to its physiologic substrate, salivary agglutinin. Taken together, our results provide new evidence that amyloid aggregation negatively impacts the functional activity of a widely studied S. mutans adhesin and are consistent with a model in which amyloidogenesis of adhesive proteins facilitates the detachment of aging biofilms. IMPORTANCE Streptococcus mutans is a keystone pathogen and causative agent of human dental caries, commonly known as tooth decay, the most prevalent infectious disease in the world. Like many pathogens, S. mutans causes disease in biofilms, which for dental decay begins with bacterial attachment to the salivary pellicle coating the tooth surface. Some strains of S. mutans are also associated with bacterial endocarditis. Amyloid aggregation was initially thought to represent only a consequence of protein mal-folding, but now, many microorganisms are known to produce functional amyloids with biofilm environments. In this study, we learned that amyloid formation diminishes the activity of a known S. mutans adhesin and that amyloid is found within the nonadherent fraction of older biofilms. This finding suggests that the transition from adhesin monomer to amyloid facilitates biofilm detachment. Knowing where and when S. mutans produces amyloid will help in developing therapeutic strategies to control tooth decay and other biofilm-related diseases.
Collapse
Affiliation(s)
- Elena Yarmola
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Ivan P. Ishkov
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | | | - Megan Menashe
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Robert L. Whitener
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Joanna R. Long
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | | | - Stephen J. Hagen
- Department of Physics, University of Florida, Gainesville, Florida, USA
| | - L. Jeannine Brady
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Johnstone KF, Herzberg MC. Antimicrobial peptides: Defending the mucosal epithelial barrier. FRONTIERS IN ORAL HEALTH 2022; 3:958480. [PMID: 35979535 PMCID: PMC9376388 DOI: 10.3389/froh.2022.958480] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
The recent epidemic caused by aerosolized SARS-CoV-2 virus illustrates the importance and vulnerability of the mucosal epithelial barrier against infection. Antimicrobial proteins and peptides (AMPs) are key to the epithelial barrier, providing immunity against microbes. In primitive life forms, AMPs protect the integument and the gut against pathogenic microbes. AMPs have also evolved in humans and other mammals to enhance newer, complex innate and adaptive immunity to favor the persistence of commensals over pathogenic microbes. The canonical AMPs are helictical peptides that form lethal pores in microbial membranes. In higher life forms, this type of AMP is exemplified by the defensin family of AMPs. In epithelial tissues, defensins, and calprotectin (complex of S100A8 and S100A9) have evolved to work cooperatively. The mechanisms of action differ. Unlike defensins, calprotectin sequesters essential trace metals from microbes, which inhibits growth. This review focuses on defensins and calprotectin as AMPs that appear to work cooperatively to fortify the epithelial barrier against infection. The antimicrobial spectrum is broad with overlap between the two AMPs. In mice, experimental models highlight the contribution of both AMPs to candidiasis as a fungal infection and periodontitis resulting from bacterial dysbiosis. These AMPs appear to contribute to innate immunity in humans, protecting the commensal microflora and restricting the emergence of pathobionts and pathogens. A striking example in human innate immunity is that elevated serum calprotectin protects against neonatal sepsis. Calprotectin is also remarkable because of functional differences when localized in epithelial and neutrophil cytoplasm or released into the extracellular environment. In the cytoplasm, calprotectin appears to protect against invasive pathogens. Extracellularly, calprotectin can engage pathogen-recognition receptors to activate innate immune and proinflammatory mechanisms. In inflamed epithelial and other tissue spaces, calprotectin, DNA, and histones are released from degranulated neutrophils to form insoluble antimicrobial barriers termed neutrophil extracellular traps. Hence, calprotectin and other AMPs use several strategies to provide microbial control and stimulate innate immunity.
Collapse
Affiliation(s)
| | - Mark C. Herzberg
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
11
|
Gao Z, Wu Z, Han Y, Zhang X, Hao P, Xu M, Huang S, Li S, Xia J, Jiang J, Yang S. Aberrant Fucosylation of Saliva Glycoprotein Defining Lung Adenocarcinomas Malignancy. ACS OMEGA 2022; 7:17894-17906. [PMID: 35664632 PMCID: PMC9161393 DOI: 10.1021/acsomega.2c01193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Aberrant glycosylation is a hallmark of cancer found during tumorigenesis and tumor progression. Lung cancer (LC) induced by oncogene mutations has been detected in the patient's saliva, and saliva glycosylation has been altered. Saliva contains highly glycosylated glycoproteins, the characteristics of which may be related to various diseases. Therefore, elucidating cancer-specific glycosylation in the saliva of healthy, non-cancer, and cancer patients can reveal whether tumor glycosylation has unique characteristics for early diagnosis. In this work, we used a solid-phase chemoenzymatic method to study the glycosylation of saliva glycoproteins in clinical specimens. The results showed that the α1,6-core fucosylation of glycoproteins was increased in cancer patients, whereas α1,2 or α1,3 fucosylation was significantly increased. We further analyzed the expression of fucosyltransferases responsible for α1,2, α1,3, and α1,6 fucosylation. The fucosylation of the saliva of cancer patients is drastically different from that of non-cancer or health controls. These results indicate that the glycoform of saliva fucosylation distinguishes LC from other diseases, and this feature has the potential to diagnose lung adenocarcinoma.
Collapse
Affiliation(s)
- Ziyuan Gao
- Center
for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
- Department
of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Pinghai Road No. 899, Suzhou 215000, China
| | - Zhen Wu
- State
Key Laboratory of Genetic Engineering, Department of Biochemistry,
School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ying Han
- School
of Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Xumin Zhang
- State
Key Laboratory of Genetic Engineering, Department of Biochemistry,
School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Piliang Hao
- School
of Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Mingming Xu
- Center
for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shan Huang
- Center
for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shuwei Li
- Nanjing
Apollomics Biotech, Inc., Nanjing, Jiangsu 210033, China
| | - Jun Xia
- Department
of Clinical Laboratory Center, Zhejiang Provincial People’s
Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou, Zhejiang 310014, China
| | - Junhong Jiang
- Department
of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Pinghai Road No. 899, Suzhou 215000, China
- Department
of Pulmonary and Critical Care Medicine, Dushu Lake Hospital, Affiliated to Soochow University, Chongwen Road No. 9, Suzhou 215000, China
| | - Shuang Yang
- Center
for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
12
|
Friedrich B, Lyer S, Janko C, Unterweger H, Brox R, Cunningham S, Dutz S, Taccardi N, Bikker FJ, Hurle K, Sebald H, Lenz M, Spiecker E, Fester L, Hackstein H, Strauß R, Boccaccini AR, Bogdan C, Alexiou C, Tietze R. Scavenging of bacteria or bacterial products by magnetic particles functionalized with a broad-spectrum pathogen recognition receptor motif offers diagnostic and therapeutic applications. Acta Biomater 2022; 141:418-428. [PMID: 34999260 DOI: 10.1016/j.actbio.2022.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/21/2021] [Accepted: 01/03/2022] [Indexed: 11/19/2022]
Abstract
Sepsis is a dysregulated host response of severe bloodstream infections, and given its frequency of occurrence and high mortality rate, therapeutic improvements are imperative. A reliable biomimetic strategy for the targeting and separation of bacterial pathogens in bloodstream infections involves the use of the broad-spectrum binding motif of human GP-340, a pattern-recognition receptor of the scavenger receptor cysteine rich (SRCR) superfamily that is expressed on epithelial surfaces but not found in blood. Here we show that these peptides, when conjugated to superparamagnetic iron oxide nanoparticles (SPIONs), can separate various bacterial endotoxins and intact microbes (E. coli, S. aureus, P. aeruginosa and S. marcescens) with high efficiency, especially at low and thus clinically relevant concentrations. This is accompanied by a subsequent strong depletion in cytokine release (TNF, IL-6, IL-1β, Il-10 and IFN-γ), which could have a direct therapeutic impact since escalating immune responses complicates severe bloodstream infections and sepsis courses. SPIONs are coated with aminoalkylsilane and capture peptides are orthogonally ligated to this surface. The particles behave fully cyto- and hemocompatible and do not interfere with host structures. Thus, this approach additionally aims to dramatically reduce diagnostic times for patients with suspected bloodstream infections and accelerate targeted antibiotic therapy. STATEMENT OF SIGNIFICANCE: Sepsis is often associated with excessive release of cytokines. This aspect and slow diagnostic procedures are the major therapeutic obstacles. The use of magnetic particles conjugated with small peptides derived from the binding motif of a broad-spectrum mucosal pathogen recognition protein GP-340 provides a highly efficient scavenging platform. These peptides are not found in blood and therefore are not subject to inhibitory mechanisms like in other concepts (mannose binding lectine, aptamers, antibodies). In this work, data are shown on the broad bacterial binding spectrum, highly efficient toxin depletion, which directly reduces the release of cytokines. Host cells are not affected and antibiotics not adsorbed. The particle bound microbes can be recultured without restriction and thus be used directly for diagnostics.
Collapse
Affiliation(s)
- Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Germany
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Germany
| | - Regine Brox
- Department of Transfusion Medicine and Hemostaseology, Universitätsklinikum Erlangen, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, Universitätsklinikum Erlangen, Germany
| | - Silvio Dutz
- Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau, Germany
| | - Nicola Taccardi
- Institute of Chemical Reaction Engineering, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Floris J Bikker
- Department of Oral Biochemistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), the Netherlands
| | - Katrin Hurle
- GeoZentrum Nordbayern, Mineralogy, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Heidi Sebald
- Immunologie und Hygiene, Mikrobiologisches Institut - Klinische Mikrobiologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Malte Lenz
- Institute of Micro- and Nanostructure Research and Center for Nanoanalysis and Electron Microscopy (CENEM), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Interdisciplinary Center for Nanostructure Films (IZNF), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Erdmann Spiecker
- Institute of Micro- and Nanostructure Research and Center for Nanoanalysis and Electron Microscopy (CENEM), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Interdisciplinary Center for Nanostructure Films (IZNF), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Lars Fester
- Institute of Anatomy and Cell Biology Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, Universitätsklinikum Erlangen, Germany
| | - Richard Strauß
- Department of Medicine 1, Universitätsklinikum Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Christian Bogdan
- Immunologie und Hygiene, Mikrobiologisches Institut - Klinische Mikrobiologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Germany
| | - Rainer Tietze
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen, Germany.
| |
Collapse
|
13
|
Kiefer A, Plattner E, Ruppel R, Weiss C, Zhou-Suckow Z, Mall M, Renner M, Müller H. DMBT1 is upregulated in cystic fibrosis, affects ciliary motility, and is reduced by acetylcysteine. Mol Cell Pediatr 2022; 9:4. [PMID: 35249163 PMCID: PMC8898207 DOI: 10.1186/s40348-022-00136-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 01/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is the most common genetic disorder in the Caucasian population. Despite remarkable improvements in morbidity and mortality during the last decades, the disease still limits survival and reduces quality of life of affected patients. Moreover, CF still represents substantial economic burden for healthcare systems. Inflammation and infection already start in early life and play important roles in pulmonary impairment. The aim of this study is to analyze the potential role of DMBT1, a protein with functions in inflammation, angiogenesis, and epithelial differentiation, in CF. RESULTS Immunohistochemically DMBT1 protein expression was upregulated in lung tissues of CF patients compared to healthy controls. Additionally, pulmonary expression of Dmbt1 was approximately 6-fold increased in an established transgenic mouse model of CF-like lung disease (ENaC tg) compared to wild-type mice as detected by qRT-PCR. Since acetylcysteine (ACC) has been shown to reduce inflammatory markers in the airways, its potential influence on DMBT1 expression was analyzed. A549 cells stably transfected with an expression plasmid encoding the largest (8kb) DMBT1 variant (DMBT1+ cells) or an empty vector control (DMBT1- cells) and incubated with ACC both showed significantly reduced DMBT1 concentrations in the culture medium (p = 0.0001). To further elucidate the function of DMBT1 in pulmonary airways, respiratory epithelial cells were examined by phase contrast microscopy. Addition of human recombinant DMBT1 resulted in altered cilia motility and irregular beat waves (p < 0.0001) suggesting a potential effect of DMBT1 on airway clearance. CONCLUSIONS DMBT1 is part of inflammatory processes in CF and may be used as a potential biomarker for CF lung disease and a potential tool to monitor CF progression. Furthermore, DMBT1 has a negative effect on ciliary motility thereby possibly compromising airway clearance. Application of ACC, leading to reduced DMBT1 concentrations, could be a potential therapeutic option for CF patients.
