1
|
Ji Y, Sun C, Wu S. Transcriptomic and Biochemical Analysis of the Antimicrobial Mechanism of Lipopeptide Iturin W against Staphylococcus aureus. Int J Mol Sci 2024; 25:9949. [PMID: 39337437 PMCID: PMC11432370 DOI: 10.3390/ijms25189949] [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: 08/20/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Staphylococcus aureus is one of the most serious pathogens threatening food safety and public health. We have previously showed that iturin W exhibited obvious antifungal activity on plant pathogens. In the present study, we found iturin W, especially C14 iturin W, showed strong antimicrobial activity against S. aureus, and the antimicrobial mechanism of C14 iturin W was further investigated by transcriptomic analysis and a related biochemical experiment. The results showed that C14 iturin W can reduce the expression levels of genes associated with the reactive oxygen species (ROS) scavenging enzyme and genes involved in arginine biosynthesis, thus leading to the increase in ROS levels of S. aureus. Furthermore, C14 iturin W can also interfere with proton dynamics, which is crucial for cells to regulate various biological possesses. Therefore, ROS accumulation and change in proton motive force are import ways for C14 iturin W to exert the antimicrobial activity. In addition, C14 iturin W can also reduce the expression levels of genes related to virulence factors and decrease the production of enterotoxins and hemolysins in S. aureus, indicating that C14 iturin W has a good potential in food and pharmaceutical fields to reduce the harm caused by S. aureus in the future.
Collapse
Affiliation(s)
- Yingyu Ji
- College of Life Sciences, Qingdao University, Qingdao 266071, China;
| | - Chaomin Sun
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| | - Shimei Wu
- College of Life Sciences, Qingdao University, Qingdao 266071, China;
| |
Collapse
|
2
|
Lee J, Jo J, Wan J, Seo H, Han SW, Shin YJ, Kim DH. In Vitro Evaluation of Probiotic Properties and Anti-Pathogenic Effects of Lactobacillus and Bifidobacterium Strains as Potential Probiotics. Foods 2024; 13:2301. [PMID: 39063385 PMCID: PMC11276478 DOI: 10.3390/foods13142301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Probiotics restore gut microbial balance, thereby providing health-promoting effects to the host. They have long been suggested for managing intestinal disorders caused by pathogens and for improving gut health. This study evaluated the probiotic properties and anti-pathogenic effects of specific probiotic strains against the intestinal pathogens Staphylococcus aureus and Escherichia coli. The tested strains-Lactiplantibacillus plantarum LC27, Limosilactobacillus reuteri NK33, Lacticaseibacillus rhamnosus NK210, Bifidobacterium longum NK46, and Bifidobacterium bifidum NK175-were able to survive harsh conditions simulating gastric and intestinal fluids. These strains exhibited good auto-aggregation abilities (41.8-92.3%) and ideal hydrophobicity (30.9-85.6% and 38.3-96.1% for xylene and chloroform, respectively), along with the ability to co-aggregate with S. aureus (40.6-68.2%) and E. coli (38.6-75.2%), indicating significant adhesion levels to Caco-2 cells. Furthermore, these strains' cell-free supernatants (CFSs) demonstrated antimicrobial and antibiofilm activity against S. aureus and E. coli. Additionally, these strains inhibited gas production by E. coli through fermentative activity. These findings suggest that the strains tested in this study have potential as novel probiotics to enhance gut health.
Collapse
Affiliation(s)
- Jaekoo Lee
- PB Business Department, NVP Healthcare Inc., Suwon 16209, Republic of Korea; (J.L.); (J.J.); (J.W.); (H.S.); (S.-W.H.)
- Department of Food Regulatory Science, Korea University, Sejong 30019, Republic of Korea
| | - Jaehyun Jo
- PB Business Department, NVP Healthcare Inc., Suwon 16209, Republic of Korea; (J.L.); (J.J.); (J.W.); (H.S.); (S.-W.H.)
| | - Jungho Wan
- PB Business Department, NVP Healthcare Inc., Suwon 16209, Republic of Korea; (J.L.); (J.J.); (J.W.); (H.S.); (S.-W.H.)
| | - Hanseul Seo
- PB Business Department, NVP Healthcare Inc., Suwon 16209, Republic of Korea; (J.L.); (J.J.); (J.W.); (H.S.); (S.-W.H.)
| | - Seung-Won Han
- PB Business Department, NVP Healthcare Inc., Suwon 16209, Republic of Korea; (J.L.); (J.J.); (J.W.); (H.S.); (S.-W.H.)
| | - Yoon-Jung Shin
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| |
Collapse
|
3
|
Wang X, Zhang T, Li W, Zhang M, Zhao L, Wang N, Zhang X, Zhang B. Dietary supplementation with Macleaya cordata extract alleviates intestinal injury in broiler chickens challenged with lipopolysaccharide by regulating gut microbiota and plasma metabolites. Front Immunol 2024; 15:1414869. [PMID: 39100674 PMCID: PMC11294198 DOI: 10.3389/fimmu.2024.1414869] [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: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction The prevention and mitigation of intestinal immune challenge is crucial for poultry production. This study investigated the effects of dietary Macleaya cordata extract (MCE) supplementation on the prevention of intestinal injury in broiler chickens challenged with lipopolysaccharide (LPS). Methods A total of 256 one-day-old male Arbor Acres broilers were randomly divided into 4 treatment groups using a 2×2 factorial design with 2 MCE supplemental levels (0 and 400 mg/kg) and 2 LPS challenge levels (0 and 1 mg/kg body weight). The experiment lasted for 21 d. Results and discussion The results showed that MCE supplementation increased the average daily feed intake during days 0-14. MCE supplementation and LPS challenge have an interaction on the average daily gain during days 15-21. MCE supplementation significantly alleviated the decreased average daily gain of broiler chickens induced by LPS. MCE supplementation increased the total antioxidant capacity and the activity of catalase and reduced the level of malondialdehyde in jejunal mucosa. MCE addition elevated the villus height and the ratio of villus height to crypt depth of the ileum. MCE supplementation decreased the mRNA expression of pro-inflammatory cytokines interleukin (IL)-6 and IL-8 in the jejunum. MCE addition mitigated LPS-induced mRNA up-expression of pro-inflammatory factors IL-1β and IL-17 in the jejunum. MCE supplementation increased the abundance of probiotic bacteria (such as Lactobacillus and Blautia) and reduced the abundance of pathogenic bacteria (such as Actinobacteriota, Peptostretococcaceae, and Rhodococcus), leading to alterations in gut microbiota composition. MCE addition altered several metabolic pathways such as Amino acid metabolism, Nucleotide metabolism, Energy metabolism, Carbohydrate metabolism, and Lipid metabolism in broilers. In these pathways, MCE supplementation increased the levels of L-aspartic acid, L-Glutamate, L-serine, etc., and reduced the levels of phosphatidylcholine, phosphatidylethanolamine, thromboxane B2, 13-(S)-HODPE, etc. In conclusion, dietary supplementation of 400 mg/kg MCE effectively improved the growth performance and intestinal function in LPS-challenged broiler chickens, probably due to the modulation of gut microbiota and plasma metabolites.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Beibei Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
4
|
Petronio Petronio G, Di Naro M, Venditti N, Guarnieri A, Cutuli MA, Magnifico I, Medoro A, Foderà E, Passarella D, Nicolosi D, Di Marco R. Targeting S. aureus Extracellular Vesicles: A New Putative Strategy to Counteract Their Pathogenic Potential. Pharmaceutics 2024; 16:789. [PMID: 38931910 PMCID: PMC11207539 DOI: 10.3390/pharmaceutics16060789] [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: 04/24/2024] [Revised: 05/27/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long-term inflammatory skin disease atopic dermatitis is characterized by dry skin, itching, and eczematous lesions. During inflammation skin barrier protein impairment promotes S. aureus colonisation in the inflamed skin, worsening AD patient's clinical condition. Proteomic analysis revealed the presence of several immune evasion proteins and virulence factors in S. aureus extracellular vesicles (EVs), suggesting a possible role for these proteins in the pathophysiology of atopic dermatitis. The objective of this study is to assess the efficacy of a wall fragment obtained from a patented strain of C. acnes DSM28251 (c40) and its combination with a mucopolysaccharide carrier (HAc40) in counteract the pathogenic potential of EVs produced by S. aureus ATCC 14458. Results obtained from in vitro studies on HaCaT keratinocyte cells showed that HAc40 and c40 treatment significantly altered the size and pathogenicity of S. aureus EVs. Specifically, EVs grew larger, potentially reducing their ability to interact with the target cells and decreasing cytotoxicity. Additionally, the overexpression of the tight junctions mRNA zona occludens 1 (ZO1) and claudin 1 (CLDN1) following EVs exposure was decreased by HAc40 and c40 treatment, indicating a protective effect on the epidermal barrier's function. These findings demonstrate how Hac40 and c40 may mitigate the harmful effects of S. aureus EVs. Further investigation is needed to elucidate the exact mechanisms underlying this interaction and explore the potential clinical utility of c40 and its mucopolysaccharide carrier conjugate HAc40 in managing atopic dermatitis.
Collapse
Affiliation(s)
- Giulio Petronio Petronio
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Maria Di Naro
- Department of Drug and Health Sciences, Università degli Studi di Catania, 95125 Catania, Italy
| | - Noemi Venditti
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
- UO Laboratorio Analisi, Responsible Research Hospital, 86100 Campobasso, Italy
| | - Antonio Guarnieri
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | | | | | - Alessandro Medoro
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Emanuele Foderà
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Daniela Passarella
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Daria Nicolosi
- Department of Drug and Health Sciences, Università degli Studi di Catania, 95125 Catania, Italy
| | - Roberto Di Marco
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| |
Collapse
|
5
|
So YJ, Park OJ, Kwon Y, Im J, Lee D, Yun SH, Cho K, Yun CH, Han SH. Bacillus subtilis Induces Human Beta Defensin-2 Through its Lipoproteins in Human Intestinal Epithelial Cells. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10224-4. [PMID: 38376819 DOI: 10.1007/s12602-024-10224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/21/2024]
Abstract
Human intestinal epithelial cells (IECs) play an important role in maintaining gut homeostasis by producing antimicrobial peptides (AMPs). Bacillus subtilis, a commensal bacterium, is considered a probiotic. Although its protective effects on intestinal health are widely reported, the key component of B. subtilis responsible for its beneficial effects remains elusive. In this study, we tried to identify the key molecules responsible for B. subtilis-induced AMPs and their molecular mechanisms in a human IEC line, Caco-2. B. subtilis increased human beta defensin (HBD)-2 mRNA expression in a dose- and time-dependent manner. Among the B. subtilis microbe-associated molecular patterns, lipoprotein (LPP) substantially increased the mRNA expression and protein production of HBD-2, whereas lipoteichoic acid and peptidoglycan did not show such effects. Those results were confirmed in primary human IECs. In addition, both LPP recognition and HBD-2 secretion mainly took place on the apical side of fully differentiated and polarized Caco-2 cells through Toll-like receptor 2-mediated JNK/p38 MAP kinase/AP-1 and NF-κB pathways. HBD-2 efficiently inhibited the growth of the intestinal pathogens Staphylococcus aureus and Bacillus cereus. Furthermore, LPPs pre-incubated with lipase or proteinase K decreased LPP-induced HBD-2 expression, suggesting that the lipid and protein moieties of LPP are crucial for HBD-2 expression. Q Exactive Plus mass spectrometry identified 35 B. subtilis LPP candidates within the LPP preparation, and most of them were ABC transporters. Taken together, these results suggest that B. subtilis promotes HBD-2 secretion in human IECs mainly with its LPPs, which might enhance the protection from intestinal pathogens.