Collapse
Affiliation(s)
- Alexander Kiefer
- Department of Pediatrics, University Hospital Erlangen, University of Erlangen-Nürnberg, Loschgestr. 15, 91054, Erlangen, Germany.,Department of Pediatric Pneumology and Allergology, St. Hedwig's Hospital of the Order of St. John, University Children's Hospital Regensburg (KUNO), Steinmetzstr. 1-3, 93049, Regensburg, Germany
| | - Erika Plattner
- Department of Pediatrics, University Hospital Erlangen, University of Erlangen-Nürnberg, Loschgestr. 15, 91054, Erlangen, Germany
| | - Renate Ruppel
- Department of Pediatrics, University Hospital Erlangen, University of Erlangen-Nürnberg, Loschgestr. 15, 91054, Erlangen, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Zhe Zhou-Suckow
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Im Neuenheimer Feld, 69120, Heidelberg, Germany
| | - Marcus Mall
- Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Marcus Renner
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Hanna Müller
- Department of Pediatrics, Neonatology and Pediatric Intensive Care, University of Marburg, Baldingerstraße, 35043, Marburg, Germany. .,Department of Pediatrics, Division of Neonatology and Pediatric Intensive Care, University Hospital Erlangen, University of Erlangen-Nürnberg, Loschgestr. 15, 91054, Erlangen, Germany.
| |
Collapse
|
14
|
Manzer HS, Villarreal RI, Doran KS. Targeting the BspC-vimentin interaction to develop anti-virulence therapies during Group B streptococcal meningitis. PLoS Pathog 2022; 18:e1010397. [PMID: 35316308 PMCID: PMC8939794 DOI: 10.1371/journal.ppat.1010397] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 12/21/2022] Open
Abstract
Bacterial infections are a major cause of morbidity and mortality worldwide and the rise of antibiotic resistance necessitates development of alternative treatments. Pathogen adhesins that bind to host cells initiate disease pathogenesis and represent potential therapeutic targets. We have shown previously that the BspC adhesin in Group B Streptococcus (GBS), the leading cause of bacterial neonatal meningitis, interacts with host vimentin to promote attachment to brain endothelium and disease development. Here we determined that the BspC variable (V-) domain contains the vimentin binding site and promotes GBS adherence to brain endothelium. Site directed mutagenesis identified a binding pocket necessary for GBS host cell interaction and development of meningitis. Using a virtual structure-based drug screen we identified compounds that targeted the V-domain binding pocket, which blocked GBS adherence and entry into the brain in vivo. These data indicate the utility of targeting the pathogen-host interface to develop anti-virulence therapeutics.
Collapse
Affiliation(s)
- Haider S. Manzer
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Ricardo I. Villarreal
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Kelly S. Doran
- University of Colorado Anschutz Medical Campus, Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| |
Collapse
|
15
|
Diez S, Besendörfer M, Weyerer V, Hartmann A, Moosmann J, Weiss C, Renner M, Müller H. DMBT1 expression and neutrophil-to-lymphocyte ratio during necrotizing enterocolitis are influenced by impaired perfusion due to cardiac anomalies. Mol Cell Pediatr 2022; 9:1. [PMID: 34989914 PMCID: PMC8739415 DOI: 10.1186/s40348-021-00133-9] [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: 06/08/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Deleted in malignant brain tumors 1 (DMBT1) is involved in innate immunity and epithelial differentiation. It has been proven to play a role in various states of inflammation or hypoxia of fetal gastrointestinal and pulmonary diseases. Discrimination of pathogenesis in necrotizing enterocolitis (NEC) based on cardiac status improves the understanding of NEC in different patient subgroups. We aimed at examining DMBT1 expressions regarding their association with cardiac status leading to impaired intestinal perfusion, intraoperative bacteria proof, and a fulminant course of NEC. METHODS Twenty-eight patients with NEC were treated surgically between 2010 and 2019 at our institution. DMBT1 expression was examined in intestinal sections using immunohistochemistry to detect DMBT1 protein. Associations of clinical parameters and DMBT1 expression were analyzed. RESULTS We examined DMBT1 levels in 10 patients without cardiac defects and 18 patients with persisting ductus arteriosus (PDA) and congenital heart defects (CHD). Compared to patients without cardiac malformations, DMBT1 levels tended to score higher in patients with PDA/CHD (p = 0.2113) and were negatively correlated with C-reactive protein in these infants (p = 0.0172; r = - 0.5533). The number of DMBT1-expressing macrophages was elevated in the PDA/CHD-subgroup (p = 0.0399). Ratios of neutrophils and monocytes to lymphocytes were significantly higher in infants with PDA/CHD (p = 0.0319 and 0.0493). DMBT1 expression was significantly associated with positive bacterial culture of intraoperative swabs (p = 0.0252) and DMBT1 expression of the serosa was associated with a fulminant course of NEC (p = 0.0239). CONCLUSIONS This study demonstrates that DMBT1 expression may be influenced by cardiac anomalies with an impaired intestinal perfusion in the neonatal intestine. NEC in PDA/CHD infants is associated with more DMBT1-positive macrophages and a significantly elevated neutrophil-to-lymphocyte ratio.
Collapse
Affiliation(s)
- Sonja Diez
- Pediatric Surgery, Department for General Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Loschgestraße 15, 91054, Erlangen, Germany.
| | - Manuel Besendörfer
- Pediatric Surgery, Department for General Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Loschgestraße 15, 91054, Erlangen, Germany
| | - Veronika Weyerer
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 8-10, 91054, Erlangen, Germany
| | - Julia Moosmann
- Department of Pediatric Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Loschgestraße 15, 91054, Erlangen, Germany
| | - Christel Weiss
- Department of Medical Statistics & Biomathematics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Haus 3, Ebene 4, 68167, Mannheim, Germany
| | - Marcus Renner
- Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Hanna Müller
- Division of Neonatology and Pediatric Intensive Care, Department of Pediatrics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Loschgestraße 15, 91054, Erlangen, Germany.,Neonatology and Pediatric Intensive Care, Department of Pediatrics, University of Marburg, Baldingerstraße, 35033, Marburg, Germany
| |
Collapse
|
16
|
Mukherjee P, Chattopadhyay A, Grijalva V, Dorreh N, Lagishetty V, Jacobs JP, Clifford BL, Vallim T, Mack JJ, Navab M, Reddy ST, Fogelman AM. Oxidized phospholipids cause changes in jejunum mucus that induce dysbiosis and systemic inflammation. J Lipid Res 2022; 63:100153. [PMID: 34808192 PMCID: PMC8953663 DOI: 10.1016/j.jlr.2021.100153] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 10/26/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
We previously reported that adding a concentrate of transgenic tomatoes expressing the apoA-I mimetic peptide 6F (Tg6F) to a Western diet (WD) ameliorated systemic inflammation. To determine the mechanism(s) responsible for these observations, Ldlr-/- mice were fed chow, a WD, or WD plus Tg6F. We found that a WD altered the taxonomic composition of bacteria in jejunum mucus. For example, Akkermansia muciniphila virtually disappeared, while overall bacteria numbers and lipopolysaccharide (LPS) levels increased. In addition, gut permeability increased, as did the content of reactive oxygen species and oxidized phospholipids in jejunum mucus in WD-fed mice. Moreover, gene expression in the jejunum decreased for multiple peptides and proteins that are secreted into the mucous layer of the jejunum that act to limit bacteria numbers and their interaction with enterocytes including regenerating islet-derived proteins, defensins, mucin 2, surfactant A, and apoA-I. Following WD, gene expression also decreased for Il36γ, Il23, and Il22, cytokines critical for antimicrobial activity. WD decreased expression of both Atoh1 and Gfi1, genes required for the formation of goblet and Paneth cells, and immunohistochemistry revealed decreased numbers of goblet and Paneth cells. Adding Tg6F ameliorated these WD-mediated changes. Adding oxidized phospholipids ex vivo to the jejunum from mice fed a chow diet reproduced the changes in gene expression in vivo that occurred when the mice were fed WD and were prevented with addition of 6F peptide. We conclude that Tg6F ameliorates the WD-mediated increase in oxidized phospholipids that cause changes in jejunum mucus, which induce dysbiosis and systemic inflammation.
Collapse
Affiliation(s)
- Pallavi Mukherjee
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | | | - Victor Grijalva
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Nasrin Dorreh
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA; UCLA Microbiome Center, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA; UCLA Microbiome Center, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; The Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System Los Angeles, Los Angeles, CA, USA
| | | | - Thomas Vallim
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA; Department of Biological Chemistry, Los Angeles, CA, USA
| | - Julia J Mack
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Mohamad Navab
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, Los Angeles, CA, USA
| |
Collapse
|
17
|
Gao Z, Xu M, Yue S, Shan H, Xia J, Jiang J, Yang S. Abnormal sialylation and fucosylation of saliva glycoproteins: Characteristics of lung cancer-specific biomarkers. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 3:100079. [PMID: 35005612 PMCID: PMC8718573 DOI: 10.1016/j.crphar.2021.100079] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 10/31/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022] Open
Abstract
Dysregulated surface glycoproteins play an important role in tumor cell proliferation and progression. Abnormal glycosylation of these glycoproteins may activate tumor signal transduction and lead to tumor development. The tumor microenvironment alters its molecular composition, some of which regulate protein glycosylation biosynthesis. The glycosylation of saliva proteins in lung cancer patients is different from healthy controls, in which the glycans of cancer patients are highly sialylated and hyperfucosylated. Most studies have shown that O-glycans from cancer are truncated O-glycans, while N-glycans contain fucoses and sialic acids. Because glycosylation analysis is challenging, there are few reports on how glycosylation of saliva proteins is related to the occurrence or progression of lung cancer. In this review, we discussed glycoenzymes involved in protein glycosylation, their changes in tumor microenvironment, potential tumor biomarkers present in body fluids, and abnormal glycosylation of saliva or lung glycoproteins. We further explored the effect of glycosylation changes on tumor signal transduction, and emphasized the role of receptor tyrosine kinases in tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ziyuan Gao
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital to Soochow University, Suzhou, Jiangsu, 215125, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University; Suzhou Jiangsu, 215006, China
| | - Mingming Xu
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Shuang Yue
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Huang Shan
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Jun Xia
- Department of Clinical Laboratory Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Junhong Jiang
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital to Soochow University, Suzhou, Jiangsu, 215125, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University; Suzhou Jiangsu, 215006, China
| | - Shuang Yang
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| |
Collapse
|
18
|
Katsumata T, Nguyen-Tra Le M, Kawada-Matsuo M, Taniguchi Y, Ouhara K, Oogai Y, Nakata M, Mizuno N, Nishitani Y, Komatsuzawa H. KATSUMATA et al.Comprehensive characterization of sortase A-dependent surface proteins in Streptococcus mutansComprehensive characterization of sortase A-dependent surface proteins in Streptococcus mutans. Microbiol Immunol 2021; 66:145-156. [PMID: 34888908 DOI: 10.1111/1348-0421.12958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
Streptococcus mutans, a cariogenic pathogen, adheres to the tooth surface and forms a biofilm. Bacterial cell surface proteins are associated with adherence to substrates. Sortase A (SrtA) mediates the localization of proteins with an LPXTG motif-containing proteins to the cell surface by covalent binding to peptidoglycan. In S. mutans UA159, 6 SrtA-dependent proteins, SpaP, WapA, WapE, DexA, FruA, and GbpC, were identified. Although some of these proteins were characterized, a comprehensive analysis of the 6 proteins has not been reported. In this study, we constructed mutants deficient in each of these proteins and the SrtA-deficient mutant. The SrtA-deficient mutant showed drastically decreased binding to salivary components, biofilm formation, bacterial coaggregation activity, hydrophobicity, and cellular matrix binding (collagen type I, fibronectin, and laminin). The SpaP-deficient mutant showed significantly reduced binding to salivary components and partially increased coaggregation with Porphyromonas gingivalis, and decreased hydrophobicity, and collagen binding. The WapA-deficient mutant showed slightly decreased coaggregation with Fusobacterium nucleatum. Although the SrtA-deficient mutant showed drastically altered phenotypes, all SrtA-dependent protein-deficient mutants, except the SpaP-deficient mutant, did not show considerable alterations in binding to salivary components. These results indicate that the 6 proteins may coordinately contribute to these activities. In addition, using genomic data of 125 S. mutans strains, we compared the amino acid sequences of each surface protein and found many variations among strains, which may affect the phenotype of cell surface proteins in S. mutans. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tamaki Katsumata
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mi Nguyen-Tra Le
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuri Taniguchi
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masanobu Nakata
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
19
|
Salivary Trefoil Factor Family (TFF) Peptides and Their Roles in Oral and Esophageal Protection: Therapeutic Potential. Int J Mol Sci 2021; 22:ijms222212221. [PMID: 34830103 PMCID: PMC8624312 DOI: 10.3390/ijms222212221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Human saliva is a complex body fluid with more than 3000 different identified proteins. Besides rheological and lubricating properties, saliva supports wound healing and acts as an antimicrobial barrier. TFF peptides are secreted from the mucous acini of the major and minor salivary glands and are typical constituents of normal saliva; TFF3 being the predominant peptide compared with TFF1 and TFF2. Only TFF3 is easily detectable by Western blotting. It occurs in two forms, a disulfide-linked homodimer (Mr: 13k) and a high-molecular-mass heterodimer with IgG Fc binding protein (FCGBP). TFF peptides are secretory lectins known for their protective effects in mucous epithelia; the TFF3 dimer probably has wound-healing properties due to its weak motogenic effect. There are multiple indications that FCGBP and TFF3-FCGBP play a key role in the innate immune defense of mucous epithelia. In addition, homodimeric TFF3 interacts in vitro with the salivary agglutinin DMBT1gp340. Here, the protective roles of TFF peptides, FCGBP, and DMBT1gp340 in saliva are discussed. TFF peptides are also used to reduce radiotherapy- or chemotherapy-induced oral mucositis. Thus, TFF peptides, FCGBP, and DMBT1gp340 are promising candidates for better formulations of artificial saliva, particularly improving wound healing and antimicrobial effects even in the esophagus.
Collapse
|
20
|
Genetic Variants of DMBT1 and SFTPD and Disease Severity in Paediatric Inflammatory Bowel Disease—A Polish Population-Based Study. CHILDREN 2021; 8:children8110946. [PMID: 34828659 PMCID: PMC8618964 DOI: 10.3390/children8110946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 11/16/2022]
Abstract
Deleted in malignant brain tumours 1 protein (DMBT1) and surfactant protein D (SFTPD) are antimicrobial peptides previously linked to inflammatory bowel disease (IBD) susceptibility. This study attempts to link the most potential IBD-associated polymorphisms in DMBT1 and SFTPD with the disease severity in children. A total of 406 IBD patients (Crohn’s disease (CD) n = 214 and ulcerative colitis (UC) n = 192) were genotyped using hydrolysis probe assay. Clinical expression was described by disease activity scales, albumin and C-reactive protein levels, localisation and behaviour (Paris classification), systemic steroid, immunosuppressive, biological, and surgical treatment, number of exacerbation-caused hospitalisations, relapses and nutritional status. IBD patients with the risk genotype (AA) in DMBT1 rs2981804 had more frequent biological treatment (AA: vs. AG/GG; p = 0.012), concomitant diseases (AA vs. AG vs. GG; p = 0.015) and cutaneous manifestations (AA vs. AG/GG, p = 0.008). In UC, rs2981804 genotypes might be linked with albumin concentrations at diagnosis (AA vs. AG vs. GG; p = 0.009). In CD, DMBT1 rs2981745 was significantly associated with the number of severe relapses per year of disease (p = 0.020) and time-to-immunosuppression (p = 0.045). SFTPD was seemingly found to be associated with age at first immunosuppression in IBD (CC vs. CT vs. TT; p = 0.048). In conclusion, selected polymorphisms of DMBT1 and SFTPD might be associated with some disease severity measures in children with IBD. However, the magnitude of associations and their clinical relevance might be minor.
Collapse
|
21
|
Glycoprotein 340's scavenger receptor cysteine-rich domain promotes adhesion of Staphylococcus aureus and Pseudomonas aeruginosa to contact lens polymers. Infect Immun 2021; 90:e0033921. [PMID: 34662210 DOI: 10.1128/iai.00339-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Contact lenses are biomaterials worn on the eye to correct refractive errors. Bacterial adhesion and colonization of these lenses results in adverse events such as microbial keratitis. The adsorption of tear proteins to contact lens materials enhances bacterial adhesion. Glycoprotein 340 (Gp340), a tear component, is known to promote microbial colonization in the oral cavity, however, it has not been investigated in any contact lens-related adverse event. Therefore, this study examined the adsorption of Gp340 and its recombinantly expressed scavenger receptor cysteine rich (iSRCR1Gp340) domain on two common contact lens materials, etafilcon A and lotrafilcon B, and the concomitant effects on the adherence of clinical isolates of microbial keratitis causative agents, Pseudomonas aeruginosa (PA6206, PA6294), and Staphylococcus aureus (SA38, USA300). Across all strains and materials, iSRCR1Gp340 enhanced adherence of bacteria in a dose-dependent manner. However, iSRCR1Gp340 did not modulate lysozyme's and lactoferrin's effects on bacterial adhesion to the contact lens. The Gp340 binding surface protein SraP significantly enhanced USA300 binding to iSRCR1Gp340-coated lenses. In addition, iSRCR1Gp340-coated surfaces had significantly diminished biofilms with the SraP mutant (ΔSraP), and with the Sortase A mutant (ΔSrtA), there was a further reduction in biofilms, indicating the likely involvement of additional surface proteins. Finally, the binding affinities between iSRCR1Gp340 and SraP were determined using surface plasmon resonance (SPR), where the complete SraP binding region displayed nanomolar affinity, whereas its smaller fragments adhered with micromolar affinities. This study concludes that Gp340 and its SRCR domains play an important role in bacterial adhesion to the contact lens.
Collapse
|
22
|
Skerrett-Byrne DA, Nixon B, Bromfield EG, Breen J, Trigg NA, Stanger SJ, Bernstein IR, Anderson AL, Lord T, Aitken RJ, Roman SD, Robertson SA, Schjenken JE. Transcriptomic analysis of the seminal vesicle response to the reproductive toxicant acrylamide. BMC Genomics 2021; 22:728. [PMID: 34625024 PMCID: PMC8499523 DOI: 10.1186/s12864-021-07951-1] [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: 02/26/2021] [Accepted: 08/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The seminal vesicles synthesise bioactive factors that support gamete function, modulate the female reproductive tract to promote implantation, and influence developmental programming of offspring phenotype. Despite the significance of the seminal vesicles in reproduction, their biology remains poorly defined. Here, to advance understanding of seminal vesicle biology, we analyse the mouse seminal vesicle transcriptome under normal physiological conditions and in response to acute exposure to the reproductive toxicant acrylamide. Mice were administered acrylamide (25 mg/kg bw/day) or vehicle control daily for five consecutive days prior to collecting seminal vesicle tissue 72 h following the final injection. RESULTS A total of 15,304 genes were identified in the seminal vesicles with those encoding secreted proteins amongst the most abundant. In addition to reproductive hormone pathways, functional annotation of the seminal vesicle transcriptome identified cell proliferation, protein synthesis, and cellular death and survival pathways as prominent biological processes. Administration of acrylamide elicited 70 differentially regulated (fold-change ≥1.5 or ≤ 0.67) genes, several of which were orthogonally validated using quantitative PCR. Pathways that initiate gene and protein synthesis to promote cellular survival were prominent amongst the dysregulated pathways. Inflammation was also a key transcriptomic response to acrylamide, with the cytokine, Colony stimulating factor 2 (Csf2) identified as a top-ranked upstream driver and inflammatory mediator associated with recovery of homeostasis. Early growth response (Egr1), C-C motif chemokine ligand 8 (Ccl8), and Collagen, type V, alpha 1 (Col5a1) were also identified amongst the dysregulated genes. Additionally, acrylamide treatment led to subtle changes in the expression of genes that encode proteins secreted by the seminal vesicle, including the complement regulator, Complement factor b (Cfb). CONCLUSIONS These data add to emerging evidence demonstrating that the seminal vesicles, like other male reproductive tract tissues, are sensitive to environmental insults, and respond in a manner with potential to exert impact on fetal development and later offspring health.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - James Breen
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,South Australian Genomics Centre (SAGC), South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.,Computational & Systems Biology Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia. .,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
23
|
Structure-function characterization of Streptococcus intermedius surface antigen Pas. J Bacteriol 2021; 203:e0017521. [PMID: 34339301 DOI: 10.1128/jb.00175-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus intermedius, an oral commensal bacterium, is found at various sites including subgingival dental plaque, purulent infections, and in cystic fibrosis lungs. Oral streptococci utilize proteins on their surface to adhere to tissues and/or surfaces localizing the bacteria, which subsequently leads to the development of biofilms, colonization and infection. Among the 19 genomically annotated cell-wall attached surface proteins on S. intermedius, Pas is an adhesin that belongs to the Antigen I/II (AgI/II) family. Here we have structurally and functionally characterized Pas, particularly focusing on its microbial-host as well as microbial-microbial interactions. The crystal structures of VPas and C123Pas show high similarity with AgI/II of S. mutans. VPas hosts a conserved metal binding site, and likewise the C123Pas structure retains its conserved metal binding sites and isopeptide bonds within its three DEv-IgG domains. Pas interacts with nanomolar affinity to lung alveolar glycoprotein 340 (Gp340), its scavenger receptor cysteine rich domains (SRCRs) and with fibrinogen. Both Candida albicans and Pseudomonas aeruginosa, the opportunistic pathogens that cohabitate with S. intermedius in the lungs of CFTR patients were studied in dual-species biofilm studies. The Pas deficient mutant (Δpas) displayed significant reduction in dual biofilm formation with C. albicans. In similar studies with P. aeruginosa, Pas did not mediate the biofilm formation with either the acute isolate (PAO1), or the chronic isolate (FRD1). However, the Sortase A deficient mutant (ΔsrtA) displayed reduced biofilm formation with both C. albicans and P. aeruginosa FRD1. Taken together, our findings highlight the role of Pas in both microbial-host and interkingdom interactions and expose its potential role in disease outcomes. Importance Streptococcus intermedius, an oral commensal bacterium, has been clinically observed in subgingival dental plaque, purulent infections, and in cystic fibrosis lungs. In this study, we have (a) determined the crystal structure of the V- and C-regions of Pas; (b) shown that its surface protein Pas adheres to fibrinogen, which could potentially ferry the microbe through the blood stream from the oral cavity; (c) characterized Pas's high affinity adherence to lung alveolar protein Gp340 that could fixate the microbe on lung epithelial cells; and (d) most importantly shown that these surface proteins on the oral commensal S. intermedius enhances biofilms of known pathogens Candida albicans and Pseudomonas aeruginosa.
Collapse
|
24
|
Rae J, Hackney J, Huang K, Keir M, Herman A. Identification of an IL-22-Dependent Gene Signature as a Pharmacodynamic Biomarker. Int J Mol Sci 2021; 22:8205. [PMID: 34360971 PMCID: PMC8347589 DOI: 10.3390/ijms22158205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
Interleukin-22 (IL-22) plays a role in epithelial barrier function and repair, and may provide benefits in conditions like inflammatory bowel disease. However, limited human data are available to assess the clinical effect of IL-22 administration. This study used a human intestinal cell line to identify an IL-22-dependent gene signature that could serve as a pharmacodynamic biomarker for IL-22 therapy. The response to IL-22Fc (UTTR1147A, an Fc-stabilized version of IL-22) was assessed in HT-29 cells by microarray, and the selected responsive genes were confirmed by qPCR. HT-29 cells demonstrated dose-dependent increases in STAT3 phosphorylation and multiple gene expression changes in response to UTTR1147A. Genes were selected that were upregulated by UTTR1147A, but to a lesser extent by IL-6, which also signals via STAT3. IL-1R1 was highly upregulated by UTTR1147A, and differential gene expression patterns were observed in response to IL-22Fc in the presence of IL-1β. An IL-22-dependent gene signature was identified that could serve as a pharmacodynamic biomarker in intestinal biopsies to support the clinical development of an IL-22 therapeutic. The differential gene expression pattern in the presence of IL-1β suggests that an inflammatory cytokine milieu in the disease setting could influence the clinical responses to IL-22.