Collapse
Affiliation(s)
- Yoon Ju So
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeongkag Kwon
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung-Ho Yun
- Center for Research Equipment, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Kun Cho
- Center for Research Equipment, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institutes of Green Bio Science & Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
6
|
Yoon KN, Lee SJ, Keum GB, Song KY, Park JH, Song BS, Yu SY, Cho JH, Kim ES, Doo H, Kwak J, Kim S, Eun JB, Lee JH, Kim HB, Lee JH, Kim JK. Characteristics of Lactococcus petauri GB97 lysate isolated from porcine feces and its in vitro and in vivo effects on inflammation, intestinal barrier function, and gut microbiota composition in mice. Microbiol Spectr 2024; 12:e0133423. [PMID: 38019021 PMCID: PMC10782967 DOI: 10.1128/spectrum.01334-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/06/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE Weaning is a crucial step in piglet management to improve pork production. During the weaning phase, disruption of epithelial barrier function and intestinal inflammation can lead to decreased absorption of nutrients and diarrhea. Therefore, maintaining a healthy intestine, epithelial barrier function, and gut microbiota composition in this crucial phase is strategic for optimal weaning in pigs. We isolated a lysate of Lactococcus petauri GB97 (LPL97) from healthy porcine feces and evaluated its anti-inflammatory activities, barrier integrity, and gut microbial changes in LPS-induced murine macrophages and DSS-induced colitis mice. We found that LPL97 regulated the immune response by downregulating the TLR4/NF-κB/MAPK signaling pathway both in vitro and in vivo. Furthermore, LPL97 alleviated the disruption of intestinal epithelial integrity and gut microbiota dysbiosis in colitis mice. This study indicates that LPL97 has the potential to be developed as an alternative feed additive to antibiotics for the swine industry.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, South Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, South Korea
| | - Soo-Jeong Lee
- Department of Food and Animal Biotechnology, Seoul National University, Seoul, South Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
| | - Gi Beom Keum
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Ki-Young Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, South Korea
| | - Jong-Heum Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, South Korea
| | - Beom-Seok Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, South Korea
| | - Seung Yeob Yu
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Jae Hyoung Cho
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Eun Sol Kim
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Hyunok Doo
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Jinok Kwak
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Sheena Kim
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, South Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Ju-Hoon Lee
- Department of Food and Animal Biotechnology, Seoul National University, Seoul, South Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, South Korea
| |
Collapse
|
7
|
Goc A, Sumera W, Rath M, Niedzwiecki A. Inhibition of α-hemolysin activity of Staphylococcus aureus by theaflavin 3,3'-digallate. PLoS One 2023; 18:e0290904. [PMID: 37651426 PMCID: PMC10470925 DOI: 10.1371/journal.pone.0290904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023] Open
Abstract
The ongoing rise in antibiotic resistance, and a waning of the introduction of new antibiotics, has resulted in limited treatment options for bacterial infections, including these caused by methicillin-resistant Staphylococcus aureus, leaving the world in a post-antibiotic era. Here, we set out to examine mechanisms by which theaflavin 3,3'-digallate (TF3) might act as an anti-hemolytic compound. In the presented study, we found that TF3 has weak bacteriostatic and bactericidal effects on Staphylococcus aureus, and strong inhibitory effect towards the hemolytic activity of its α-hemolysin (Hla) including its production and secretion. A supportive SPR assay reinforced these results and further revealed binding of TF3 to Hla with KD = 4.57×10-5 M. Interestingly, TF3 was also able to protect human primary keratinocytes from Hla-induced cell death, being at the same time non-toxic for them. Further analysis of TF3 properties revealed that TF3 blocked Hla-prompting immune reaction by inhibiting production and secretion of IL1β, IL6, and TNFα in vitro and in vivo, through affecting NFκB activity. Additionally, we observed that TF3 also markedly attenuated S. aureus-induced barrier disruption, by inhibiting Hla-triggered E-cadherin and ZO-1 impairment. Overall, by blocking activity of Hla, TF3 subsequently subdued the inflammation and protected the epithelial barrier, which is considered as beneficial to relieving skin injury.
Collapse
Affiliation(s)
- Anna Goc
- Department of Infectious Diseases, Dr. Rath Research Institute, San Jose, California, United States of America
| | - Waldemar Sumera
- Department of Infectious Diseases, Dr. Rath Research Institute, San Jose, California, United States of America
| | - Matthias Rath
- Department of Infectious Diseases, Dr. Rath Research Institute, San Jose, California, United States of America
| | - Aleksandra Niedzwiecki
- Department of Infectious Diseases, Dr. Rath Research Institute, San Jose, California, United States of America
| |
Collapse
|
8
|
Celebi O, Taghizadehghalehjoughi A, Celebi D, Mesnage R, Golokhvast KS, Arsene AL, Spandidos DA, Tsatsakis A. Effect of the combination of Lactobacillus acidophilus (probiotic) with vitamin K3 and vitamin E on Escherichia coli and Staphylococcus aureus: An in vitro pathogen model. Mol Med Rep 2023; 27:119. [PMID: 37144488 PMCID: PMC10196883 DOI: 10.3892/mmr.2023.13006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 05/06/2023] Open
Abstract
The gut microbiota plays a key role in maintaining health and regulating the host's immune response. The use of probiotics and concomitant vitamins can increase mucus secretion by improving the intestinal microbial population and prevent the breakdown of tight junction proteins by reducing lipopolysaccharide concentration. Changes in the intestinal microbiome mass affect multiple metabolic and physiological functions. Studies on how this microbiome mass and the regulation in the gastrointestinal tract are affected by probiotic supplements and vitamin combinations have attracted attention. The current study evaluated vitamins K and E and probiotic combinations effects on Escherichia coli and Staphylococcus aureus. Minimal inhibition concentrations of vitamins and probiotics were determined. In addition, inhibition zone diameters, antioxidant activities and immunohistochemical evaluation of the cell for DNA damage were performed to evaluate the effects of vitamins and probiotics. At the specified dose intervals, L. acidophilus and vitamin combinations inhibit the growth of Escherichia coli and Staphylococcus aureus. It could thus contribute positively to biological functions by exerting immune system‑strengthening activities.
Collapse
Affiliation(s)
- Ozgur Celebi
- Department of Medical Microbiology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | | | - Demet Celebi
- Department of Microbiology, Faculty of Veterinary Medicine, Ataturk University, 25240 Erzurum, Turkey
- Vaccine Application and Development Center, Ataturk University, 25240 Erzurum, Turkey
| | - Robin Mesnage
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London WC2R 2LS, United Kingdom
| | | | - Andreea Letitia Arsene
- Department of General and Pharmaceutical Microbiology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
9
|
Majumder S, Sackey T, Viau C, Park S, Xia J, Ronholm J, George S. Genomic and phenotypic profiling of Staphylococcus aureus isolates from bovine mastitis for antibiotic resistance and intestinal infectivity. BMC Microbiol 2023; 23:43. [PMID: 36803552 PMCID: PMC9940407 DOI: 10.1186/s12866-023-02785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is one of the prevalent etiological agents of contagious bovine mastitis, causing a significant economic burden on the global dairy industry. Given the emergence of antibiotic resistance (ABR) and possible zoonotic spillovers, S aureus from mastitic cattle pose threat to both veterinary and public health. Therefore, assessment of their ABR status and pathogenic translation in human infection models is crucial. RESULTS In this study, 43 S. aureus isolates associated with bovine mastitis obtained from four different Canadian provinces (Alberta, Ontario, Quebec, and Atlantic provinces) were tested for ABR and virulence through phenotypic and genotypic profiling. All 43 isolates exhibited crucial virulence characteristics such as hemolysis, and biofilm formation, and six isolates from ST151, ST352, and ST8 categories showed ABR. Genes associated with ABR (tetK, tetM, aac6', norA, norB, lmrS, blaR, blaZ, etc.), toxin production (hla, hlab, lukD, etc.), adherence (fmbA, fnbB, clfA, clfB, icaABCD, etc.), and host immune invasion (spa, sbi, cap, adsA, etc.) were identified by analyzing whole-genome sequences. Although none of the isolates possessed human adaptation genes, both groups of ABR and antibiotic-susceptible isolates demonstrated intracellular invasion, colonization, infection, and death of human intestinal epithelial cells (Caco-2), and Caenorhabditis elegans. Notably, the susceptibilities of S. aureus towards antibiotics such as streptomycin, kanamycin, and ampicillin were altered when the bacteria were internalized in Caco-2 cells and C. elegans. Meanwhile, tetracycline, chloramphenicol, and ceftiofur were comparatively more effective with ≤ 2.5 log10 reductions of intracellular S. aureus. CONCLUSIONS This study demonstrated the potential of S. aureus isolated from mastitis cows to possess virulence characteristics enabling invasion of intestinal cells thus calling for developing therapeutics capable of targeting drug-resistant intracellular pathogens for effective disease management.
Collapse
Affiliation(s)
- Satwik Majumder
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Trisha Sackey
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Charles Viau
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Soyoun Park
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Jianguo Xia
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada ,grid.14709.3b0000 0004 1936 8649Department of Animal Science, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Jennifer Ronholm
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada ,grid.14709.3b0000 0004 1936 8649Department of Animal Science, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Saji George
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9, Canada.
| |
Collapse
|
10
|
Complexification of In Vitro Models of Intestinal Barriers, A True Challenge for a More Accurate Alternative Approach. Int J Mol Sci 2023; 24:ijms24043595. [PMID: 36835003 PMCID: PMC9958734 DOI: 10.3390/ijms24043595] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
The use of cell models is common to mimic cellular and molecular events in interaction with their environment. In the case of the gut, the existing models are of particular interest to evaluate food, toxicants, or drug effects on the mucosa. To have the most accurate model, cell diversity and the complexity of the interactions must be considered. Existing models range from single-cell cultures of absorptive cells to more complex combinations of two or more cell types. This work describes the existing solutions and the challenges that remain to be solved.
Collapse
|
11
|
Wan Y, Wang X, Bai T, Zheng X, Yang L, Li Q, Wang X. Lysine Inhibits Hemolytic Activity of Staphylococcus aureus and Its Application in Food Model Contaminated with Staphylococcus aureus. Toxins (Basel) 2022; 14:toxins14120867. [PMID: 36548764 PMCID: PMC9786064 DOI: 10.3390/toxins14120867] [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: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022] Open
Abstract
Alpha-hemolysin (Hla) is one of the important exotoxins of Staphylococcus aureus (S. aureus) and can be used as a target to reduce the virulence of S. aureus. This study explored the inhibitory effect of Lysine (Lys) on Hla and its application in food safety. Lys significantly inhibited the expression of Hla at sub-inhibitory concentrations and directly interacted with Hla to interfere with its oligomerization and thus significantly inhibited its hemolytic activity. Notably, Lys attenuated S. aureus damage to mouse small intestine and Caco-2 cells and delayed mouse mortality. In the food model, Lys inhibited the expression of Hla of S. aureus and had no significant effect on the sensory score. Moreover, Lys had no obvious damage effect on the main organs of mice, which indicated that Lys has good biocompatibility and has the potential to be used in the food industry as an anti-S. aureus preparation.
Collapse
|
12
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
13
|
Yamada S, Noda T, Okabe K, Yanagida S, Nishida M, Kanda Y. SARS-CoV-2 induces barrier damage and inflammatory responses in the human iPSC-derived intestinal epithelium. J Pharmacol Sci 2022; 149:139-146. [PMID: 35641026 PMCID: PMC9060709 DOI: 10.1016/j.jphs.2022.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 01/25/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly spread and led to global health crises. COVID-19 causes well-known respiratory failure and gastrointestinal symptoms, such as diarrhea, nausea, and vomiting. Thus, human gastrointestinal cell models are urgently needed for COVID-19 research; however, it is difficult to obtain primary human intestinal cells. In this study, we examined whether human induced pluripotent stem cell (iPSC)-derived small intestinal epithelial cells (iPSC-SIECs) could be used as a SARS-CoV-2 infection model. We observed that iPSC-SIECs, such as absorptive and Paneth cells, were infected with SARS-CoV-2, and remdesivir treatment decreased intracellular SARS-CoV-2 replication in iPSC-SIECs. SARS-CoV-2 infection decreased expression levels of tight junction markers, ZO-3 and CLDN1, and transepithelial electrical resistance (TEER), which evaluates the integrity of tight junction dynamics. In addition, SARS-CoV-2 infection increased expression levels of proinflammatory genes, which are elevated in patients with COVID-19. These findings suggest iPSC-SIECs as a useful in vitro model for elucidating COVID-19 pathology and drug development.