Collapse
Affiliation(s)
- Julie Rae
- OMNI Biomarker Development, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Jason Hackney
- Bioinformatics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA; (J.H.); (K.H.)
| | - Kevin Huang
- Bioinformatics, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA; (J.H.); (K.H.)
| | - Mary Keir
- OMNI Biomarker Discovery, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA;
| | - Ann Herman
- OMNI Biomarker Development, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA;
| |
Collapse
|
25
|
Nexoe AB, Pedersen AA, von Huth S, Sorensen GL, Holmskov U, Jiang PP, Detlefsen S, Husby S, Rathe M. No effect of deleted in malignant brain tumors 1 deficiency on chemotherapy induced murine intestinal mucositis. Sci Rep 2021; 11:14687. [PMID: 34282203 PMCID: PMC8289998 DOI: 10.1038/s41598-021-94076-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Mucositis is a serious adverse effect of chemotherapeutic treatment. During intestinal mucositis, the mucosal barrier is compromised, increasing the risk of severe infections. Mucositis necessitates dose reduction or pauses in treatment, which affect the outcome of the treatment. Deleted in malignant brain tumors 1 (DMBT1) is a secreted scavenger protein with effects on innate immunity and epithelial regeneration. We have previously shown that jejunal DMBT1 expression is increased in piglets during chemotherapeutic treatment. We hypothesized that DMBT1 ameliorates doxorubicin-induced mucositis. Individually-caged Dmbt1+/+ (WT) and Dmbt1-/- (KO) female mouse littermates received intraperitoneal injections of either doxorubicin or saline. They were euthanized after three (D3) or seven days (D7). Weight loss was monitored every day, and serum citrulline levels were measured at termination. Intestinal tissue was analyzed for the expression of DMBT1 and proinflammatory cytokines (IL-1β, IL-6, and TNF). Specimens from the small intestines and colon were scored for inflammation and epithelial and mucosal architecture changes. We detected no effect of DMBT1 on weight loss, serum citrulline levels, expression of proinflammatory cytokines, or histologic damage. We detected a significant increase in crypt depth in WT mice compared to that in KO mice on D3. In conclusion, DMBT1 does not affect doxorubicin-induced mucositis in mice.
Collapse
Affiliation(s)
- Anders B Nexoe
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
- Department of Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Andreas A Pedersen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Sebastian von Huth
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Grith L Sorensen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Ping-Ping Jiang
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Steffen Husby
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Mathias Rathe
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark.
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
26
|
Gu X, Zhang J, Li J, Wang Z, Feng J, Li J, Pan K, Ni X, Zeng D, Jing B, Zhang D. Effects of Bacillus cereus PAS38 on Immune-Related Differentially Expressed Genes of Spleen in Broilers. Probiotics Antimicrob Proteins 2021; 12:425-438. [PMID: 31243733 DOI: 10.1007/s12602-019-09567-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study mainly explored the immunomodulatory mechanisms of the probiotic Bacillus cereus PAS38 (PB) on broiler spleen. A total of 120 avian white feather broilers were randomly divided into 4 groups (N = 30), as follows: control (CNTL, fed with basal diet), PB (fed with diet supplemented with probiotic B. cereus PAS38), vaccine (VAC, fed with basal diet and injected with Newcastle disease virus vaccine), and vaccine + PB group (PBVAC, fed with basal diet supplemented with B. cereus PAS38 and injected with NDV vaccine). The experiment was conducted for 42 days. Twelve spleens were collected from four different groups, weighed, and cut into histological sections, and transcriptome analysis was performed using RNA-seq. Results of the spleen and histological section relative weights showed that feeding with probiotic B. cereus PAS38 and vaccination had a similar tendency to promote spleen development. Compared with the CNTL group, 21 immune-related genes were significantly downregulated in the PB and PBVAC groups. These genes were mainly involved in attenuating inflammatory response. The upregulated antimicrobial peptide NK-lysin and guanylate-binding protein 1 expression levels indicated that this strain enhanced the body's antimicrobial capacity. B. cereus PAS38 also amplified the broilers' immune response to the vaccine, which mainly reflected on nonspecific immunity. Hence, probiotic B. cereus PAS38 can regulate and promote the immune function of broilers.
Collapse
Affiliation(s)
- Xiaoxiao Gu
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jiao Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jiajun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Zhenhua Wang
- Chengdu Vocational College of Agricultural Science and Technology, Chengdu, 611100, China
| | - Jie Feng
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jianzhen Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
- Chengdu Vocational College of Agricultural Science and Technology, Chengdu, 611100, China
| | - Kangcheng Pan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China.
| | - Xueqin Ni
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Dong Zeng
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Bo Jing
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Dongmei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| |
Collapse
|
27
|
Blickwedel J, Bagci S, Alsat EA, Strizek B, Renner M, Müller A, Müller H. DMBT1 amount in amniotic fluid depends on gestational age. J Matern Fetal Neonatal Med 2021; 35:7058-7064. [PMID: 34107846 DOI: 10.1080/14767058.2021.1937103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Amniotic fluid is a mixture containing many different proteins as immunomodulatory peptides and growth factors. The glycoprotein Deleted in Malignant Brain Tumors 1 (DMBT1) is participated in innate immunity, angiogenesis and epithelial differentiation. We analyzed the DMBT1 concentration in amniotic fluid during gestation. METHODS DMBT1 concentration was quantified by ELISA. Amniotic fluid samples were collected from preterm and term neonates. Effects of maternal or neonatal parameters were analyzed. To evaluate the source of DMBT1 we examined RNA of fetal tissue in relation to DMBT1 expression. RESULTS The median DMBT1 concentration in amniotic fluid was 54.4 ng/ml. Amniotic fluid obtained <28 weeks of gestation revealed significantly lower DMBT1 concentrations compared to ≥28 weeks. We found a positive correlation between DMBT1 concentration and gestational age (p = .026). The fetal DMBT1 expression was pronounced in the gastrointestinal tract. CONCLUSIONS The results showed that DMBT1 concentrations in amniotic fluid correlate with the gestational age during gestation and that the fetal gastrointestinal tract is a potential source of DMBT1. BRIEF RATIONALE Amniotic fluid contains not only nutrients, but also many immunomodulatory peptides and growth factors. Deleted in Malignant Brain Tumors 1 (DMBT1) is an innate immunity protein with functions in epithelial differentiation and angiogenesis. The aim of this research was to study the DMBT1 content and the factors affecting its concentration in amniotic fluid during gestation. In summary, the results obtained in this study showed that DMBT1 is a component of amniotic fluid and that DMBT1 concentrations in amniotic fluid correlate with gestational age. In addition to this, the fetal gastrointestinal tract is a potential source of DMBT1 detected in amniotic fluid.
Collapse
Affiliation(s)
- Jana Blickwedel
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - Soyhan Bagci
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - Ebru Ailen Alsat
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - Brigitte Strizek
- Department of Obstetrics and Prenatal Medicine, University of Bonn, Bonn, Germany
| | - Marcus Renner
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Andreas Müller
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - Hanna Müller
- Neonatology and Pediatric Intensive Care, Department of Pediatrics, University of Marburg, Marburg, Germany.,Division of Neonatology and Pediatric Intensive Care, Department of Pediatrics, University Hospital Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
28
|
Samaha E, Vierlinger K, Weinhappel W, Godnic-Cvar J, Nöhammer C, Koczan D, Thiesen HJ, Yanai H, Fraifeld VE, Ziesche R. Expression Profiling Suggests Loss of Surface Integrity and Failure of Regenerative Repair as Major Driving Forces for Chronic Obstructive Pulmonary Disease Progression. Am J Respir Cell Mol Biol 2021; 64:441-452. [PMID: 33524306 DOI: 10.1165/rcmb.2020-0270oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) poses a major risk for public health, yet remarkably little is known about its detailed pathophysiology. Definition of COPD as nonreversible pulmonary obstruction revealing more about spatial orientation than about mechanisms of pathology may be a major reason for this. We conducted a controlled observational study allowing for simultaneous assessment of clinical and biological development in COPD. Sixteen healthy control subjects and 104 subjects with chronic bronchitis, with or without pulmonary obstruction at baseline, were investigated. Using both the extent of and change in bronchial obstruction as main scoring criteria for the analysis of gene expression in lung tissue, we identified 410 genes significantly associated with progression of COPD. One hundred ten of these genes demonstrated a distinctive expression pattern, with their functional annotations indicating participation in the regulation of cellular coherence, membrane integrity, growth, and differentiation, as well as inflammation and fibroproliferative repair. The regulatory pattern indicates a sequentially unfolding pathology that centers on a two-step failure of surface integrity commencing with a loss of epithelial coherence as early as chronic bronchitis. Decline of regenerative repair starting in Global Initiative for Chronic Obstructive Lung Disease stage I then activates degradation of extracellular-matrix hyaluronan, causing structural failure of the bronchial wall that is only resolved by scar formation. Although they require independent confirmation, our findings provide the first tangible pathophysiological concept of COPD to be further explored.Clinical trial registered with www.clinicaltrials.gov (NCT00618137).
Collapse
Affiliation(s)
- Eslam Samaha
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Klemens Vierlinger
- Department of Health and Environment, Austrian Institute of Technology, Vienna, Austria
| | - Wolfgang Weinhappel
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Jasminka Godnic-Cvar
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christa Nöhammer
- Department of Health and Environment, Austrian Institute of Technology, Vienna, Austria
| | - Dirk Koczan
- Department of Immunology, University of Rostock, Rostock, Germany; and
| | | | - Hagai Yanai
- Faculty of Health Sciences, Beer-Sheva Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vadim E Fraifeld
- Faculty of Health Sciences, Beer-Sheva Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rolf Ziesche
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
Hoffmann W. Trefoil Factor Family (TFF) Peptides and Their Links to Inflammation: A Re-evaluation and New Medical Perspectives. Int J Mol Sci 2021; 22:ijms22094909. [PMID: 34066339 PMCID: PMC8125380 DOI: 10.3390/ijms22094909] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Trefoil factor family peptides (TFF1, TFF2, TFF3), together with mucins, are typical exocrine products of mucous epithelia. Here, they act as a gastric tumor suppressor (TFF1) or they play different roles in mucosal innate immune defense (TFF2, TFF3). Minute amounts are also secreted as endocrine, e.g., by the immune and central nervous systems. As a hallmark, TFF peptides have different lectin activities, best characterized for TFF2, but also TFF1. Pathologically, ectopic expression occurs during inflammation and in various tumors. In this review, the role of TFF peptides during inflammation is discussed on two levels. On the one hand, the expression of TFF1-3 is regulated by inflammatory signals in different ways (upstream links). On the other hand, TFF peptides influence inflammatory processes (downstream links). The latter are recognized best in various Tff-deficient mice, which have completely different phenotypes. In particular, TFF2 is secreted by myeloid cells (e.g., macrophages) and lymphocytes (e.g., memory T cells), where it modulates immune reactions triggering inflammation. As a new concept, in addition to lectin-triggered activation, a hypothetical lectin-triggered inhibition of glycosylated transmembrane receptors by TFF peptides is discussed. Thus, TFFs are promising players in the field of glycoimmunology, such as galectins and C-type lectins.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
30
|
Lee HS, Gonzalez-Gil A, Drake V, Li TA, Schnaar RL, Kim J. Induction of the endogenous sialoglycan ligand for eosinophil death receptor Siglec-8 in chronic rhinosinusitis with hyperplastic nasal polyposis. Glycobiology 2021; 31:1026-1036. [PMID: 33755113 DOI: 10.1093/glycob/cwab018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/15/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Siglec-8, an immune-inhibitory sialoglycan binding lectin (S8), is expressed on the surface of eosinophils and mast cells, potent mediators of allergic inflammation. When S8 engages endogenous sialoglycan ligands, eosinophils undergo apoptosis and mast cell mediator release is inhibited. In the human airway, Siglec-8 ligands (S8L) are sialylated keratan sulfate chains carried on isoforms of the protein Deleted in Malignant Brain Tumors-1 (DMBT1), an immunoregulatory protein that we recently identified as the endogenous ligand for S8, DMBT1S8. We herein report that S8L is overexpressed in chronic rhinosinusitis with nasal polyposis (CRSwNP), a prevalent eosinophilic laden airway disease. Quantification and comparison of the degree to which DMBT1 carries the S8L by immunoblot analysis and lectin blot overlay, respectively, from nasal lavage showed that the S8L/DMBT1 ratio was significantly increased in CRSwNP vs control or CRS patients. We identified the histological sites of S8L and DMBT1 expression in fresh surgically resected human nasal polyps. Histochemistry of diseased polyps and adjacent non-diseased middle turbinate tissue from CRSwNP demonstrated colocalization of S8L and DMBT1 with highest staining in submucosal glands >> epithelium > stoma. S8L expression was specifically elevated in the submucosal glands and epithelium of polyp tissue compared to middle turbinate. We hypothesize that expression of the isoform of DMBT1 carrying the Siglec-8 binding sialoglycan, DMBT1S8, is induced in polyps of CRSwNP specifically at the site of disease, is produced in the submucosal glands of polyps and secreted into the lumen of the sinonasal cavity as a host response to mitigate eosinophil-mediated inflammation.
Collapse
Affiliation(s)
- Hyun Sil Lee
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA.,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Anabel Gonzalez-Gil
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Virginia Drake
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - T August Li
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Ronald L Schnaar
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Jean Kim
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA.,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| |
Collapse
|
31
|
Han G, Sinjab A, Hara K, Treekitkarnmongkol W, Brennan P, Chang K, Bogatenkova E, Sanchez-Espiridion B, Behrens C, Solis LM, Gao B, Girard L, Zhang J, Sepesi B, Cascone T, Byers LA, Gibbons DL, Chen J, Moghaddam SJ, Ostrin EJ, Scheet P, Fujimoto J, Shay J, Heymach JV, Minna JD, Dubinett S, Wistuba II, Stevenson CS, Spira AE, Wang L, Kadara H. Single-Cell Expression Landscape of SARS-CoV-2 Receptor ACE2 and Host Proteases in Normal and Malignant Lung Tissues from Pulmonary Adenocarcinoma Patients. Cancers (Basel) 2021; 13:cancers13061250. [PMID: 33809063 PMCID: PMC7998226 DOI: 10.3390/cancers13061250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The coronavirus disease 2019 (COVID-19) pandemic continues to spread rapidly on a global scale. When presenting with severe respiratory complications, COVID-19 results in markedly high death rates, particularly among patients with comorbidities such as cancer. Motivated by the ongoing global health crisis, we leveraged a growing in-house cohort of pulmonary tissues from lung cancer patients to analyze, at high resolution, the expression of host proteins implicated in the entryway of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into lung epithelial cells. Our results identify key pathways in lung pathobiology and inflammation that offer the potential to identify novel markers and therapeutic targets that can be repurposed for clinical management of COVID-19, particularly among lung cancer patients, a population that represents over half a million individuals in the United States alone. Abstract The novel coronavirus SARS-CoV-2 is the causative agent of the COVID-19 pandemic. Severely symptomatic COVID-19 is associated with lung inflammation, pneumonia, and respiratory failure, thereby raising concerns of elevated risk of COVID-19-associated mortality among lung cancer patients. Angiotensin-converting enzyme 2 (ACE2) is the major receptor for SARS-CoV-2 entry into lung cells. The single-cell expression landscape of ACE2 and other SARS-CoV-2-related genes in pulmonary tissues of lung cancer patients remains unknown. We sought to delineate single-cell expression profiles of ACE2 and other SARS-CoV-2-related genes in pulmonary tissues of lung adenocarcinoma (LUAD) patients. We examined the expression levels and cellular distribution of ACE2 and SARS-CoV-2-priming proteases TMPRSS2 and TMPRSS4 in 5 LUADs and 14 matched normal tissues by single-cell RNA-sequencing (scRNA-seq) analysis. scRNA-seq of 186,916 cells revealed epithelial-specific expression of ACE2, TMPRSS2, and TMPRSS4. Analysis of 70,030 LUAD- and normal-derived epithelial cells showed that ACE2 levels were highest in normal alveolar type 2 (AT2) cells and that TMPRSS2 was expressed in 65% of normal AT2 cells. Conversely, the expression of TMPRSS4 was highest and most frequently detected (75%) in lung cells with malignant features. ACE2-positive cells co-expressed genes implicated in lung pathobiology, including COPD-associated HHIP, and the scavengers CD36 and DMBT1. Notably, the viral scavenger DMBT1 was significantly positively correlated with ACE2 expression in AT2 cells. We describe normal and tumor lung epithelial populations that express SARS-CoV-2 receptor and proteases, as well as major host defense genes, thus comprising potential treatment targets for COVID-19 particularly among lung cancer patients.
Collapse
Affiliation(s)
- Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | - Kieko Hara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | - Warapen Treekitkarnmongkol
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | - Patrick Brennan
- Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.B.); (E.B.)
| | - Kyle Chang
- Guardant Health, Redwood City, CA 94063, USA;
| | - Elena Bogatenkova
- Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.B.); (E.B.)
| | - Beatriz Sanchez-Espiridion
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | - Carmen Behrens
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.B.); (J.Z.); (T.C.); (L.A.B.); (D.L.G.); (J.V.H.)
| | - Luisa M. Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | - Boning Gao
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, TX 75390, USA; (B.G.); (L.G.); (J.D.M.)
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, TX 75390, USA; (B.G.); (L.G.); (J.D.M.)
| | - Jianjun Zhang
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.B.); (J.Z.); (T.C.); (L.A.B.); (D.L.G.); (J.V.H.)
| | - Boris Sepesi
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77005, USA;
| | - Tina Cascone
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.B.); (J.Z.); (T.C.); (L.A.B.); (D.L.G.); (J.V.H.)
| | - Lauren A. Byers
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.B.); (J.Z.); (T.C.); (L.A.B.); (D.L.G.); (J.V.H.)
| | - Don L. Gibbons
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.B.); (J.Z.); (T.C.); (L.A.B.); (D.L.G.); (J.V.H.)
| | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.); (S.J.M.)
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.); (S.J.M.)
| | - Edwin J. Ostrin
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Paul Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA;
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | - Jerry Shay
- Department of Cell Biology, University of Texas Southwestern, Dallas, TX 75390, USA;
| | - John V. Heymach
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.B.); (J.Z.); (T.C.); (L.A.B.); (D.L.G.); (J.V.H.)
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern, Dallas, TX 75390, USA; (B.G.); (L.G.); (J.D.M.)
| | - Steven Dubinett
- Department of Medicine, The University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Ignacio I. Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
| | | | - Avrum E. Spira
- Lung Cancer Initiative at Johnson and Johnson, Cambridge, MA 02142, USA; (C.S.S.); (A.E.S.)
- Section of Computational Biomedicine, Boston University, Boston, MA 02215, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (L.W.); (H.K.)
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (K.H.); (W.T.); (B.S.-E.); (L.M.S.); (J.F.); (I.I.W.)
- Correspondence: (L.W.); (H.K.)
| |
Collapse
|
32
|
Davey PA, Power AM, Santos R, Bertemes P, Ladurner P, Palmowski P, Clarke J, Flammang P, Lengerer B, Hennebert E, Rothbächer U, Pjeta R, Wunderer J, Zurovec M, Aldred N. Omics-based molecular analyses of adhesion by aquatic invertebrates. Biol Rev Camb Philos Soc 2021; 96:1051-1075. [PMID: 33594824 DOI: 10.1111/brv.12691] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Many aquatic invertebrates are associated with surfaces, using adhesives to attach to the substratum for locomotion, prey capture, reproduction, building or defence. Their intriguing and sophisticated biological glues have been the focus of study for decades. In all but a couple of specific taxa, however, the precise mechanisms by which the bioadhesives stick to surfaces underwater and (in many cases) harden have proved to be elusive. Since the bulk components are known to be based on proteins in most organisms, the opportunities provided by advancing 'omics technologies have revolutionised bioadhesion research. Time-consuming isolation and analysis of single molecules has been either replaced or augmented by the generation of massive data sets that describe the organism's translated genes and proteins. While these new approaches have provided resources and opportunities that have enabled physiological insights and taxonomic comparisons that were not previously possible, they do not provide the complete picture and continued multi-disciplinarity is essential. This review covers the various ways in which 'omics have contributed to our understanding of adhesion by aquatic invertebrates, with new data to illustrate key points. The associated challenges are highlighted and priorities are suggested for future research.
Collapse
Affiliation(s)
- Peter A Davey
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, U.K
| | - Anne Marie Power
- Ryan Institute, School of Natural Sciences, National University of Ireland Galway, Room 226, Galway, H91 TK33, Ireland
| | - Romana Santos
- Departamento de Biologia Animal, Faculdade de Ciências, Centro de Ciências do Mar e do Ambiente (MARE), Universidade de Lisboa, Lisbon, 1749-016, Portugal
| | - Philip Bertemes
- Institute of Zoology and Center of Molecular Biosciences Innsbruck, University of Innsbruck, Technikerstrasse 25, Innsbruck, 6020, Austria
| | - Peter Ladurner
- Institute of Zoology and Center of Molecular Biosciences Innsbruck, University of Innsbruck, Technikerstrasse 25, Innsbruck, 6020, Austria
| | - Pawel Palmowski
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, U.K
| | - Jessica Clarke
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, U.K
| | - Patrick Flammang
- Biology of Marine Organisms and Biomimetics Unit, Research Institute for Biosciences, University of Mons, Place du Parc 23, Mons, 7000, Belgium
| | - Birgit Lengerer
- Institute of Zoology and Center of Molecular Biosciences Innsbruck, University of Innsbruck, Technikerstrasse 25, Innsbruck, 6020, Austria
| | - Elise Hennebert
- Laboratory of Cell Biology, Research Institute for Biosciences, University of Mons, Place du Parc 23, Mons, 7000, Belgium
| | - Ute Rothbächer
- Institute of Zoology and Center of Molecular Biosciences Innsbruck, University of Innsbruck, Technikerstrasse 25, Innsbruck, 6020, Austria
| | - Robert Pjeta
- Institute of Zoology and Center of Molecular Biosciences Innsbruck, University of Innsbruck, Technikerstrasse 25, Innsbruck, 6020, Austria
| | - Julia Wunderer
- Institute of Zoology and Center of Molecular Biosciences Innsbruck, University of Innsbruck, Technikerstrasse 25, Innsbruck, 6020, Austria
| | - Michal Zurovec
- Biology Centre of the Czech Academy of Sciences and Faculty of Sciences, University of South Bohemia, České Budějovice, 370 05, Czech Republic
| | - Nick Aldred
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, U.K
| |
Collapse
|
33
|
Nakata M, Kreikemeyer B. Genetics, Structure, and Function of Group A Streptococcal Pili. Front Microbiol 2021; 12:616508. [PMID: 33633705 PMCID: PMC7900414 DOI: 10.3389/fmicb.2021.616508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Streptococcus pyogenes (Group A Streptococcus; GAS) is an exclusively human pathogen. This bacterial species is responsible for a large variety of infections, ranging from purulent but mostly self-limiting oropharynx/skin diseases to streptococcal sequelae, including glomerulonephritis and rheumatic fever, as well as life-threatening streptococcal toxic-shock syndrome. GAS displays a wide array of surface proteins, with antigenicity of the M protein and pili utilized for M- and T-serotyping, respectively. Since the discovery of GAS pili in 2005, their genetic features, including regulation of expression, and structural features, including assembly mechanisms and protein conformation, as well as their functional role in GAS pathogenesis have been intensively examined. Moreover, their potential as vaccine antigens has been studied in detail. Pilus biogenesis-related genes are located in a discrete section of the GAS genome encoding fibronectin and collagen binding proteins and trypsin-resistant antigens (FCT region). Based on the heterogeneity of genetic composition and DNA sequences, this region is currently classified into nine distinguishable forms. Pili and fibronectin-binding proteins encoded in the FCT region are known to be correlated with infection sites, such as the skin and throat, possibly contributing to tissue tropism. As also found for pili of other Gram-positive bacterial pathogens, GAS pilin proteins polymerize via isopeptide bonds, while intramolecular isopeptide bonds present in the pilin provide increased resistance to degradation by proteases. As supported by findings showing that the main subunit is primarily responsible for T-serotyping antigenicity, pilus functions and gene expression modes are divergent. GAS pili serve as adhesins for tonsillar tissues and keratinocyte cell lines. Of note, a minor subunit is considered to have a harpoon function by which covalent thioester bonds with host ligands are formed. Additionally, GAS pili participate in biofilm formation and evasion of the immune system in a serotype/strain-specific manner. These multiple functions highlight crucial roles of pili during the onset of GAS infection. This review summarizes the current state of the art regarding GAS pili, including a new mode of host-GAS interaction mediated by pili, along with insights into pilus expression in terms of tissue tropism.