Collapse
Affiliation(s)
- Shigeru Yamada
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Takamasa Noda
- Department of Psychiatry, National Center of Neurology and Psychiatry, Tokyo, Japan,Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan,Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan,Department of Brain Bioregulatory Science, The Jikei University Graduate School of Medicine, Tokyo, Japan
| | - Kaori Okabe
- Department of Psychiatry, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Motohiro Nishida
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan,National Institute for Physiological Sciences and Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan,Corresponding author. Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, 210-9501, Japan. Fax: +81 44 270 1065
| |
Collapse
|
14
|
Bacterial Toxins from Staphylococcus aureus and Bordetella bronchiseptica Predispose the Horse's Respiratory Tract to Equine Herpesvirus Type 1 Infection. Viruses 2022; 14:v14010149. [PMID: 35062352 PMCID: PMC8778808 DOI: 10.3390/v14010149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Respiratory disease in horses is caused by a multifactorial complex of infectious agents and environmental factors. An important pathogen in horses is equine herpesvirus type 1 (EHV-1). During co-evolution with this ancient alphaherpesvirus, the horse’s respiratory tract has developed multiple antiviral barriers. However, these barriers can become compromised by environmental threats. Pollens and mycotoxins enhance mucosal susceptibility to EHV-1 by interrupting cell junctions, allowing the virus to reach its basolateral receptor. Whether bacterial toxins also play a role in this impairment has not been studied yet. Here, we evaluated the role of α-hemolysin (Hla) and adenylate cyclase (ACT), toxins derived from the facultative pathogenic bacterium Staphylococcus aureus (S. aureus) and the primary pathogen Bordetella bronchiseptica (B. bronchiseptica), respectively. Equine respiratory mucosal explants were cultured at an air–liquid interface and pretreated with these toxins, prior to EHV-1 inoculation. Morphological analysis of hematoxylin–eosin (HE)-stained sections of the explants revealed a decreased epithelial thickness upon treatment with both toxins. Additionally, the Hla toxin induced detachment of epithelial cells and a partial loss of cilia. These morphological changes were correlated with increased EHV-1 replication in the epithelium, as assessed by immunofluorescent stainings and confocal microscopy. In view of these results, we argue that the ACT and Hla toxins increase the susceptibility of the epithelium to EHV-1 by disrupting the epithelial barrier function. In conclusion, this study is the first to report that bacterial exotoxins increase the horse’s sensitivity to EHV-1 infection. Therefore, we propose that horses suffering from infection by S. aureus or B. bronchiseptica may be more susceptible to EHV-1 infection.
Collapse
|
15
|
Gao Y, Xu Y, Yin J. Selenomethionine Ameliorates Cognitive Impairment, Decreases Hippocampal Oxidative Stress and Attenuates Dysbiosis in D-Galactose-Treated Mice. Antioxidants (Basel) 2022; 11:antiox11010111. [PMID: 35052615 PMCID: PMC8772940 DOI: 10.3390/antiox11010111] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/25/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
The prevalence of age-related cognitive impairment is increasing as the proportion of older individuals in the population grows. It is therefore necessary and urgent to find agents to prevent or ameliorate age-related cognitive impairment. Selenomethionine (SeMet) is a natural amino acid occurring in yeast and Brazil nuts. It mitigates cognitive impairment in an Alzheimer’s disease mouse model, however, whether it works on age-related cognitive impairment remains unknown. In this study, SeMet significantly improved the performance of D-galactose-treated mice in the novel object recognition test, passive avoidance task and Morris water maze test. SeMet reversed D-galactose-induced reduction of hippocampal acetylcholine levels, suppression of choline acetyltransferase activity and activation of acetyl cholinesterase. It decreased D-galactose-induced oxidative stress and increased the selenoprotein P levels in the hippocampus. Besides, it attenuated D-galactose-induced dysbiosis by increasing the α-diversity and modulating the taxonomic structure. Correlations between certain taxa and physiological parameters were observed. Our results provide evidence of the effectiveness of SeMet on ameliorating D-galactose-induced cognitive impairment and suggest SeMet has potential to be used in the prevention or adjuvant treatment of age-related cognitive impairment.
Collapse
Affiliation(s)
| | - Yongquan Xu
- Correspondence: (Y.X.); (J.Y.); Tel.: +86-571-8665-0594 (Y.X.); +86-571-8665-0031 (J.Y.)
| | - Junfeng Yin
- Correspondence: (Y.X.); (J.Y.); Tel.: +86-571-8665-0594 (Y.X.); +86-571-8665-0031 (J.Y.)
| |
Collapse
|
16
|
Endothelial glycocalyx degradation during sepsis: Causes and consequences. Matrix Biol Plus 2021; 12:100094. [PMID: 34917925 PMCID: PMC8668992 DOI: 10.1016/j.mbplus.2021.100094] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022] Open
Abstract
The endothelial glycocalyx is a ubiquitous intravascular structure essential for vascular homeostasis. During sepsis, the glycocalyx is degraded via the collective action of a variety of redundant sheddases, the regulation of which remains the focus of active investigation. Septic loss of the glycocalyx imparts both local vascular injury (leading to acute respiratory distress syndrome and acute kidney injury) as well as the systemic consequences of circulating glycosaminoglycan fragments (leading to cognitive dysfunction). Glycocalyx degradation during sepsis is potentially shaped by clinically-modifiable factors, suggesting opportunities for therapeutic intervention to mitigate the end-organ consequences of sepsis.
The glycocalyx is a ubiquitous structure found on endothelial cells that extends into the vascular lumen. It is enriched in proteoglycans, which are proteins attached to the glycosaminoglycans heparan sulfate, chondroitin sulfate, dermatan sulfate, keratan sulfate, and hyaluronic acid. In health and disease, the endothelial glycocalyx is a central regulator of vascular permeability, inflammation, coagulation, and circulatory tonicity. During sepsis, a life-threatening syndrome seen commonly in hospitalized patients, the endothelial glycocalyx is degraded, significantly contributing to its many clinical manifestations. In this review we discuss the intrinsically linked mechanisms responsible for septic endothelial glycocalyx destruction: glycosaminoglycan degradation and proteoglycan cleavage. We then examine the consequences of local endothelial glycocalyx loss to several organ systems and the systemic consequences of shed glycocalyx constituents. Last, we explore clinically relevant non-modifiable and modifiable factors that exacerbate or protect against endothelial glycocalyx shedding during sepsis.
Collapse
Key Words
- ADAM, A Disintegrin and Metalloproteinase
- ANP, Atrial Natriuretic Peptide
- ARDS, Acute respiratory distress syndrome
- Ang2, Angiopoietin-2
- DAMP, Damage-associated Molecular Pattern
- Endothelial glycocalyx
- FFP, Fresh Frozen Plasma
- GAG, Glycosaminoglycan
- Glycosaminoglycans
- HPSE-1/2, Heparanase-1/2
- LPS, Lipopolysaccharide
- MMP, Matrix Metalloproteinase
- PG, Proteoglycan
- Proteoglycans
- Sepsis
- TIMP, Tissue inhibitors of matrix metalloproteinase
Collapse
|
17
|
Hoffmann P, Burmester M, Langeheine M, Brehm R, Empl MT, Seeger B, Breves G. Caco-2/HT29-MTX co-cultured cells as a model for studying physiological properties and toxin-induced effects on intestinal cells. PLoS One 2021; 16:e0257824. [PMID: 34618824 PMCID: PMC8496855 DOI: 10.1371/journal.pone.0257824] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/12/2021] [Indexed: 12/28/2022] Open
Abstract
Infectious gastrointestinal diseases are frequently caused by toxins secreted by pathogens which may impair physiological functions of the intestines, for instance by cholera toxin or by heat-labile enterotoxin. To obtain a functional model of the human intestinal epithelium for studying toxin-induced disease mechanisms, differentiated enterocyte-like Caco-2 cells were co-cultured with goblet cell-like HT29-MTX cells. These co-cultures formed a functional epithelial barrier, as characterized by a high electrical resistance and the presence of physiological intestinal properties such as glucose transport and chloride secretion which could be demonstrated electrophysiologically and by measuring protein expression. When the tissues were exposed to cholera toxin or heat-labile enterotoxin in the Ussing chamber, cholera toxin incubation resulted in an increase in short-circuit currents, indicating an increase in apical chloride secretion. This is in line with typical cholera toxin-induced secretory diarrhea in humans, while heat-labile enterotoxin only showed an increase in short-circuit-current in Caco-2 cells. This study characterizes for the first time the simultaneous measurement of physiological properties on a functional and structural level combined with the epithelial responses to bacterial toxins. In conclusion, using this model, physiological responses of the intestine to bacterial toxins can be investigated and characterized. Therefore, this model can serve as an alternative to the use of laboratory animals for characterizing pathophysiological mechanisms of enterotoxins at the intestinal level.
Collapse
Affiliation(s)
- Pascal Hoffmann
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Burmester
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Langeheine
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ralph Brehm
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Michael T. Empl
- Institute for Food Toxicology, University of Veterinary Medicine, Hannover, Germany
| | - Bettina Seeger
- Institute for Food Toxicology, University of Veterinary Medicine, Hannover, Germany
| | - Gerhard Breves
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
18
|
Mergani A, Wanes D, Schecker N, Branitzki-Heinemann K, Naim HY, von Köckritz-Blickwede M. Staphylococcus aureus Infection Influences the Function of Intestinal Cells by Altering the Lipid Raft-Dependent Sorting of Sucrase-Isomaltase. Front Cell Dev Biol 2021; 9:699970. [PMID: 34490251 PMCID: PMC8418112 DOI: 10.3389/fcell.2021.699970] [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: 04/24/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is an important nosocomial and community-acquired facultative intracellular pathogen. Many studies have reported that S. aureus infections are associated with intestinal symptoms, but little is known about the molecular mechanisms implicated in S. aureus-induced alterations of intestinal functions. In this study, we investigated the implication of lipid rafts in the interaction of S. aureus with Caco-2 cells. To assess potential alterations in the lipid raft structure and effects on the hydrolytic function, we utilized sucrase–isomaltase (SI) as the major intestinal α-glucosidase that is associated with and sorted to the apical membrane via lipid rafts. Seven days post-confluent, Caco-2 cells were infected with S. aureus Newman and further incubated for an additional 2 days. After 48 h, the levels of SI expression as well as the enzymatic function of this protein were assessed in the infected versus non-infected cells. Analysis of the sorting behavior of SI to the apical membrane constituted another crucial aspect in studying the effects of S. aureus on Caco-2 cells. For this purpose, the apical membranes or brush border membranes (BBMs; referred to as P2 fraction) were separated in both infected and non-infected cells from the basolateral and intracellular membranes (referred to as P1 fraction) by employing a cationic-based procedure using CaCl2. The data show that there is no significant change in the overall expression levels of SI in the infected versus non-infected cells as assessed by Western blotting analysis using monoclonal anti-SI antibodies. By contrast, a significant decrease in the localization as well as the specific hydrolytic activities of SI toward sucrose and isomaltose (Palatinose) was observed in the BBM (P2 fraction) in Caco-2 cells 48 h post-infection. Concomitantly, the specific SI activities increased in the basolateral membrane/intracellular fraction (P1). Noteworthy, the specific activity of SI in the BBM of infected cells was markedly reduced as compared with that of the non-infected counterparts. The data accumulated from this study strongly suggest that infections with S. aureus influence the final step in the lipid raft-associated trafficking of human SI and thereby may trigger secondary functional gastrointestinal disorders.