Collapse
Affiliation(s)
- Masanobu Nakata
- Department of Oral Microbiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University of Rostock, Rostock, Germany
| |
Collapse
|
34
|
Boziki M, Grigoriadis N, Papaefthymiou A, Doulberis M, Polyzos SA, Gavalas E, Deretzi G, Karafoulidou E, Kesidou E, Taloumtzis C, Theotokis P, Sofou E, Katsinelos P, Vardaka E, Fludaras I, Touloumtzi M, Koukoufiki A, Simeonidou C, Liatsos C, Kountouras J. The trimebutine effect on Helicobacter pylori-related gastrointestinal tract and brain disorders: A hypothesis. Neurochem Int 2021; 144:104938. [PMID: 33535070 DOI: 10.1016/j.neuint.2020.104938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/17/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
The localization of bacterial components and/or metabolites in the central nervous system may elicit neuroinflammation and/or neurodegeneration. Helicobacter pylori (a non-commensal symbiotic gastrointestinal pathogen) infection and its related metabolic syndrome have been implicated in the pathogenesis of gastrointestinal tract and central nervous system disorders, thus medications affecting the nervous system - gastrointestinal tract may shape the potential of Helicobacter pylori infection to trigger these pathologies. Helicobacter pylori associated metabolic syndrome, by impairing gut motility and promoting bacterial overgrowth and translocation, might lead to brain pathologies. Trimebutine maleate is a prokinetic drug that hastens gastric emptying, by inducing the release of gastrointestinal agents such as motilin and gastrin. Likewise, it appears to protect against inflammatory signal pathways, involved in inflammatory disorders including brain pathologies. Trimebutine maleate also acts as an antimicrobial agent and exerts opioid agonist effect. This study aimed to investigate a hypothesis regarding the recent advances in exploring the potential role of gastrointestinal tract microbiota dysbiosis-related metabolic syndrome and Helicobacter pylori in the pathogenesis of gastrointestinal tract and brain diseases. We hereby proposed a possible neuroprotective role for trimebutine maleate by altering the dynamics of the gut-brain axis interaction, thus suggesting an additional effect of trimebutine maleate on Helicobacter pylori eradication regimens against these pathologies.
Collapse
Affiliation(s)
- Marina Boziki
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Nikolaos Grigoriadis
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Apostolis Papaefthymiou
- Department of Gastroenterology, University Hospital of Larissa, Larissa, 41110, Greece; Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece; First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Michael Doulberis
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece; First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece; Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, Aarau, 5001, Switzerland
| | - Stergios A Polyzos
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece; First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Emmanuel Gavalas
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece
| | - Georgia Deretzi
- Department of Neurology, Papageorgiou General Hospital, Thessaloniki, 56429, Macedonia, Greece
| | - Eleni Karafoulidou
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Evangelia Kesidou
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Charilaos Taloumtzis
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece; 424 General Military Hospital of Thessaloniki, Department of Gastroenterology, Thessaloniki, 56429, Macedonia, Greece
| | - Paschalis Theotokis
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Electra Sofou
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Panagiotis Katsinelos
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece
| | - Elisabeth Vardaka
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece; Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, 574 00, Thessaloniki, Macedonia, Greece
| | - Ioannis Fludaras
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece
| | - Maria Touloumtzi
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece
| | - Argiro Koukoufiki
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece
| | - Constantina Simeonidou
- Laboratory of Experimental Physiology, Department of Physiology and Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54124, Macedonia, Greece
| | - Christos Liatsos
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece; Department of Gastroenterology, 401 Army General Hospital of Athens, Athens, 115 25, Greece
| | - Jannis Kountouras
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, 546 42, Macedonia, Greece.
| |
Collapse
|
35
|
Manzer HS, Nobbs AH, Doran KS. The Multifaceted Nature of Streptococcal Antigen I/II Proteins in Colonization and Disease Pathogenesis. Front Microbiol 2020; 11:602305. [PMID: 33329493 PMCID: PMC7732690 DOI: 10.3389/fmicb.2020.602305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022] Open
Abstract
Streptococci are Gram-positive bacteria that belong to the natural microbiota of humans and animals. Certain streptococcal species are known as opportunistic pathogens with the potential to cause severe invasive disease. Antigen I/II (AgI/II) family proteins are sortase anchored cell surface adhesins that are nearly ubiquitous across streptococci and contribute to many streptococcal diseases, including dental caries, respiratory tract infections, and meningitis. They appear to be multifunctional adhesins with affinities to various host substrata, acting to mediate attachment to host surfaces and stimulate immune responses from the colonized host. Here we will review the literature including recent work that has demonstrated the multifaceted nature of AgI/II family proteins, focusing on their overlapping and distinct functions and their important contribution to streptococcal colonization and disease.
Collapse
Affiliation(s)
- Haider S. Manzer
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angela H. Nobbs
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Kelly S. Doran
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
36
|
Reichhardt MP, Messing M, Andersson S, Kolho KL, Meri S. Intestinal SALSA/dmbt1 levels are decreased in prematurely born infants. Scand J Immunol 2020; 93:e12987. [PMID: 33047342 DOI: 10.1111/sji.12987] [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: 04/25/2020] [Revised: 09/09/2020] [Accepted: 10/06/2020] [Indexed: 11/29/2022]
Abstract
The first months of life represent a crucial time period for an infant. Alongside establishing the early microbiome, the mucosal immunological homeostasis is being developed. Both processes may be perturbed in prematurely born infants. The glycoprotein SALSA plays a role in mucosal inflammation and microbial clearance. It is one of the most abundant molecules on the intestinal mucosal surfaces in early life. SALSA binds to many types of microbes and host defence molecules like IgA, C1q and collectin molecules. We here describe the development in faecal SALSA levels during the first three months of life. During these 90 days, the median SALSA level in full-term babies decreased from 1100 μg/mL (range 49-17 000 μg/mL) to 450 μg/mL (range 33-1000 μg/mL). Lower levels of SALSA were observed in prematurely born infants in the same time period. Our novel observation thus indicates an impact of prematurity on an important component of the infant intestinal immune system. Changes in SALSA in early life may have an effect on the early establishment of the human microbiome.
Collapse
Affiliation(s)
- Martin Parnov Reichhardt
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Marcel Messing
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Sture Andersson
- Department of Pediatrics and Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Kaija-Leena Kolho
- Department of Pediatrics and Children's Hospital, University of Helsinki, Helsinki, Finland.,Faculty of Medicine and HealthTechnology, Tampere University, Tampere, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
37
|
Papini E, Tavano R, Mancin F. Opsonins and Dysopsonins of Nanoparticles: Facts, Concepts, and Methodological Guidelines. Front Immunol 2020; 11:567365. [PMID: 33154748 PMCID: PMC7587406 DOI: 10.3389/fimmu.2020.567365] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/25/2020] [Indexed: 11/13/2022] Open
Abstract
Understanding the effects mediated by a set of nanoparticle (NP)-bound host biomolecules, often indicated with the umbrella term of NP corona, is essential in nanomedicine, nanopharmacology, and nanotoxicology. Among the NP-adsorbed proteome, some factors mediate cell binding, endocytosis, and clearing by macrophages and other phagocytes (opsonins), while some others display few affinities for the cell surface (dysopsonins). The functional mapping of opsonins and dysopsonins is instrumental to design long-circulating and nanotoxicologically safe next-generation nanotheranostics. In this review, we critically analyze functional data identifying specific proteins with opsonin or dysopsonin properties. Special attention is dedicated to the following: (1) the simplicity or complexity of the NP proteome and its modulation, (2) the role of specific host proteins in mediating the stealth properties of uncoated or polymer-coated NPs, and (3) the ability of the innate immune system, and, in particular, of the complement proteins, to mediate NP clearance by phagocytes. Emerging species-specific peculiarities, differentiating humans from preclinical animal models (the murine especially), are highlighted throughout this overview. The operative definition of opsonin and dysopsonin and the measurement schemes to assess their in vitro efficacy is critically re-examined. This provides a shared and unbiased approach useful for NP opsonin and dysopsonin systematic identification.
Collapse
Affiliation(s)
- Emanuele Papini
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Centre for Innovative Biotechnological Research, University of Padua, Padua, Italy
| | - Regina Tavano
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Centre for Innovative Biotechnological Research, University of Padua, Padua, Italy
| | - Fabrizio Mancin
- Department of Chemical Sciences, University of Padua, Padua, Italy
| |
Collapse
|
38
|
Gonzalez-Gil A, Li TA, Porell RN, Fernandes SM, Tarbox HE, Lee HS, Aoki K, Tiemeyer M, Kim J, Schnaar RL. Isolation, identification, and characterization of the human airway ligand for the eosinophil and mast cell immunoinhibitory receptor Siglec-8. J Allergy Clin Immunol 2020; 147:1442-1452. [PMID: 32791164 DOI: 10.1016/j.jaci.2020.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/24/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The immunoinhibitory receptor Siglec-8 on the surface of human eosinophils and mast cells binds to sialic acid-containing ligands in the local milieu, resulting in eosinophil apoptosis, inhibition of mast cell degranulation, and suppression of inflammation. Siglec-8 ligands were found on postmortem human trachea and bronchi and on upper airways in 2 compartments, cartilage and submucosal glands, but they were surprisingly absent from the epithelium. We hypothesized that Siglec-8 ligands in submucosal glands and ducts are normally transported to the airway mucus layer, which is lost during tissue preparation. OBJECTIVE Our aim was to identify the major Siglec-8 sialoglycan ligand on the mucus layer of human airways. METHODS Human upper airway mucus layer proteins were recovered during presurgical nasal lavage of patients at a sinus clinic. Proteins were resolved by gel electrophoresis and blotted, and Siglec-8 ligands detected. Ligands were purified by size exclusion and affinity chromatography, identified by proteomic mass spectrometry, and validated by electrophoretic and histochemical colocalization. The affinity of Siglec-8 binding to purified human airway ligand was determined by inhibition of glycan binding. RESULTS A Siglec-8-ligand with a molecular weight of approximately 1000 kDa was found in all patient nasal lavage samples. Purification and identification revealed deleted in malignant brain tumors 1 (DMBT1) (also known by the aliases GP340 and SALSA), a large glycoprotein with multiple O-glycosylation repeats. Immunoblotting, immunohistochemistry, and enzyme treatments confirmed that Siglec-8 ligand on the human airway mucus layer is an isoform of DMBT1 carrying O-linked sialylated keratan sulfate chains (DMBT1S8). Quantitative inhibition revealed that DMBT1S8 has picomolar affinity for Siglec-8. CONCLUSION A distinct DMBT1 isoform, DMBT1S8, is the major high-avidity ligand for Siglec-8 on human airways.