Collapse
Affiliation(s)
- AhmedElmontaser Mergani
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hanover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hanover, Germany
| | - Dalanda Wanes
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Natalie Schecker
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Katja Branitzki-Heinemann
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hanover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hanover, Germany
| | - Hassan Y Naim
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hanover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hanover, Germany
| |
Collapse
|
19
|
Krones D, Rühling M, Becker KA, Kunz TC, Sehl C, Paprotka K, Gulbins E, Fraunholz M. Staphylococcus aureus α-Toxin Induces Acid Sphingomyelinase Release From a Human Endothelial Cell Line. Front Microbiol 2021; 12:694489. [PMID: 34394034 PMCID: PMC8358437 DOI: 10.3389/fmicb.2021.694489] [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: 04/13/2021] [Accepted: 07/05/2021] [Indexed: 11/14/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is well known to express a plethora of toxins of which the pore-forming hemolysin A (α-toxin) is the best-studied cytolysin. Pore-forming toxins (PFT) permeabilize host membranes during infection thereby causing concentration-dependent effects in host cell membranes ranging from disordered ion fluxes to cytolysis. Host cells possess defense mechanisms against PFT attack, resulting in endocytosis of the breached membrane area and delivery of repair vesicles to the insulted plasma membrane as well as a concurrent release of membrane repair enzymes. Since PFTs from several pathogens have been shown to recruit membrane repair components, we here investigated whether staphylococcal α-toxin is able to induce these mechanisms in endothelial cells. We show that S. aureus α-toxin induced increase in cytosolic Ca2+ in endothelial cells, which was accompanied by p38 MAPK phosphorylation. Toxin challenge led to increased endocytosis of an extracellular fluid phase marker as well as increased externalization of LAMP1-positive membranes suggesting that peripheral lysosomes are recruited to the insulted plasma membrane. We further observed that thereby the lysosomal protein acid sphingomyelinase (ASM) was released into the cell culture medium. Thus, our results show that staphylococcal α-toxin triggers mechanisms in endothelial cells, which have been implicated in membrane repair after damage of other cell types by different toxins.
Collapse
Affiliation(s)
- David Krones
- Chair of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Marcel Rühling
- Chair of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Katrin Anne Becker
- Institute of Molecular Biology, University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Tobias C Kunz
- Chair of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Carolin Sehl
- Institute of Molecular Biology, University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Kerstin Paprotka
- Chair of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University of Duisburg-Essen, University Hospital, Essen, Germany.,Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Martin Fraunholz
- Chair of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
20
|
Raineri EJM, Altulea D, van Dijl JM. Staphylococcal trafficking and infection - from 'nose to gut' and back. FEMS Microbiol Rev 2021; 46:6321165. [PMID: 34259843 PMCID: PMC8767451 DOI: 10.1093/femsre/fuab041] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/11/2021] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus is an opportunistic human pathogen, which is a leading cause of infections worldwide. The challenge in treating S. aureus infection is linked to the development of multidrug-resistant strains and the mechanisms employed by this pathogen to evade the human immune defenses. In addition, S. aureus can hide asymptomatically in particular ‘protective’ niches of the human body for prolonged periods of time. In the present review, we highlight recently gained insights in the role of the human gut as an endogenous S. aureus reservoir next to the nasopharynx and oral cavity. In addition, we address the contribution of these ecological niches to staphylococcal transmission, including the roles of particular triggers as modulators of the bacterial dissemination. In this context, we present recent advances concerning the interactions between S. aureus and immune cells to understand their possible roles as vehicles of dissemination from the gut to other body sites. Lastly, we discuss the factors that contribute to the switch from colonization to infection. Altogether, we conclude that an important key to uncovering the pathogenesis of S. aureus infection lies hidden in the endogenous staphylococcal reservoirs, the trafficking of this bacterium through the human body and the subsequent immune responses.
Collapse
Affiliation(s)
- Elisa J M Raineri
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dania Altulea
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Abstract
Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host's defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.
Collapse
Affiliation(s)
- Mariane Pivard
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
22
|
Nataraj BH, Ramesh C, Mallappa RH. Extractable surface proteins of indigenous probiotic strains confer anti-adhesion knack and protect against methicillin-resistant Staphylococcus aureus induced epithelial hyperpermeability in HT-29 cell line. Microb Pathog 2021; 158:104974. [PMID: 34015494 DOI: 10.1016/j.micpath.2021.104974] [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: 01/14/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 11/28/2022]
Abstract
Probiotic intervention has been long believed to have beneficial effects on human health by curbing the intestinal colonization of pathogens. However, the application of live probiotics therapy may not be an ideal approach to circumvent the infections of superbug origin due to the risk of horizontal antibiotic resistance genes transfer. In this study, the anti-adhesion ability of extractable cell surface proteins from two indigenous potential probiotic strains (Lactiplantibacillus plantarum A5 and Limosilactobacillus fermentum Lf1) and two standard reference strains (Lactobacillus acidophilus NCFM and Lacticaseibacillus rhamnosus LGG) was evaluated against clinical isolates of Methicillin-Resistant Staphylococcus aureus (MRSA) on porcine gastric mucin and HT-29 cells. The surface proteins from the probiotic strains were extracted by treatment with 5 M lithium chloride. The surface protein quantification and SDS-PAGE profiling indicated that the yield and protein patterns were strain-specific. Surface proteins significantly hampered the mucoadhesion of MRSA isolates via protective, competitive, and displacement. Similarly, the treatment with surface proteins probiotic strains displayed anti-adhesion against MRSA isolates on HT-29 cells without affecting the viability of the cell line. Surface proteins treatment to the confluent monolayer of HT-29 cells maintained the epithelial integrity; however, MRSA isolates (109 cells/mL) showed considerable alteration in the epithelial integrity by exacerbating the FITC-dextran transflux. Contrarily, the co-treatment with surface proteins with MRSA isolates significantly lowered the FITC-dextran transflux across the differentiated HT-29 monolayer. Overall, the findings of this study suggest that probiotic-derived surface proteins could be the novel biotherapeutics to combat the MRSA colonization and their concomitant intestinal infections.
Collapse
Affiliation(s)
| | - Chette Ramesh
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Rashmi Hogarehalli Mallappa
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India.
| |
Collapse
|
23
|
Antibiotic Resistance Crisis: An Update on Antagonistic Interactions between Probiotics and Methicillin-Resistant Staphylococcus aureus (MRSA). Curr Microbiol 2021; 78:2194-2211. [PMID: 33881575 DOI: 10.1007/s00284-021-02442-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
Antimicrobial resistance (AMR) havoc is a global multifaceted crisis endowing a significant challenge for the successful eradication of devastating pathogens. Methicillin-Resistant Staphylococcus aureus (MRSA) is an enduring superbug involved in causing devastating infections. Although MRSA is a frequent colonizer of human skin, wound, and anterior nares, the intestinal colonization of MRSA has greatly increased the risk of inducing MRSA-associated colitis besides creating a conducive environment for horizontal transfer of resistant genes to commensal microbes. On the other hand, staphylococcal resistance to last-resort antibiotics has urged the development of novel antimicrobial agents for the effective decolonization of MRSA. In response, probiotics and their metabolites (postbiotics) have been proposed as the adjunct therapeutic avenues. Probiotics exhibit a multitude of anti-MRSA actions (anti-bacterial, anti-biofilm, anti-virulence, anti-drug resistance, co-aggregation, and anti-quorum sensing) through the production of numerous antagonistic compounds such as organic acids, hydrogen peroxide, low molecular weight compounds, biosurfactants, bacteriocins, and bacteriocins like inhibitory substances. Besides, probiotics stabilize the epithelial barrier function and positively modulate the host immune system via regulating various signal transduction mechanisms. Preclinical and human intervention studies have suggested that probiotics outcompete with MRSA by exhibiting anti-colonization mechanisms via protective, competitive, and displacement mode. In this review, we aim to highlight the dynamics of MRSA associated virulence and drug resistance properties, and how probiotics antagonize MRSA through various mechanism of action.
Collapse
|
24
|
Schuler F, Barth PJ, Niemann S, Schaumburg F. A Narrative Review on the Role of Staphylococcus aureus Bacteriuria in S. aureus Bacteremia. Open Forum Infect Dis 2021; 8:ofab158. [PMID: 34189162 PMCID: PMC8233567 DOI: 10.1093/ofid/ofab158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/25/2021] [Indexed: 11/12/2022] Open
Abstract
Staphylococcus aureus bacteriuria (SABU) can occur in patients with S. aureus bacteremia (SAB). However, little is known on the (molecular) pathomechanisms of the renal passage of S. aureus. This review discusses the epidemiology and pathogenesis of SABU in patients with SAB and identifies knowledge gaps. The literature search was restricted to the English language. The prevalence of SABU in patients with SAB is 7.8%-39% depending on the study design. The main risk factor for SABU is urinary tract catheterization. SABU in SAB patients is associated with increased mortality. Given present evidence, hematogenous seeding-as seen in animal models-and the development of micro-abscesses best describe the translocation of S. aureus from blood to urine. Virulence factors that might be involved are adhesion factors, sortase A, and coagulase, among others. Other potential routes of bacterial translocation (eg, transcytosis, paracytosis, translocation via "Trojan horses") were identified as knowledge gaps.
Collapse
Affiliation(s)
- Franziska Schuler
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Peter J Barth
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
25
|
Effects of Bovine Colostrum with or without Egg on In Vitro Bacterial-Induced Intestinal Damage with Relevance for SIBO and Infectious Diarrhea. Nutrients 2021; 13:nu13031024. [PMID: 33809940 PMCID: PMC8004259 DOI: 10.3390/nu13031024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 02/08/2023] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) occurs commonly, is difficult to treat, and frequently recurs. Bovine colostrum (BC) and chicken eggs contain immunoglobulins and other components that possess antimicrobial, immunoregulatory, and growth factor activities; however, it is not known if they have the ability to reduce injury caused by the presence of bacteria associated with SIBO (Streptococcus, Escherichia coli, Staphylococcus, Bacteroides, Klebsiella, Enterococcus, and Proteus) and infectious diarrhea (enteropathogenic Escherichia coli, Salmonella). We examined the effects of BC, egg, or the combination, on bacterial growth and bacteria-induced changes in transepithelial electrical resistance (TEER) and bacterial translocation across confluent Caco-2 monolayers. BC, egg, or the combination did not affect bacterial growth. Adding bacteria to monolayers reduced TEER and (with minor variations among species) increased bacterial translocation, increased monolayer apoptosis (increased caspase-3 and Baxα, reduced Bcl2), increased intercellular adhesion molecule 1 (ICAM-1), and reduced cell adhesion molecules zonulin1 (ZO1) and claudin-1. BC, egg, or the combination reduced these effects (all p < 0.01) and caused additional increases in vascular endothelial growth factor (VEGF) and Heat Shock Protein 70 (Hsp70) expression. We conclude that BC ± egg strengthens mucosal integrity against a battery of bacteria relevant for SIBO and for infectious diarrhea. Oral BC ± egg may have clinical value for these conditions, especially SIBO where eradication of precipitating organisms may be difficult to achieve.
Collapse
|
26
|
Tight Junctions as a Key for Pathogens Invasion in Intestinal Epithelial Cells. Int J Mol Sci 2021; 22:ijms22052506. [PMID: 33801524 PMCID: PMC7958858 DOI: 10.3390/ijms22052506] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Tight junctions play a major role in maintaining the integrity and impermeability of the intestinal barrier. As such, they act as an ideal target for pathogens to promote their translocation through the intestinal mucosa and invade their host. Different strategies are used by pathogens, aimed at directly destabilizing the junctional network or modulating the different signaling pathways involved in the modulation of these junctions. After a brief presentation of the organization and modulation of tight junctions, we provide the state of the art of the molecular mechanisms leading to permeability breakdown of the gut barrier as a consequence of tight junctions’ attack by pathogens, including bacteria, viruses, fungi, and parasites.
Collapse
|
27
|
Dash S, Duraivelan K, Samanta D. Cadherin-mediated host-pathogen interactions. Cell Microbiol 2021; 23:e13316. [PMID: 33543826 DOI: 10.1111/cmi.13316] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/04/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Cell adhesion molecules mediate cell-to-cell and cell-to-matrix adhesions and play an immense role in a myriad of physiological processes during the growth and development of a multicellular organism. Cadherins belong to a major group of membrane-bound cell surface proteins that, in coordination with nectins, drive the formation and maintenance of adherens junctions for mediating cell to cell adhesion, cellular communication and signalling. Alongside adhesive function, the involvement of cadherins in mediating host-pathogen interactions has been extensively explored in recent years. In this review, we provide an in-depth understanding of microbial pathogens and their virulence factors that exploit cadherins for their strategical invasion into the host cell. Furthermore, macromolecular interactions involving cadherins and various microbial factors such as secretory toxins and adhesins lead to the disintegration of host cell junctions followed by the entry of the pathogen or triggering downstream signalling pathways responsible for successful invasion of the pathogenic microbes are discussed. Besides providing a comprehensive insight into some of the structural complexes involving cadherins and microbial factors to offer the mechanistic details of host-pathogen interactions, the current review also highlights novel constituents of various cell signalling events such as endocytosis machinery elicited upon microbial infections.
Collapse
Affiliation(s)
- Sagarika Dash
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Dibyendu Samanta
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
28
|
Olshan KL, Leonard MM, Serena G, Zomorrodi AR, Fasano A. Gut microbiota in Celiac Disease: microbes, metabolites, pathways and therapeutics. Expert Rev Clin Immunol 2020; 16:1075-1092. [PMID: 33103934 DOI: 10.1080/1744666x.2021.1840354] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Current evidence supports a vital role of the microbiota on health outcomes, with alterations in an otherwise healthy balance linked to chronic medical conditions like celiac disease (CD). Recent advances in microbiome analysis allow for unparalleled profiling of the microbes and metabolites. With the growing volume of data available, trends are emerging that support a role for the gut microbiota in CD pathogenesis. AREAS COVERED In this article, the authors review the relationship between factors such as genes and antibiotic exposure on CD onset and the intestinal microbiota. The authors also review other microbiota within the human body (like the oropharynx) that may play a role in CD pathogenesis. Finally, the authors discuss implications for disease modification and the ultimate goal of prevention. The authors reviewed literature from PubMed, EMBASE, and Web of Science. EXPERT OPINION CD serves as a unique opportunity to explore the role of the intestinal microbiota on the development of chronic autoimmune disease. While research to date provides a solid foundation, most studies have been case-control and thus do not have capacity to explore the mechanistic role of the microbiota in CD onset. Further longitudinal studies and integrated multi-omics are necessary for investigating CD pathogenesis.
Collapse
Affiliation(s)
- Katherine L Olshan
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Ali R Zomorrodi
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,European Biomedical Research Institute of Salerno (EBRIS) , Salerno, Italy
| |
Collapse
|
29
|
Abstract
Despite being regarded as an extracellular bacterium, the pathogen Staphylococcus aureus can invade and survive within human cells. The intracellular niche is considered a hideout from the host immune system and antibiotic treatment and allows bacterial proliferation. The opportunistic human pathogen Staphylococcus aureus causes serious infectious diseases that range from superficial skin and soft tissue infections to necrotizing pneumonia and sepsis. While classically regarded as an extracellular pathogen, S. aureus is able to invade and survive within human cells. Host cell exit is associated with cell death, tissue destruction, and the spread of infection. The exact molecular mechanism employed by S. aureus to escape the host cell is still unclear. In this study, we performed a genome-wide small hairpin RNA (shRNA) screen and identified the calcium signaling pathway as being involved in intracellular infection. S. aureus induced a massive cytosolic Ca2+ increase in epithelial host cells after invasion and intracellular replication of the pathogen. This was paralleled by a decrease in endoplasmic reticulum Ca2+ concentration. Additionally, calcium ions from the extracellular space contributed to the cytosolic Ca2+ increase. As a consequence, we observed that the cytoplasmic Ca2+ rise led to an increase in mitochondrial Ca2+ concentration, the activation of calpains and caspases, and eventually to cell lysis of S. aureus-infected cells. Our study therefore suggests that intracellular S. aureus disturbs the host cell Ca2+ homeostasis and induces cytoplasmic Ca2+ overload, which results in both apoptotic and necrotic cell death in parallel or succession.
Collapse
|
30
|
Bianchi F, van den Bogaart G. Vacuolar escape of foodborne bacterial pathogens. J Cell Sci 2020; 134:134/5/jcs247221. [PMID: 32873733 DOI: 10.1242/jcs.247221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The intracellular pathogens Listeria monocytogenes, Salmonella enterica, Shigella spp. and Staphylococcus aureus are major causes of foodborne illnesses. Following the ingestion of contaminated food or beverages, pathogens can invade epithelial cells, immune cells and other cell types. Pathogens survive and proliferate intracellularly via two main strategies. First, the pathogens can remain in membrane-bound vacuoles and tailor organellar trafficking to evade host-cell defenses and gain access to nutrients. Second, pathogens can rupture the vacuolar membrane and proliferate within the nutrient-rich cytosol of the host cell. Although this virulence strategy of vacuolar escape is well known for L. monocytogenes and Shigella spp., it has recently become clear that S. aureus and Salmonella spp. also gain access to the cytosol, and that this is important for their survival and growth. In this Review, we discuss the molecular mechanisms of how these intracellular pathogens rupture the vacuolar membrane by secreting a combination of proteins that lyse the membranes or that remodel the lipids of the vacuolar membrane, such as phospholipases. In addition, we also propose that oxidation of the vacuolar membrane also contributes to cytosolic pathogen escape. Understanding these escape mechanisms could aid in the identification of new therapeutic approaches to combat foodborne pathogens.
Collapse
Affiliation(s)
- Frans Bianchi
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9722GR Groningen, The Netherlands
| | - Geert van den Bogaart
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9722GR Groningen, The Netherlands .,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 9625GA Nijmegen, The Netherlands
| |
Collapse
|
31
|
Bonifacius A, Goldmann O, Floess S, Holtfreter S, Robert PA, Nordengrün M, Kruse F, Lochner M, Falk CS, Schmitz I, Bröker BM, Medina E, Huehn J. Staphylococcus aureus Alpha-Toxin Limits Type 1 While Fostering Type 3 Immune Responses. Front Immunol 2020; 11:1579. [PMID: 32849537 PMCID: PMC7427519 DOI: 10.3389/fimmu.2020.01579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus can cause life-threatening diseases, and hospital- as well as community-associated antibiotic-resistant strains are an emerging global public health problem. Therefore, prophylactic vaccines or immune-based therapies are considered as alternative treatment opportunities. To develop such novel treatment approaches, a better understanding of the bacterial virulence and immune evasion mechanisms and their potential effects on immune-based therapies is essential. One important staphylococcal virulence factor is alpha-toxin, which is able to disrupt the epithelial barrier in order to establish infection. In addition, alpha-toxin has been reported to modulate other cell types including immune cells. Since CD4+ T cell-mediated immunity is required for protection against S. aureus infection, we were interested in the ability of alpha-toxin to directly modulate CD4+ T cells. To address this, murine naïve CD4+ T cells were differentiated in vitro into effector T cell subsets in the presence of alpha-toxin. Interestingly, alpha-toxin induced death of Th1-polarized cells, while cells polarized under Th17 conditions showed a high resistance toward increasing concentrations of this toxin. These effects could neither be explained by differential expression of the cellular alpha-toxin receptor ADAM10 nor by differential activation of caspases, but might result from an increased susceptibility of Th1 cells toward Ca2+-mediated activation-induced cell death. In accordance with the in vitro findings, an alpha-toxin-dependent decrease of Th1 and concomitant increase of Th17 cells was observed in vivo during S. aureus bacteremia. Interestingly, corresponding subsets of innate lymphoid cells and γδ T cells were similarly affected, suggesting a more general effect of alpha-toxin on the modulation of type 1 and type 3 immune responses. In conclusion, we have identified a novel alpha-toxin-dependent immunomodulatory strategy of S. aureus, which can directly act on CD4+ T cells and might be exploited for the development of novel immune-based therapeutic approaches to treat infections with antibiotic-resistant S. aureus strains.
Collapse
Affiliation(s)
- Agnes Bonifacius
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Oliver Goldmann
- Department Infection Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Philippe A Robert
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maria Nordengrün
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Friederike Kruse
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hanover, Germany.,Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hanover, Germany
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical School, Hanover, Germany.,DZIF, German Center for Infectious Diseases, TTU-IICH Hannover-Braunschweig Site, Hanover, Germany
| | - Ingo Schmitz
- Department Systems-Oriented Immunology and Inflammation Research, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Department of Molecular Immunology, Ruhr-University Bochum, Bochum, Germany
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Eva Medina
- Department Infection Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
32
|
Hamouda RA, Yousuf WE, Mohammed ABA, Mohammed RS, Darwish DB, Abdeen EE. Comparative study between zinc oxide nanoparticles synthesis by biogenic and wet chemical methods in vivo and in vitro against Staphylococcus aureus. Microb Pathog 2020; 147:104384. [PMID: 32679246 DOI: 10.1016/j.micpath.2020.104384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/13/2020] [Accepted: 07/06/2020] [Indexed: 11/19/2022]
Abstract
ZnO nanoparticles (ZnO-NPs) can be used as nano medicine for Staphylococcus aureus infection, which causes deleterious effects on liver, kidney and lung tissue, as it causes catarrhal bronchitis, peri-bronchial oedema, lymphocytic granulomas, oedematous fluid and haemorrhage inside the bronchi, and interstitial pneumonia. In this research ZnO nanoparticle (ZnO-NPs) synthesis by biogenic method using green alga Ulva fasciata and by wet chemical method. Both of them tested in vitro and in vivo against Staphylococcus aureus. The characterization of ZnO-NPs was detected by U.V spectroscopy, Fourier-transform infrared (FTIR), Energy dispersive X-ray spectroscopy (EDX), X-ray diffraction (XRD), Scanning Electron Microscope (SEM) and Transmission Electron Microscope (TEM). In vivo assessment eight groups, each group contain of five rats and the treatment as follow (1) an uninfected control group; (2) an infected group; groups (3), (4), and (5) were injected with biogenic or chemical ZnO-NPs or zinc acetate, as the bulk group, respectively; and groups (6), (7) and (8) were infected and then treated in the same manner as groups (3), (4), and (5), respectively. The blood profile, biochemical parameters, phagocytic activity and histological assessment of liver, kidney and lung tissue of each rat was investigated after 20 days. The rats treated with 5 mg/1 kg natural ZnO-NPs showed improved lung characteristics, and the number of platelets in the infected groups treated with ZnO-NPs from chemical and natural sources (G6 and G7) was close to those in the control group. However, the trend was reversed for regarding lymphocytes, which remained at higher levels in uninfected animals treated with synthetic ZnO-NPs (G4) than in infected rats treated with synthetic ZnO-NPs (G7). Moreover, a significant difference in phagocytic activity was found among all groups compared to that of controls. Compared to control group rats (G1), uninfected rats injected with only natural ZnO-NPs (G3) showed a significant (P < 0.05) improvement in the phagocytic index. We propose that ZnO-NPs produced from natural sources are preferable to those produced from chemical sources for use as nano medicine for the treatment of S. aureus infection in albino rats.