Collapse
Affiliation(s)
- Anabel Gonzalez-Gil
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Md
| | - T August Li
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Ryan N Porell
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Steve M Fernandes
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Haley E Tarbox
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Hyun Sil Lee
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, Ga
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, Ga
| | - Jean Kim
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Ronald L Schnaar
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
39
|
Ma N, Zhao Y. DMBT1 suppresses cell proliferation, migration and invasion in ovarian cancer and enhances sensitivity to cisplatin through galectin-3/PI3k/Akt pathway. Cell Biochem Funct 2020; 38:801-809. [PMID: 32424818 DOI: 10.1002/cbf.3549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/13/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022]
Abstract
Ovarian cancer (OC) is one of the most common gynaecologic malignancies. Deleted in malignant brain tumors 1 (DMBT1) was considered as a tumour suppressor in multiple cancers, but there have been no systemic profiling studies of DMBT1 in OC until now. The aim of this study is to explore the role and the potential mechanism of DMBT1 in OC. mRNA levels and protein expressions of corresponding genes were detected by quantitative real-time polymerase chain reaction and western blot. Cell proliferation was detected by CCK-8 assay and cell colony formation. Cell migration and invasion were detected by wound healing and transwell assay. The combination between DMBT1 and galectin-3 was demonstrated by immunoprecipitation. We demonstrated that DMBT1 was downregulated in OC cell lines, especially SKOV3 cells. Overexpression of DMBT1 significantly inhibited cell proliferation, colony formation, migration and invasion, as well as decreased Matrix Metalloproteinase-2 (MMP-2) and MMP-7. DMBT1 caused a reduction of cell viability by treatment with cisplatin. Immunoprecipitation assay revealed a combination between DMBT1 and galectin-3. DMBT1 could decrease the expression of galectin-3 and inhibit the phosphorylation of PI3K and AKT, while overexpression of galectin-3 reversed this effect. In summary, DMBT1 might inhibit the progression of OC and improve the sensitivity of SKOV3 cells to cisplatin through galectin-3/PI3K/AKT pathway, giving a new insight into the role of DMBT1 in OC and enriching the potential strategies for OC treatment. SIGNIFICANCE OF THE STUDY: The present study focus on the role and the potential mechanism of DMBT1 in ovarian cancer (OC). We demonstrated that DMBT1 might inhibit the progression of ovarian by inhibiting cell proliferation, migration and invasion and increased the sensitivity to cisplatin through galectin-3/PI3K/AKT pathway. The findings ensure the interaction relation between DMBT1 and galectin-3 in OC, providing a novel biological marker for OC and enriching the potential strategies for OC treatment.
Collapse
Affiliation(s)
- Nan Ma
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yuqing Zhao
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
40
|
Zhang S, Huo X, Li M, Hou R, Cong X, Xu X. Oral antimicrobial activity weakened in children with electronic waste lead exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:14763-14770. [PMID: 32056098 DOI: 10.1007/s11356-020-08037-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 02/10/2020] [Indexed: 02/05/2023]
Abstract
Environmental lead (Pb) exposure can induce dysbacteriosis, impair oral health, and is associated with the development of dental caries. However, the mechanism is unclear. The aim of this study was to explore the effects of Pb toxicity on oral antimicrobial activity in children in an e-waste area. Results showed higher blood Pb levels in e-waste-exposed group children, accompanied by decreased saliva SAG (salivary agglutinin) concentrations, increased peripheral WBC (white blood cell) counts and monocyte counts, and elevated peripheral monocyte percentage. LnPb (natural logarithmic transformation of blood Pb level) was negatively correlated with saliva SAG concentration, while positively correlated with peripheral monocyte percentage. Saliva SAG concentration played a complete mediating role in the correlation of LnPb to peripheral monocyte percentage. To our knowledge, this is the first study on the relationship of environmental Pb exposure and oral antimicrobial activity in children, showing that environmental Pb exposure may weaken oral antimicrobial activity through reducing saliva SAG concentration, which may raise the risk of oral dysbacteriosis and ultimately pathogen infection.
Collapse
Affiliation(s)
- Shaocheng Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511486, Guangdong, China
| | - Minghui Li
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
| | - Ruikun Hou
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
| | - Xiaowei Cong
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China.
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
41
|
Pan X, Zhang D, Nguyen DN, Wei W, Yu X, Gao F, Sangild PT. Postnatal Gut Immunity and Microbiota Development Is Minimally Affected by Prenatal Inflammation in Preterm Pigs. Front Immunol 2020; 11:420. [PMID: 32265914 PMCID: PMC7098537 DOI: 10.3389/fimmu.2020.00420] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
Chorioamnionitis (CA), resulting from intra-amniotic inflammation, is a frequent cause of preterm birth and exposes the immature intestine to bacterial toxins and/or inflammatory mediators before birth via fetal swallowing. This may affect intestinal immune development, interacting with the effects of enteral feeding and gut microbiota colonization just after birth. Using preterm pigs as model for preterm infants, we hypothesized that prenatal exposure to gram-negative endotoxin influences postnatal bacterial colonization and gut immune development. Pig fetuses were given intra-amniotic lipopolysaccharide (LPS) 3 days before preterm delivery by cesarean section and were compared with littermate controls (CON) at birth and after 5 days of formula feeding and spontaneous bacterial colonization. Amniotic fluid was collected for analysis of leukocyte counts and cytokines, and the distal small intestine was analyzed for endotoxin level, morphology, and immune cell counts. Intestinal gene expression and microbiota were analyzed by transcriptomics and metagenomics, respectively. At birth, LPS-exposed pigs showed higher intestinal endotoxin, neutrophil/macrophage density, and shorter villi. About 1.0% of intestinal genes were affected at birth, and DMBT1, a regulator of mucosal immune defense, was identified as the hub gene in the co-expression network. Genes related to innate immune response (TLR2, LBP, CD14, C3, SFTPD), neutrophil chemotaxis (C5AR1, CSF3R, CCL5), and antigen processing (MHC II genes and CD4) were also affected, and expression levels correlated with intestinal neutrophil/macrophage density and amniotic fluid cytokine levels. On day 5, LPS and CON pigs showed similar sensitivity to necrotizing enterocolitis, endotoxin levels, morphology, immune cell counts, gene expressions, and microbiota composition (except for difference in some low-abundant species). Our results show that CA markedly affects intestinal genes at preterm birth, including genes related to immune cell infiltration. However, a few days later, following the physiological adaptations to preterm birth, CA had limited effects on intestinal structure, function, gene expression, bacterial colonization, and necrotizing enterocolitis sensitivity. We conclude that short-term, prenatal intra-amniotic inflammation is unlikely to exert marked effects on intestinal immune development in preterm neonates beyond the immediate neonatal period.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Du Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Duc Ninh Nguyen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Wei Wei
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xinxin Yu
- Lingnan Guangdong Laboratory of Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Fei Gao
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Lingnan Guangdong Laboratory of Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Per T Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Neonatology, Rigshospitalet, Copenhagen, Denmark.,Department of Pediatrics, Odense University Hospital, Odense, Denmark
| |
Collapse
|
42
|
Reichhardt MP, Loimaranta V, Lea SM, Johnson S. Structures of SALSA/DMBT1 SRCR domains reveal the conserved ligand-binding mechanism of the ancient SRCR fold. Life Sci Alliance 2020; 3:3/4/e201900502. [PMID: 32098784 PMCID: PMC7043408 DOI: 10.26508/lsa.201900502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 02/06/2023] Open
Abstract
The structures of SALSA SRCR domains 1 and 8 reveal a cation-dependent mechanism for ligand recognition, contributing to important roles in the immune system and cellular signalling. The cation-binding sites are conserved across all SRCR domains, suggesting conserved functional mechanisms. The scavenger receptor cysteine-rich (SRCR) family of proteins comprises more than 20 membrane-associated and secreted molecules. Characterised by the presence of one or more copies of the ∼110 amino-acid SRCR domain, this class of proteins have widespread functions as antimicrobial molecules, scavenger receptors, and signalling receptors. Despite the high level of structural conservation of SRCR domains, no unifying mechanism for ligand interaction has been described. The SRCR protein SALSA, also known as DMBT1/gp340, is a key player in mucosal immunology. Based on detailed structural data of SALSA SRCR domains 1 and 8, we here reveal a novel universal ligand-binding mechanism for SALSA ligands. The binding interface incorporates a dual cation-binding site, which is highly conserved across the SRCR superfamily. Along with the well-described cation dependency on most SRCR domain–ligand interactions, our data suggest that the binding mechanism described for the SALSA SRCR domains is applicable to all SRCR domains. We thus propose to have identified in SALSA a conserved functional mechanism for the SRCR class of proteins.
Collapse
Affiliation(s)
| | | | - Susan M Lea
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.,Central Oxford Structural Molecular Imaging Centre, University of Oxford, Oxford, UK
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
43
|
Zhao Y, Tao Q, Wu J, Liu H. DMBT1 has a protective effect on allergic rhinitis. Biomed Pharmacother 2019; 121:109675. [PMID: 31810134 DOI: 10.1016/j.biopha.2019.109675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE Deleted in malignant brain tumors 1 (DMBT1) is a secreted scavenger receptor cysteine-rich (SRCR) protein, predominantly expressed in nasal mucosal epithelial cells. In previous experiments, we found large amounts of DMBT1 present in the nasal cavity mucosa of allergic rhinitis (AR) patients. However, the exact role of DMBT1 in AR remains unclear. This study is to investigate the mechanism through which DMBT1 exerts its effects on AR progression. METHOD DMBT1 levels in the nasal lavage (NL) fluid and the nasal mucosa in AR and control groups were determined by ELISA and IHC. Next, mice were sensitized with ovalbumin; intranasal administrations of recombinant DMBT1 were then performed. DMBT1 in the nasal mucosa of mice were determined by IHC and WB. Nasal symptoms were estimated. IL-4 and IL-5 in BAL fluid and eosinophils infiltration in the nasal cavity were measured through ELISA and HE staining, respectively. RESULTS DMBT1 levels were found to be significantly higher in the NL fluid and nasal mucosa of AR patients and AR mice, compared to control groups (p < 0.01). Levels of IL-4 and IL-5 in BAL and infiltration of eosinophils into the nasal mucosa were significantly increased in AR mice, compared to control mice (p < 0.01). rDMBT1 significantly reduced the number of nasal sneezing and rubbings in AR mice (p < 0.01). It also inhibited the eosinophil infiltration in the nasal mucosa and significantly decreased the production of IL-4 and IL-5 in BAL (p < 0.01). CONCLUSION This study demonstrated that rDMBT1 alleviates AR progression in mice via inhibiting IL-4 and IL-5 production and eosinophils infiltration in the nasal cavity.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, China
| | - Qilei Tao
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, China
| | - Jian Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, China.
| | - Huanhai Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, China.
| |
Collapse
|
44
|
DMBT1 inhibition of Pseudomonas aeruginosa twitching motility involves its N-glycosylation and cannot be conferred by the Scavenger Receptor Cysteine-Rich bacteria-binding peptide domain. Sci Rep 2019; 9:13146. [PMID: 31511582 PMCID: PMC6739395 DOI: 10.1038/s41598-019-49543-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/22/2019] [Indexed: 01/18/2023] Open
Abstract
The scavenging capacity of glycoprotein DMBT1 helps defend mucosal epithelia against microbes. DMBT1 binding to multiple bacterial species involves its conserved Scavenger Receptor Cysteine-Rich (SRCR) domains, localized to a 16-mer consensus sequence peptide, SRCRP2. Previously, we showed that DMBT1 bound Pseudomonas aeruginosa pili, and inhibited twitching motility, a pilus-mediated movement important for virulence. Here, we determined molecular characteristics required for twitching motility inhibition. Heat-denatured DMBT1 lost capacity to inhibit twitching motility and showed reduced pili binding (~40%). Size-exclusion chromatography of Lys-C-digested native DMBT1 showed that only high-Mw fractions retained activity, suggesting involvement of the N-terminal containing repeated SRCR domains with glycosylated SRCR-Interspersed Domains (SIDs). However, individual or pooled consensus sequence peptides (SRCRPs 1 to 7) showed no activity and did not bind P. aeruginosa pili; nor did recombinant DMBT1 (aa 1–220) or another SRCR-rich glycoprotein, CD163. Enzymatic de-N-glycosylation of DMBT1, but not de-O-glycosylation, reduced its capacity to inhibit twitching motility (~57%), without reducing pili binding. Therefore, DMBT1 inhibition of P. aeruginosa twitching motility involves its N-glycosylation, its pili-binding capacity is insufficient, and it cannot be conferred by the SRCR bacteria-binding peptide domain, either alone or mixed with other unlinked SRCRPs, suggesting an additional mechanism for DMBT1-mediated mucosal defense.