Collapse
Affiliation(s)
- Ragaa A Hamouda
- Department of Biology, Faculty of Sciences and Arts Khulais, University of Jeddah, Saudi Arabia; Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt.
| | - Wesam E Yousuf
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - A B Abeer Mohammed
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Rasha Salah Mohammed
- Department of Animal and Poultry Health, Animal and Poultry Production Division, DRC, Cairo, Egypt
| | - Doaa B Darwish
- Botany Department, Faculty of Science, Mansoura University, Egypt; Department of Biology, Faculty of Science, University of Tabuk, Saudi Arabia
| | - Eman E Abdeen
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt.
| |
Collapse
|
33
|
Li L, Wang G, Cheung A, Abdelhady W, Seidl K, Xiong YQ. MgrA Governs Adherence, Host Cell Interaction, and Virulence in a Murine Model of Bacteremia Due to Staphylococcus aureus. J Infect Dis 2020; 220:1019-1028. [PMID: 31177268 DOI: 10.1093/infdis/jiz219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/26/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND MgrA is an important global virulence gene regulator in Staphylococcus aureus. In the present study, the role of mgrA in host-pathogen interactions related to virulence was explored in both methicillin-resistant S. aureus (MRSA) and methicillin-susceptible S. aureus (MSSA) strains. METHODS In vitro susceptibilities to human defense peptides (HDPs), adherence to fibronectin (Fn) and endothelial cells (ECs), EC damage, α-toxin production, expression of global regulator (eg, agr RNAIII) and its downstream effectors (eg, α-toxin [hla] and Fn binding protein A [fnbA]), MgrA binding to fnbA promoter, and the effect on HDP-induced mprF and dltA expression were analyzed. The impact of mgrA on virulence was evaluated using a mouse bacteremia model. RESULTS mgrA mutants displayed significantly higher susceptibility to HDPs, which might be related to the decreased HDP-induced mprF and dltA expression but decreased Fn and EC adherence, EC damage, α-toxin production, agr RNAIII, hla and fnbA expression, and attenuated virulence in the bacteremia model as compared to their respective parental and mgrA-complemented strains. Importantly, direct binding of MgrA to the fnbA promoter was observed. CONCLUSIONS These results suggest that mgrA mediates host-pathogen interactions and virulence and may provide a novel therapeutic target for invasive S. aureus infections.
Collapse
Affiliation(s)
- Liang Li
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance
| | - Genzhu Wang
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance
| | | | - Wessam Abdelhady
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance
| | - Kati Seidl
- University Hospital of Zurich, Switzerland
| | - Yan Q Xiong
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance.,David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
34
|
Clostridium perfringens Epsilon-Toxin Impairs the Barrier Function in MDCK Cell Monolayers in a Ca 2+-Dependent Manner. Toxins (Basel) 2020; 12:toxins12050286. [PMID: 32365779 PMCID: PMC7291203 DOI: 10.3390/toxins12050286] [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: 03/27/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022] Open
Abstract
Epsilon-toxin produced by Clostridium perfringens significantly contributes to the pathogeneses of enterotoxemia in ruminants and multiple sclerosis in humans. Epsilon-toxin forms a heptameric oligomer in the host cell membrane, promoting cell disruption. Here, we investigate the effect of epsilon-toxin on epithelial barrier functions. Epsilon-toxin impairs the barrier integrity of Madin-Darby Canine Kidney (MDCK) cells, as demonstrated by decreased transepithelial electrical resistance (TEER), increased paracellular flux marker permeability, and the decreased cellular localization of junctional proteins, such as occludin, ZO-1, and claudin-1. U73122, an endogenous phospholipase C (PLC) inhibitor, inhibited the decrease in TEER and the increase in the permeability of flux marker induced by epsilon-toxin. The application of epsilon-toxin to MDCK cells resulted in the biphasic formation of 1,2-diacylglycerol (DAG) and inositol-1,4,5-triphosphate (IP3). U73122 blocked the formation of DAG and IP3 induced by the toxin. Epsilon-toxin also specifically activated endogenous PLC-γ1. Epsilon-toxin dose-dependently increased the cytosolic calcium ion concentration ([Ca2+]i). The toxin-induced elevation of [Ca2+]i was inhibited by U73122. Cofilin is a key regulator of actin cytoskeleton turnover and tight-junction (TJ) permeability regulation. Epsilon-toxin caused cofilin dephosphorylation. These results demonstrate that epsilon-toxin induces Ca2+ influx through activating the phosphorylation of PLC-γ1 and then causes TJ opening accompanied by cofilin dephosphorylation.
Collapse
|
35
|
Joyce P, Ulmefors H, Maghrebi S, Subramaniam S, Wignall A, Jõemetsa S, Höök F, Prestidge CA. Enhancing the Cellular Uptake and Antibacterial Activity of Rifampicin through Encapsulation in Mesoporous Silica Nanoparticles. NANOMATERIALS 2020; 10:nano10040815. [PMID: 32344619 PMCID: PMC7221943 DOI: 10.3390/nano10040815] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
An urgent demand exists for the development of novel delivery systems that efficiently transport antibacterial agents across cellular membranes for the eradication of intracellular pathogens. In this study, the clinically relevant poorly water-soluble antibiotic, rifampicin, was confined within mesoporous silica nanoparticles (MSN) to investigate their ability to serve as an efficacious nanocarrier system against small colony variants of Staphylococcus aureus (SCV S. aureus) hosted within Caco-2 cells. The surface chemistry and particle size of MSN were varied through modifications during synthesis, where 40 nm particles with high silanol group densities promoted enhanced cellular uptake. Extensive biophysical analysis was performed, using quartz crystal microbalance with dissipation (QCM-D) and total internal reflection fluorescence (TIRF) microscopy, to elucidate the mechanism of MSN adsorption onto semi-native supported lipid bilayers (snSLB) and, thus, uncover potential cellular uptake mechanisms of MSN into Caco-2 cells. Such studies revealed that MSN with reduced silanol group densities were prone to greater particle aggregation on snSLB, which was expected to restrict endocytosis. MSN adsorption and uptake into Caco-2 cells correlated well with antibacterial efficacy against SCV S. aureus, with 40 nm hydrophilic particles triggering a ~2.5-log greater reduction in colony forming units, compared to the pure rifampicin. Thus, this study provides evidence for the potential to design silica nanocarrier systems with controlled surface chemistries that can be used to re-sensitise intracellular bacteria to antibiotics by delivering them to the site of infection.
Collapse
Affiliation(s)
- Paul Joyce
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; (P.J.); (S.J.); (F.H.)
| | - Hanna Ulmefors
- School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, South Australia 5090, Australia; (H.U.); (S.M.); (S.S.); (A.W.)
- ARC Centre of Excellence in Bio-Nano Science and Technology, University of South Australia, Adelaide, South Australia 5090, Australia
| | - Sajedeh Maghrebi
- School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, South Australia 5090, Australia; (H.U.); (S.M.); (S.S.); (A.W.)
- ARC Centre of Excellence in Bio-Nano Science and Technology, University of South Australia, Adelaide, South Australia 5090, Australia
| | - Santhni Subramaniam
- School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, South Australia 5090, Australia; (H.U.); (S.M.); (S.S.); (A.W.)
- ARC Centre of Excellence in Bio-Nano Science and Technology, University of South Australia, Adelaide, South Australia 5090, Australia
| | - Anthony Wignall
- School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, South Australia 5090, Australia; (H.U.); (S.M.); (S.S.); (A.W.)
- ARC Centre of Excellence in Bio-Nano Science and Technology, University of South Australia, Adelaide, South Australia 5090, Australia
| | - Silver Jõemetsa
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; (P.J.); (S.J.); (F.H.)
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden; (P.J.); (S.J.); (F.H.)
| | - Clive A. Prestidge
- School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, South Australia 5090, Australia; (H.U.); (S.M.); (S.S.); (A.W.)
- ARC Centre of Excellence in Bio-Nano Science and Technology, University of South Australia, Adelaide, South Australia 5090, Australia
- Correspondence:
| |
Collapse
|
36
|
Pan P, Bai L, Hua X, Wang Y, Jiang X, Cheng X, Song Y, Yu X. miR-155 Regulates claudin1 Expression in Humans With Intestinal Mucosa Dysfunction After Brain Injury. Transplant Proc 2020; 51:3474-3480. [PMID: 31810510 DOI: 10.1016/j.transproceed.2019.08.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/24/2019] [Accepted: 08/30/2019] [Indexed: 02/06/2023]
Abstract
Patients with craniocerebral trauma often have intestinal mucosal dysfunction, and the claudin1 protein plays an important role in intestinal mucosal function. Our previous work has shown that the expression of microRNA-155 (miR-155) in the peripheral blood of patients with craniocerebral trauma is decreased. Animal experiments also suggest that the expression of miR-155 is increased in the intestinal mucosa of mice with brain injury and the expression of claudin1 is decreased. We recruited 56 samples (35 patients with traumatic brain injury [TBI] and 21 patients without history of head trauma) to detect the expression of miR-155 on claudin1 regulation by quantitative polymerase chain reaction, reverse transcriptase polymerase chain reaction, and so on. We also used the receiver operating characteristic curve (ROC) to further evaluate the diagnostic value of the 2 biomarkers. From the results, we found that the expression level of miR-155 and claudin1 in the case group was lower than that in the control group. Human miR-155 (Hsa-miR-155) may positively regulate intestinal mucosal function by inhibiting the expression of claudin1, leading to intestinal mucosal barrier dysfunction. Combining the ROC curve data, the results further prove that miR-155 and claudin1 might be the new clinical diagnostic markers and treatment targets for the intestinal mucosal barrier dysfunction after TBI.
Collapse
Affiliation(s)
- Pengfei Pan
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Linlin Bai
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Xiaoli Hua
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Yuqiang Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Xiaofang Jiang
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Xi Cheng
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Yunlin Song
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China.
| | - Xiangyou Yu
- Department of Critical Care Medicine, the First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| |
Collapse
|
37
|
Shah J, Rouaud F, Guerrera D, Vasileva E, Popov LM, Kelley WL, Rubinstein E, Carette JE, Amieva MR, Citi S. A Dock-and-Lock Mechanism Clusters ADAM10 at Cell-Cell Junctions to Promote α-Toxin Cytotoxicity. Cell Rep 2019; 25:2132-2147.e7. [PMID: 30463011 DOI: 10.1016/j.celrep.2018.10.088] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/01/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
We previously identified PLEKHA7 and other junctional proteins as host factors mediating death by S. aureus α-toxin, but the mechanism through which junctions promote toxicity was unclear. Using cell biological and biochemical methods, we now show that ADAM10 is docked to junctions by its transmembrane partner Tspan33, whose cytoplasmic C terminus binds to the WW domain of PLEKHA7 in the presence of PDZD11. ADAM10 is locked at junctions through binding of its cytoplasmic C terminus to afadin. Junctionally clustered ADAM10 supports the efficient formation of stable toxin pores. Instead, disruption of the PLEKHA7-PDZD11 complex inhibits ADAM10 and toxin junctional clustering. This promotes toxin pore removal from the cell surface through an actin- and macropinocytosis-dependent process, resulting in cell recovery from initial injury and survival. These results uncover a dock-and-lock molecular mechanism to target ADAM10 to junctions and provide a paradigm for how junctions regulate transmembrane receptors through their clustering.
Collapse
Affiliation(s)
- Jimit Shah
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Florian Rouaud
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Diego Guerrera
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Ekaterina Vasileva
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Lauren M Popov
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William L Kelley
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1211-4 Geneva, Switzerland
| | - Eric Rubinstein
- INSERM, Université Paris-Sud, UMRS_935, 94807 Villejuif Cedex, France
| | - Jan E Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sandra Citi
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland.
| |
Collapse
|
38
|
Livingston ET, Mursalin MH, Callegan MC. A Pyrrhic Victory: The PMN Response to Ocular Bacterial Infections. Microorganisms 2019; 7:E537. [PMID: 31703354 PMCID: PMC6920826 DOI: 10.3390/microorganisms7110537] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Some tissues of the eye are susceptible to damage due to their exposure to the outside environment and inability to regenerate. Immune privilege, although beneficial to the eye in terms of homeostasis and protection, can be harmful when breached or when an aberrant response occurs in the face of challenge. In this review, we highlight the role of the PMN (polymorphonuclear leukocyte) in different bacterial ocular infections that invade the immune privileged eye at the anterior and posterior segments: keratitis, conjunctivitis, uveitis, and endophthalmitis. Interestingly, the PMN response from the host seems to be necessary for pathogen clearance in ocular disease, but the inflammatory response can also be detrimental to vision retention. This "Pyrrhic Victory" scenario is explored in each type of ocular infection, with details on PMN recruitment and response at the site of ocular infection. In addition, we emphasize the differences in PMN responses between each ocular disease and its most common corresponding bacterial pathogen. The in vitro and animal models used to identify PMN responses, such as recruitment, phagocytosis, degranulation, and NETosis, are also outlined in each ocular infection. This detailed study of the ocular acute immune response to infection could provide novel therapeutic strategies for blinding diseases, provide more general information on ocular PMN responses, and reveal areas of bacterial ocular infection research that lack PMN response studies.