Collapse
|
45
|
Shin D, Chang SY, Bogere P, Won K, Choi JY, Choi YJ, Lee HK, Hur J, Park BY, Kim Y, Heo J. Beneficial roles of probiotics on the modulation of gut microbiota and immune response in pigs. PLoS One 2019; 14:e0220843. [PMID: 31461453 PMCID: PMC6713323 DOI: 10.1371/journal.pone.0220843] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
The importance of probiotics in swine production is widely acknowledged as crucial. However, gaps still remain in the exact roles played by probiotics in modulation of gut microbiota and immune response. This study determined the roles of probiotic Lactobacillus plantarum strain JDFM LP11in gut microbiota modulation and immune response in weaned piglets. L. plantarum JDFM LP11 increased the population of lactic acid bacteria in feces and enhanced the development of villi in the small intestine. Metagenome analysis showed that microbial diversity and richness (Simpson, Shannon, ACE, Chao1) and the relative abundance of the Firmicutes were higher in weaned piglets fed probiotics. Five bacterial families were different in the relative abundance, especially; Prevotellaceae occupied the largest part of microbial community showed the most difference between two groups. Transcriptome analysis identified 25 differentially expressed genes using RNA-sequencing data of the ileum. Further gene ontology and immune DB analysis determined 8 genes associated with innate defense response and cytokine production. BPI, RSAD2, SLPI, LUM, OLFM4, DMBT1 and C6 genes were down-regulated by probiotic supplementation except PLA2G2A. PICRUSt analysis predicting functional profiling of microbial communities indicated branched amino acid biosynthesis and butyrate metabolism promoting gut development and health were increased by probiotics. Altogether, our data suggest that L. plantarum JDFM LP11 increases the diversity and richness in the microbial community, and attenuates the ileal immune gene expression towards gut inflammation, promoting intestinal development in weaned piglets.
Collapse
Affiliation(s)
- Donghyun Shin
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, Republic of Korea
| | - Sung Yong Chang
- Department of Animal Science and Institute of Milk Genomics, Chonbuk National University, Jeonju, Republic of Korea
| | - Paul Bogere
- Department of Agricultural Convergence Technology, Chonbuk National University, Jeonju, Republic of Korea
| | - KyeongHye Won
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, Republic of Korea
| | - Jae-Young Choi
- The Animal Molecular Genetics and Breeding Center, Chonbuk National University, Jeonju, Republic of Korea
| | - Yeon-Jae Choi
- International Agricultural Development and Cooperation Center, Chonbuk National University, Jeonju, Republic of Korea
| | - Hak Kyo Lee
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, Republic of Korea
- The Animal Molecular Genetics and Breeding Center, Chonbuk National University, Jeonju, Republic of Korea
| | - Jin Hur
- College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Byung-Yong Park
- College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Jaeyoung Heo
- International Agricultural Development and Cooperation Center, Chonbuk National University, Jeonju, Republic of Korea
- * E-mail:
| |
Collapse
|
46
|
Zhang S, Huo X, Zhang Y, Lu X, Xu C, Xu X. The association of PM 2.5 with airway innate antimicrobial activities of salivary agglutinin and surfactant protein D. CHEMOSPHERE 2019; 226:915-923. [PMID: 31509921 DOI: 10.1016/j.chemosphere.2019.04.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 02/05/2023]
Abstract
Fine particulate matter ≤2.5 μm (PM2.5) is a prominent global public health risk factor that can cause respiratory infection by downregulating the amounts of antimicrobial proteins and peptides (AMPs). Both salivary agglutinin (SAG) and surfactant protein D (SPD) are important AMPs in respiratory mucosal fluid, providing protection against airway pathogen invasion and infection by inducing microbial aggregation and enhancing pathogen clearance. However, the relationship between PM2.5 and these AMPs is unclear. To better understand the relationship between PM2.5 and airway innate immune defenses, we review the respiratory antimicrobial activities of SAG and SPD, as well as the adverse effects of PM2.5 on airway innate antimicrobial defense. We speculate there exists a dual effect between PM2.5 and respiratory antimicrobial activity, which means that PM2.5 suppresses respiratory antimicrobial activity through downregulating airway AMPs, while airway AMPs accelerate PM2.5 clearance by inducing PM2.5 microbial aggregation. We propose further research on the relationship between PM2.5 and these AMPs.
Collapse
Affiliation(s)
- Shaocheng Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511486, Guangdong, China
| | - Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xueling Lu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Cheng Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
47
|
The Group B Streptococcal surface antigen I/II protein, BspC, interacts with host vimentin to promote adherence to brain endothelium and inflammation during the pathogenesis of meningitis. PLoS Pathog 2019; 15:e1007848. [PMID: 31181121 PMCID: PMC6586375 DOI: 10.1371/journal.ppat.1007848] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/20/2019] [Accepted: 05/16/2019] [Indexed: 12/29/2022] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) normally colonizes healthy adults but can cause invasive disease, such as meningitis, in the newborn. To gain access to the central nervous system, GBS must interact with and penetrate brain or meningeal blood vessels; however, the exact mechanisms are still being elucidated. Here, we investigate the contribution of BspC, an antigen I/II family adhesin, to the pathogenesis of GBS meningitis. Disruption of the bspC gene reduced GBS adherence to human cerebral microvascular endothelial cells (hCMEC), while heterologous expression of BspC in non-adherent Lactococcus lactis conferred bacterial attachment. In a murine model of hematogenous meningitis, mice infected with ΔbspC mutants exhibited lower mortality as well as decreased brain bacterial counts and inflammatory infiltrate compared to mice infected with WT GBS strains. Further, BspC was both necessary and sufficient to induce neutrophil chemokine expression. We determined that BspC interacts with the host cytoskeleton component vimentin and confirmed this interaction using a bacterial two-hybrid assay, microscale thermophoresis, immunofluorescent staining, and imaging flow cytometry. Vimentin null mice were protected from WT GBS infection and also exhibited less inflammatory cytokine production in brain tissue. These results suggest that BspC and the vimentin interaction is critical for the pathogenesis of GBS meningitis. Group B Streptococcus (GBS) typically colonizes healthy adults but can cause severe disease in immune-compromised individuals, including newborns. Despite wide-spread intrapartum antibiotic prophylaxis given to pregnant women, GBS remains a leading cause of neonatal meningitis. To cause meningitis, GBS must interact with and penetrate the blood-brain barrier (BBB), which separates bacteria and immune cells in the blood from the brain. In order to develop targeted therapies to treat GBS meningitis, it is important to understand the mechanisms of BBB crossing. Here, we describe the role of the GBS surface factor, BspC, in promoting meningitis and discover the host ligand for BspC, vimentin, which is an intermediate filament protein that is constitutively expressed by endothelial cells. We determined that BspC interacts with the C-terminal domain of cell-surface vimentin to promote bacterial attachment to brain endothelial cells and that purified BspC protein can induce immune signaling pathways. In a mouse model of hematogenous meningitis, we observed that a GBS mutant lacking BspC was less virulent compared to WT GBS and resulted in less inflammatory disease. We also observed that mice lacking vimentin were protected from GBS infection. These results reveal the importance of the BspC-vimentin interaction in the progression of GBS meningitis disease.
Collapse
|
48
|
Joshi R, Yan D, Hamed O, Mostafa MM, Joshi T, Newton R, Giembycz MA. Impact of Phosphodiesterase 4 Inhibition on the Operational Efficacy, Response Maxima, and Kinetics of Indacaterol-Induced Gene Expression Changes in BEAS-2B Airway Epithelial Cells: A Global Transcriptomic Analysis. Mol Pharmacol 2019; 96:56-72. [DOI: 10.1124/mol.118.115311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/25/2019] [Indexed: 12/26/2022] Open
|
49
|
Zhang S, Huo X, Zhang Y, Huang Y, Zheng X, Xu X. Ambient fine particulate matter inhibits innate airway antimicrobial activity in preschool children in e-waste areas. ENVIRONMENT INTERNATIONAL 2019; 123:535-542. [PMID: 30622078 DOI: 10.1016/j.envint.2018.12.061] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 12/12/2018] [Accepted: 12/28/2018] [Indexed: 02/05/2023]
Abstract
Ambient fine particulate matter (PM2.5) is a risk factor for respiratory diseases. Previous studies suggest that PM2.5 exposure may down-regulate airway antimicrobial proteins and peptides (AMPs), thereby accelerating airway pathogen infection. However, epidemiological research is scarce. Hence, we estimated the associations between individual PM2.5 chronic daily intake (CDI) and the levels of the airway AMP salivary agglutinin (SAG), as well as peripheral leukocyte counts and pro-inflammatory cytokines, of preschool children in Guiyu (an e-waste area) and Haojiang (a reference area located 31.6 km to the east of Guiyu). We recruited 581 preschool children from Guiyu and Haojiang, of which 222 were included in this study for a matching design (Guiyu: n = 110 vs. Haojiang: n = 112). Air PM2.5 pollution data was collected to calculate individual PM2.5 CDI. The mean concentration of PM2.5 in Guiyu was higher than in Haojiang, resulting in a higher individual PM2.5 CDI. Concomitantly, saliva SAG levels were lower in Guiyu children (5.05 ng/mL) than in Haojiang children (8.68 ng/mL), and were negatively correlated with CDI. Additionally, peripheral counts of white blood cells, and the concentrations of interleukin-8 and tumor necrosis factor-alpha, in Guiyu children were greater than in Haojiang children, and were positively associated with CDI. Similar results were found for neutrophils and monocytes. To our knowledge, this is the first study on the relationship between PM2.5 exposure and innate airway antimicrobial activity in children, in an e-waste area, showing that PM2.5 pollution may weaken airway antimicrobial activity by down-regulation of saliva SAG levels, which might accelerate airway pathogen infection in children.
Collapse
Affiliation(s)
- Shaocheng Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 511486, Guangdong, China
| | - Yu Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9713, GZ, the Netherlands
| | - Yu Huang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xiangbin Zheng
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
50
|
Nelson CS, Fouda GG, Permar SR. Pediatric HIV-1 Acquisition and Lifelong Consequences of Infant Infection. CURRENT IMMUNOLOGY REVIEWS 2019; 15:131-138. [PMID: 33223981 PMCID: PMC7678020 DOI: 10.2174/1573395514666180531074047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/11/2018] [Accepted: 05/24/2018] [Indexed: 11/22/2022]
Abstract
Increased availability of antiretroviral therapy to pregnant and breastfeeding women in resource-limited areas has proven remarkably successful at reducing HIV vertical transmission rates over the past several decades. Yet, still more than 170,000 children are infected annually due to failures in therapy implementation, monitoring, and adherence. Mother-to-child transmission (MTCT) of HIV-1 can occur at one of several distinct stages of infant development - intrauterine, intrapartum, and postpartum. The heterogeneity of the maternal-fetal interface at each of these modes of transmission poses a challenge for the implementation of immune interventions to prevent all modes of HIV MTCT. However, using mother-infant human cohorts and nonhuman primate models of infant simian immunodeficiency virus (SIV) acquisition, investigators have made important observation about the biology of pediatric HIV infection and have identified unique protective immune factors for each mode of transmission. Knowledge of immune factors protective against HIV MTCT will be critical to the development of targeted immune therapies to prevent infant HIV acquisition and to bring an end to the pediatric AIDS epidemic.
Collapse
Affiliation(s)
- Cody S. Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Genevieve G.A. Fouda
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Sallie R. Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|