Collapse
Affiliation(s)
- Erin T. Livingston
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (E.T.L.); (M.H.M.)
| | - Md Huzzatul Mursalin
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (E.T.L.); (M.H.M.)
| | - Michelle C. Callegan
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (E.T.L.); (M.H.M.)
- Department of Ophthalmology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Neuroscience, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| |
Collapse
|
39
|
Seike S, Takehara M, Kobayashi K, Nagahama M. Clostridium perfringens Delta-Toxin Damages the Mouse Small Intestine. Toxins (Basel) 2019; 11:toxins11040232. [PMID: 31013617 PMCID: PMC6520758 DOI: 10.3390/toxins11040232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/08/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
Clostridium perfringens strains B and C cause fatal intestinal diseases in animals. The secreted pore-forming toxin delta-toxin is one of the virulence factors of the strains, but the mechanism of intestinal pathogenesis is unclear. Here, we investigated the effects of delta-toxin on the mouse ileal loop. Delta-toxin caused fluid accumulation and intestinal permeability to fluorescein isothiocyanate (FITC)-dextran in the mouse ileal loop in a dose- and time-dependent manner. Treatment with delta-toxin induced significant histological damage and shortening of villi. Delta-toxin activates a disintegrin and metalloprotease (ADAM) 10, leading to the cleavage of E-cadherin, the epithelial adherens junction protein, in human intestinal epithelial Caco-2 cells. In this study, E-cadherin immunostaining in mouse intestinal epithelial cells was almost undetectable 1 h after toxin treatment. ADAM10 inhibitor (GI254023X) blocked the toxin-induced fluid accumulation and E-cadherin loss in the mouse ileal loop. Delta-toxin stimulated the shedding of intestinal epithelial cells. The shedding cells showed the accumulation of E-cadherin in intracellular vesicles and the increased expression of active caspase-3. Our findings demonstrate that delta-toxin causes intestinal epithelial cell damage through the loss of E-cadherin cleaved by ADAM10.
Collapse
Affiliation(s)
- Soshi Seike
- Laboratory of Molecular Microbiological Science, Faculty of Pharmaceutical Sciences, Hiroshima International University, Kure, Hiroshima 737-0112, Japan.
| | - Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| |
Collapse
|
40
|
Kalsi KK, Garnett JP, Patkee W, Weekes A, Dockrell ME, Baker EH, Baines DL. Metformin attenuates the effect of Staphylococcus aureus on airway tight junctions by increasing PKCζ-mediated phosphorylation of occludin. J Cell Mol Med 2018; 23:317-327. [PMID: 30450773 PMCID: PMC6307806 DOI: 10.1111/jcmm.13929] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/29/2018] [Indexed: 01/03/2023] Open
Abstract
Airway epithelial tight junction (TJ) proteins form a resistive barrier to the external environment, however, during respiratory bacterial infection TJs become disrupted compromising barrier function. This promotes glucose flux/accumulation into the lumen which acts as a nutrient source for bacterial growth. Metformin used for the treatment of diabetes increases transepithelial resistance (TEER) and partially prevents the effect of bacteria but the mechanisms of action are unclear. We investigated the effect of metformin and Staphylococcus aureus on TJ proteins, zonula occludins (ZO)-1 and occludin in human airway epithelial cells (H441). We also explored the role of AMP-activated protein kinase (AMPK) and PKCζ in metformin-induced effects. Pretreatment with metformin prevented the S. aureus-induced changes in ZO-1 and occludin. Metformin also promoted increased abundance of full length over smaller cleaved occludin proteins. The nonspecific PKC inhibitor staurosporine reduced TEER but did not prevent the effect of metformin indicating that the pathway may involve atypical PKC isoforms. Investigation of TJ reassembly after calcium depletion showed that metformin increased TEER more rapidly and promoted the abundance and localization of occludin at the TJ. These effects were inhibited by the AMPK inhibitor, compound C and the PKCζ pseudosubstrate inhibitor (PSI). Metformin increased phosphorylation of occludin and acetyl-coA-carboxylase but only the former was prevented by PSI. This study demonstrates that metformin improves TJ barrier function by promoting the abundance and assembly of full length occludin at the TJ and that this process involves phosphorylation of the protein via an AMPK-PKCζ pathway.
Collapse
Affiliation(s)
- Kameljit K. Kalsi
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - James P. Garnett
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
- Present address:
Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - Wishwanath Patkee
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Alexina Weekes
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Mark E. Dockrell
- South West Thames Institute for Renal ResearchSt Helier HospitalCarshaltonUK
| | - Emma H. Baker
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Deborah L. Baines
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| |
Collapse
|
41
|
Murphy J, Ramezanpour M, Drilling A, Roscioli E, Psaltis AJ, Wormald PJ, Vreugde S. In vitro characteristics of an airway barrier-disrupting factor secreted by Staphylococcus aureus. Int Forum Allergy Rhinol 2018; 9:187-196. [PMID: 30431711 DOI: 10.1002/alr.22232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/25/2018] [Accepted: 10/05/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Staphylococcus aureus is a major contributor to the pathophysiology of chronic rhinosinusitis (CRS). Previous research has shown that S. aureus-secreted products disrupt the airway barrier. METHODS S. aureus ATCC 13565 and 25923 strains were grown at exponential, postexponential, and stationary phases. Microbial conditioned media (CM) was collected from the cultures and ultrafiltered (UF). Liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) was performed on the UF-CM. UF-CM was subjected to heat and protease treatment, size fractionation, and ultracentrifugation (UC) separation. Human nasal epithelial cells grown at air-liquid interface (HNEC-ALI) cultures were exposed to purified alpha hemolysin (Hla), staphylococcal enterotoxin A (SEA), lipoteichoic acid (LTA), and UF-CM. Barrier function outcomes were measured by transepithelial electrical resistance (TEER) and apparent permeability (Papp). UC fraction exposed cultures were subjected to immunofluorescence microscopy for tight junction (TJ) protein zonula occludens-1 (ZO-1). RESULTS LC-ESI-MS/MS identified 107 proteins, with Hla being most abundant. Hla, SEA, and LTA did not alter the HNEC-ALI barrier as measured by TEER or Papp. Barrier disruption caused by UF-CM peaked in the postexponential phase, was sensitive to heat and protease treatment, >30-kDa in size, and enriched in the UC fraction. HNEC-ALI exposed to UF-CM and UC demonstrated loss of ZO-1 localization. CONCLUSION These results suggest that the S. aureus factor responsible for TJ disruption in HNEC-ALI cultures is either a protein-macromolecule or a combination of secreted factors. The product is enriched in the UC fraction, suggesting it is associated with large structures such as membrane components or vesicles.
Collapse
Affiliation(s)
- Jae Murphy
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Mahnaz Ramezanpour
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Amanda Drilling
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Eugene Roscioli
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Alkis James Psaltis
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Peter-John Wormald
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sarah Vreugde
- Department of Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| |
Collapse
|
42
|
Smith A, Moravcova S, Treibel TA, Colque-Navarro P, Mollby R, Moon JC, Hamilton-Davies C. Relationship between endotoxin core, staphylococcal and varicella antibody levels and outcome following aortic valve replacement surgery: a prospective observational study. Perioper Med (Lond) 2018; 7:20. [PMID: 30250733 PMCID: PMC6146602 DOI: 10.1186/s13741-018-0101-z] [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: 05/21/2018] [Accepted: 08/02/2018] [Indexed: 11/10/2022] Open
Abstract
Background Morbidity and mortality following cardiac valve surgery is high. Immunity is an important contributor to outcome. This study examines the relationship of staphylococcal and endotoxin antibody levels to outcome following cardiac surgery. Methods Using enzyme-linked immunosorbent assays (ELISA), we measured pre-operative levels of antibodies to endotoxin core (EndoCAb); 3 common staphylococcal epitopes and varicella on saved serum of 60 adult patients scheduled to undergo elective primary surgical aortic valve replacement (AVR). Primary outcome measure was post-operative length of stay (LOS) in hospital with secondary outcomes being development of infective complications, length of stay on the intensive care unit (ICU) and 30-day mortality. Patients were quartiled according to antibody levels and outcomes compared between the quartile groups using Mann-Whitney tests for length of stay and Fisher's test for development of infection. Results Sixty patients (34 M, 26 F) were recruited with mean age 73 years (IQR 66-78), mean body mass index (BMI) 27.7 (IQR 25-31) and EuroSCORE II 1.44 (0.95-1.99). Those patients in the lower quartile for pre-operative antibody level had a longer post-operative stay than the upper quartile. EndoCAb (median IgG level Q1 42.2 MU/ml vs Q4 256 MU/ml) 9 vs 6 days, p = 0.025; alpha-toxin (median IgG level Q1 63 U vs Q4 558 U) 10 vs 7 days, p = 0.034; teichoic acid (median IgG level Q1 14 U vs Q4 419 U) 10 vs 8 days, p = 0.441; staphylococcal enterotoxin A (median IgG level Q1 55 U vs Q4 427 U) 9 vs 7 days, p = 0.865; varicella zoster (median IgG level Q1 1.325 U vs Q4 2.54 U) 8 vs 7 days, p = 1.0; and combined antibody levels 10 vs 6 days, p = 0.017. There were no differences in the number developing post-operative infections for each antibody type. The combined antibody analysis suggested a reduction in proportion of individuals developing infection from the upper vs lower quartile: 0 vs 0.33, p = 0.042. Conclusions This study again suggests the inverse relationship between endotoxin core antibody levels and outcome following aortic valve surgery as well as suggesting a similar relationship with antibodies to staphylococcus. There is no such relationship for antibody levels against an organism not providing a peri-operative threat. Understanding this relationship may enable therapeutic manipulation of immune status, re-evaluation of risk and further investigation of the low immune state. Trial registration The patients in this study are a sub-group of the RELIEF AS study.ClinicalTrials.gov identifier NCT02174471.
Collapse
Affiliation(s)
- Andrew Smith
- 1Queen Mary University London, London, UK.,2University College London, London, UK.,6Department for Peri-operative Medicine, St Bartholomew's Hospital, First floor, KGV Building, West Smithfield, London, UK
| | | | - Thomas A Treibel
- 4Institute for Cardiovascular Sciences, University College London, London, UK
| | | | | | - James C Moon
- 4Institute for Cardiovascular Sciences, University College London, London, UK
| | | |
Collapse
|
43
|
Bouillot S, Reboud E, Huber P. Functional Consequences of Calcium Influx Promoted by Bacterial Pore-Forming Toxins. Toxins (Basel) 2018; 10:toxins10100387. [PMID: 30257425 PMCID: PMC6215193 DOI: 10.3390/toxins10100387] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 02/06/2023] Open
Abstract
Bacterial pore-forming toxins induce a rapid and massive increase in cytosolic Ca2+ concentration due to the formation of pores in the plasma membrane and/or activation of Ca2+-channels. As Ca2+ is an essential messenger in cellular signaling, a sustained increase in Ca2+ concentration has dramatic consequences on cellular behavior, eventually leading to cell death. However, host cells have adapted mechanisms to protect against Ca2+ intoxication, such as Ca2+ efflux and membrane repair. The final outcome depends upon the nature and concentration of the toxin and on the cell type. This review highlights the repercussions of Ca2+ overload on the induction of cell death, repair mechanisms, cellular adhesive properties, and the inflammatory response.
Collapse
Affiliation(s)
- Stéphanie Bouillot
- Université Grenoble Alpes, CNRS ERL5261, CEA BIG-BCI, INSERM UMR1036, Grenoble 38054, France.
| | - Emeline Reboud
- Université Grenoble Alpes, CNRS ERL5261, CEA BIG-BCI, INSERM UMR1036, Grenoble 38054, France.
| | - Philippe Huber
- Université Grenoble Alpes, CNRS ERL5261, CEA BIG-BCI, INSERM UMR1036, Grenoble 38054, France.
| |
Collapse
|
44
|
Cutaneous Barriers and Skin Immunity: Differentiating A Connected Network. Trends Immunol 2018; 39:315-327. [DOI: 10.1016/j.it.2018.02.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 12/12/2022]
|
45
|
Kim S, Goel R, Kumar A, Qi Y, Lobaton G, Hosaka K, Mohammed M, Handberg EM, Richards EM, Pepine CJ, Raizada MK. Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. Clin Sci (Lond) 2018; 132:701-718. [PMID: 29507058 PMCID: PMC5955695 DOI: 10.1042/cs20180087] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/21/2018] [Accepted: 03/02/2018] [Indexed: 12/11/2022]
Abstract
Recent evidence indicates a link between gut pathology and microbiome with hypertension (HTN) in animal models. However, whether this association exists in humans is unknown. Thus, our objectives in the present study were to test the hypotheses that high blood pressure (BP) patients have distinct gut microbiomes and that gut-epithelial barrier function markers and microbiome composition could predict systolic BP (SBP). Fecal samples, analyzed by shotgun metagenomics, displayed taxonomic and functional changes, including altered butyrate production between patients with high BP and reference subjects. Significant increases in plasma of intestinal fatty acid binding protein (I-FABP), lipopolysaccharide (LPS), and augmented gut-targetting proinflammatory T helper 17 (Th17) cells in high BP patients demonstrated increased intestinal inflammation and permeability. Zonulin, a gut epithelial tight junction protein regulator, was markedly elevated, further supporting gut barrier dysfunction in high BP. Zonulin strongly correlated with SBP (R2 = 0.5301, P<0.0001). Two models predicting SBP were built using stepwise linear regression analysis of microbiome data and circulating markers of gut health, and validated in a separate cohort by prediction of SBP from zonulin in plasma (R2 = 0.4608, P<0.0001). The mouse model of HTN, chronic angiotensin II (Ang II) infusion, was used to confirm the effects of butyrate and gut barrier function on the cardiovascular system and BP. These results support our conclusion that intestinal barrier dysfunction and microbiome function are linked to HTN in humans. They suggest that manipulation of gut microbiome and its barrier functions could be the new therapeutic and diagnostic avenues for HTN.
Collapse
Affiliation(s)
- Seungbum Kim
- Department of Physiology and Functional, University of Florida, Gainesville, FL, U.S.A
| | - Ruby Goel
- Department of Physiology and Functional, University of Florida, Gainesville, FL, U.S.A
| | - Ashok Kumar
- Department of Physiology and Functional, University of Florida, Gainesville, FL, U.S.A
| | - Yanfei Qi
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, U.S.A
| | - Gil Lobaton
- Department of Physiology and Functional, University of Florida, Gainesville, FL, U.S.A
| | - Koji Hosaka
- Department of Neurosurgery, University of Florida, Gainesville, FL, U.S.A
| | - Mohammed Mohammed
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, U.S.A
| | - Eileen M Handberg
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, U.S.A
| | - Elaine M Richards
- Department of Physiology and Functional, University of Florida, Gainesville, FL, U.S.A
| | - Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, U.S.A.
| | - Mohan K Raizada
- Department of Physiology and Functional, University of Florida, Gainesville, FL, U.S.A.
| |
Collapse
|
46
|
Hasan S, Kulkarni NN, Asbjarnarson A, Linhartova I, Osicka R, Sebo P, Gudmundsson GH. Bordetella pertussis Adenylate Cyclase Toxin Disrupts Functional Integrity of Bronchial Epithelial Layers. Infect Immun 2018; 86:e00445-17. [PMID: 29203545 PMCID: PMC5820963 DOI: 10.1128/iai.00445-17] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022] Open
Abstract
The airway epithelium restricts the penetration of inhaled pathogens into the underlying tissue and plays a crucial role in the innate immune defense against respiratory infections. The whooping cough agent, Bordetella pertussis, adheres to ciliated cells of the human airway epithelium and subverts its defense functions through the action of secreted toxins and other virulence factors. We examined the impact of B. pertussis infection and of adenylate cyclase toxin-hemolysin (CyaA) action on the functional integrity of human bronchial epithelial cells cultured at the air-liquid interface (ALI). B. pertussis adhesion to the apical surface of polarized pseudostratified VA10 cell layers provoked a disruption of tight junctions and caused a drop in transepithelial electrical resistance (TEER). The reduction of TEER depended on the capacity of the secreted CyaA toxin to elicit cAMP signaling in epithelial cells through its adenylyl cyclase enzyme activity. Both purified CyaA and cAMP-signaling drugs triggered a decrease in the TEER of VA10 cell layers. Toxin-produced cAMP signaling caused actin cytoskeleton rearrangement and induced mucin 5AC production and interleukin-6 (IL-6) secretion, while it inhibited the IL-17A-induced secretion of the IL-8 chemokine and of the antimicrobial peptide beta-defensin 2. These results indicate that CyaA toxin activity compromises the barrier and innate immune functions of Bordetella-infected airway epithelia.
Collapse
Affiliation(s)
- Shakir Hasan
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | | | | | - Irena Linhartova
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | - Radim Osicka
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | - Peter Sebo
- Institute of Microbiology of the CAS, v.v.i., Prague, Czech Republic
| | | |
Collapse
|
47
|
Cai W, Ran Y, Li Y, Wang B, Zhou L. Intestinal microbiome and permeability in patients with autoimmune hepatitis. Best Pract Res Clin Gastroenterol 2017; 31:669-673. [PMID: 29566910 DOI: 10.1016/j.bpg.2017.09.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/16/2017] [Indexed: 01/31/2023]
Abstract
Autoimmune hepatitis (AIH) is a severe inflammatory liver disease. The underlying mechanisms remain unclear, but recent studies provided new perspectives on altered intestinal microbiome and permeability in AIH animal models and patients, highlighting gut-liver crosstalk in the pathogenesis of AIH. Transgenic AIH mice carrying HLA-DR3 showed reduced diversity and total load of gut microbiota. Germ-free mice are resistant to concanavalin A-induced liver injury, whereas enterogenouss antigens induce the activation of natural killer T cells participating in concanavalin A-induced liver injury, supporting the close relationship between microbiota and AIH. Moreover, 'molecular mimicry' provides a plausible interpretation of the immune reactions between microorganic antigens and liver autoantigens, for instance, cytochrome P4502D6, the target of cross-reactivity between virus and self. Nevertheless, direct evidence for the intestinal microbiome and permeability in AIH is still limited. The relationship between AIH susceptibilities and an intestinal microbiome shaped by drugs, diets or genes needs further study.
Collapse
Affiliation(s)
- Wangfeng Cai
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, PR China
| | - Ying Ran
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, PR China
| | - Yanni Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, PR China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, PR China.
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, PR China.
| |
Collapse
|
48
|
Van Cleemput J, Poelaert KCK, Laval K, Maes R, Hussey GS, Van den Broeck W, Nauwynck HJ. Access to a main alphaherpesvirus receptor, located basolaterally in the respiratory epithelium, is masked by intercellular junctions. Sci Rep 2017; 7:16656. [PMID: 29192251 PMCID: PMC5709510 DOI: 10.1038/s41598-017-16804-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/16/2017] [Indexed: 11/17/2022] Open
Abstract
The respiratory epithelium of humans and animals is frequently exposed to alphaherpesviruses, originating from either external exposure or reactivation from latency. To date, the polarity of alphaherpesvirus infection in the respiratory epithelium and the role of respiratory epithelial integrity herein has not been studied. Equine herpesvirus type 1 (EHV1), a well-known member of the alphaherpesvirus family, was used to infect equine respiratory mucosal explants and primary equine respiratory epithelial cells (EREC), grown at the air-liquid interface. EHV1 binding to and infection of mucosal explants was greatly enhanced upon destruction of the respiratory epithelium integrity with EGTA or N-acetylcysteine. EHV1 preferentially bound to and entered EREC at basolateral cell surfaces. Restriction of infection via apical inoculation was overcome by disruption of intercellular junctions. Finally, basolateral but not apical EHV1 infection of EREC was dependent on cellular N-linked glycans. Overall, our findings demonstrate that integrity of the respiratory epithelium is crucial in the host’s innate defence against primary alphaherpesvirus infections. In addition, by targeting a basolaterally located receptor in the respiratory epithelium, alphaherpesviruses have generated a strategy to efficiently escape from host defence mechanisms during reactivation from latency.
Collapse
Affiliation(s)
- Jolien Van Cleemput
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Katrien C K Poelaert
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Kathlyn Laval
- Department of Molecular Biology and Princeton Neuroscience Institute, Princeton University, 119 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, 08544, USA
| | - Roger Maes
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, 784 Wilson Road, East Lansing, Michigan, 48824, USA
| | - Gisela S Hussey
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, 784 Wilson Road, East Lansing, Michigan, 48824, USA
| | - Wim Van den Broeck
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Hans J Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium.
| |
Collapse
|
49
|
Horn J, Stelzner K, Rudel T, Fraunholz M. Inside job: Staphylococcus aureus host-pathogen interactions. Int J Med Microbiol 2017; 308:607-624. [PMID: 29217333 DOI: 10.1016/j.ijmm.2017.11.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a notorious opportunistic pathogen causing a plethora of diseases. Recent research established that once phagocytosed by neutrophils and macrophages, a certain percentage of S. aureus is able to survive within these phagocytes which thereby even may contribute to dissemination of the pathogen. S. aureus further induces its uptake by otherwise non-phagocytic cells and the ensuing intracellular cytotoxicity is suggested to lead to tissue destruction, whereas bacterial persistence within cells is thought to lead to immune evasion and chronicity of infections. We here review recent work on the S. aureus host pathogen interactions with a focus on the intracellular survival of the pathogen.
Collapse
Affiliation(s)
- Jessica Horn
- Chair of Microbiology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Kathrin Stelzner
- Chair of Microbiology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Thomas Rudel
- Chair of Microbiology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Martin Fraunholz
- Chair of Microbiology, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|
50
|
Seike S, Takehara M, Takagishi T, Miyamoto K, Kobayashi K, Nagahama M. Delta-toxin from Clostridium perfringens perturbs intestinal epithelial barrier function in Caco-2 cell monolayers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:428-433. [PMID: 28988777 DOI: 10.1016/j.bbamem.2017.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/26/2017] [Accepted: 10/03/2017] [Indexed: 01/02/2023]
Abstract
Clostridium perfringens delta-toxin is a β-barrel-pore-forming toxin (β-PFT) and a presumptive virulence factor of type B and C strains, which are causative organisms of fatal intestinal diseases in animals. We showed previously that delta-toxin causes cytotoxicity via necrosis in sensitive cells. Here, we examined the effect of delta-toxin on intestinal membrane integrity. Delta-toxin led to a reduction in transepithelial electrical resistance (TEER) and increased the permeability of fluorescence isothiocyanate-conjugated dextran in human intestinal epithelial Caco-2 cells without changing the tight junction proteins, such as zonula occludens-1 (ZO-1), occludin, and claudin-1. On the other hand, delta-toxin reduced the cellular levels of adherence junction protein E-cadherin before cell injury. A disintegrin and metalloprotease (ADAM) 10 facilitates E-cadherin cleavage and was identified as the cellular receptor for alpha-toxin, a β-PFT produced by Staphylococcus aureus. ADAM10 inhibitor (GI254023X) blocked the toxin-induced decrease in TEER and cleavage of E-cadherin. Delta-toxin enhanced ADAM10 activity in a dose- and time-dependent manner. Furthermore, delta-toxin colocalized with ADAM10. These results indicated that ADAM10 plays a key role in delta-toxin-induced intestinal injury.
Collapse
Affiliation(s)
- Soshi Seike
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Teruhisa Takagishi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Kazuaki Miyamoto
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| |
Collapse
|