1
|
Gopakumar G, Coppo MJC, Diaz-Méndez A, Hartley CA, Devlin JM. Clinical assessment and transcriptome analysis of host immune responses in a vaccination-challenge study using a glycoprotein G deletion mutant vaccine strain of infectious laryngotracheitis virus. Front Immunol 2025; 15:1458218. [PMID: 39926602 PMCID: PMC11802539 DOI: 10.3389/fimmu.2024.1458218] [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: 07/02/2024] [Accepted: 12/17/2024] [Indexed: 02/11/2025] Open
Abstract
A glycoprotein-G-deleted live-attenuated vaccine strain of the infectious laryngotracheitis virus (ILTV), ΔgG-ILTV, is safe and efficacious against ILTV challenge. In the current study, the transcriptome of peripheral blood mononuclear cells (PBMCs) of the ΔgG-ILTV-vaccinated group of specific-pathogen-free chickens were compared to those of the nonvaccinated group at 7 days post-vaccination. Tracheal transcriptomes after challenge with virulent ILTV were compared between groups of the non-vaccinated-challenged and the vaccinated-challenged as well as the non-vaccinated-challenged and the uninfected chickens at 4 to 5 days post-challenge. The clinical outcomes after challenge between these groups were also evaluated. Significant differences were observed in the tracheal transcriptome of the non-vaccinated-challenged birds compared to the other two groups. Enriched gene ontologies and pathways that indicated heightened immune responses and impairments to ciliary and neuronal functions, cell junction components, and potential damages to cartilaginous and extracellular components in the trachea of the non-vaccinated-challenged birds were consistent with their severe tracheal pathology compared to the other two groups. On the contrary, the absence of any difference in the tracheal transcriptome between the vaccinated-challenged and the uninfected birds were reflected by the preservation of tracheal mucosal integrity in both groups and mild infiltration of leukocytes in the vaccinated-challenged birds. The results from this study demonstrated that vaccination with ΔgG-ILTV prevented the changes in tracheal transcriptome induced during ILTV challenge, resulting in clinical protection. Additionally, these results also provide insights into the molecular mechanisms underlying the tracheal pathology induced by ILTV infection.
Collapse
Affiliation(s)
- Gayathri Gopakumar
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Mauricio J. C. Coppo
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
- Escuela de Medicina Veterinaria, Universidad Andrés Bello, Concepción, Chile
| | - Andrés Diaz-Méndez
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Carol A. Hartley
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne M. Devlin
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
2
|
Wang Z, Li L, Li W, Yan H, Yuan Y. Salidroside Alleviates Furan-Induced Impaired Gut Barrier and Inflammation via Gut Microbiota-SCFA-TLR4 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16484-16495. [PMID: 38990698 DOI: 10.1021/acs.jafc.4c02433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
As a food contaminant that can be quickly absorbed through the gastrointestinal system, furan has been shown to disrupt the intestinal flora and barrier. Investigation of the intestinal toxicity mechanism of furan is of great significance to health. We previously identified the regulatory impact of salidroside (SAL) against furan-provoked intestinal damage, and the present work further explored whether the alleviating effect of SAL against furan-caused intestinal injury was based on the intestinal flora; three models, normal, pseudo-germ-free, and fecal microbiota transplantation (FMT), were established, and the changes in intestinal morphology, barrier, and inflammation were observed. Moreover, 16S rDNA sequencing observed the variation of the fecal flora associated with inflammation and short-chain fatty acids (SCFAs). Results obtained from the LC-MS/MS suggested that SAL increased furan-inhibited SCFA levels, activated the mRNA expressions of SCFA receptors (GPR41, GPR43, and GPR109A), and inhibited the furan-activated TLR4/MyD88/NF-κB signaling. Analysis of protein-protein interaction further confirmed the aforementioned effects of SAL, which inhibited furan-induced barrier damage and intestinal inflammation.
Collapse
Affiliation(s)
- Ziyue Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Lu Li
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Wenliang Li
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiyang Yan
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yuan Yuan
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| |
Collapse
|
3
|
Mantzios T, Kiousi DE, Brellou GD, Papadopoulos GA, Economou V, Vasilogianni M, Kanari E, Petridou E, Giannenas I, Tellez-Isaias G, Pappa A, Galanis A, Tsiouris V. Investigation of Potential Gut Health Biomarkers in Broiler Chicks Challenged by Campylobacter jejuni and Submitted to a Continuous Water Disinfection Program. Pathogens 2024; 13:356. [PMID: 38787208 PMCID: PMC11124259 DOI: 10.3390/pathogens13050356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
The exploration of novel biomarkers to assess poultry health is of paramount importance, not only to enhance our understanding of the pathogenicity of zoonotic agents but also to evaluate the efficacy of novel treatments as alternatives to antibiotics. The present study aimed to investigate potential gut health biomarkers in broiler chicks challenged by Campylobacter jejuni and subjected to a continuous water disinfection program. A total of 144 one-day-old hatched broiler chicks were randomly allocated to four treatment groups with four replicates each, according to the following experimental design: Group A received untreated drinking water; Group B received drinking water treated with 0.01-0.05% v/v Cid 2000™ (hydrogen peroxide, acetic acid and paracetic acid); Group C was challenged by C. jejuni and received untreated drinking water; and Group D was challenged by C. jejuni and received drinking water treated with 0.01-0.05% v/v Cid 2000™. The use of Cid 2000™ started on day 1 and was applied in intervals until the end of the experiment at 36 days, while the C. jejuni challenge was applied on day 18. Potential biomarkers were investigated in serum, feces, intestinal tissue, intestinal content, and liver samples of broilers. Statistical analysis revealed significant increases (p < 0.001) in serum cortisol levels in C. jejuni-challenged broilers. Serum fluorescein isothiocyanate dextran (FITC-d) increased significantly (p = 0.004) in broilers challenged by C. jejuni and treated with drinking water disinfectant, while fecal ovotransferrin concentration also increased significantly (p < 0.001) in broilers that received the drinking water disinfectant alone. The gene expression levels of occludin (p = 0.003) and mucin-2 (p < 0.001) were significantly upregulated in broilers challenged by C. jejuni, while mucin-2 significantly increased in birds that were challenged and received the drinking water disinfectant (p < 0.001). TLR-4 expression levels were significantly (p = 0.013) decreased in both groups that received the drinking water disinfectant, compared to the negative control group. Finally, the C. jejuni challenge significantly increased (p = 0.032) the crypt depth and decreased (p = 0.021) the villus height-to-crypt-depth ratio in the ileum of birds, while the tested disinfectant product increased (p = 0.033) the villus height in the jejunum of birds. Furthermore, the counts of C. jejuni in the ceca of birds (p = 0.01), as well as its translocation rate to the liver of broilers (p = 0.001), were significantly reduced by the addition of the water disinfectant. This research contributes to novel insights into the intricate interplay of water disinfection and/or C. jejuni challenge with potential intestinal biomarkers. In addition, it emphasizes the need for continued research to unveil the underlying mechanisms, expands our understanding of broiler responses to these challenges and identifies breakpoints for further investigations.
Collapse
Affiliation(s)
- Tilemachos Mantzios
- Unit of Avian Medicine, Clinic of Farm Animals, School of Veterinary Medicine, Aristotle University of Thessaloniki, 546 27 Thessaloniki, Greece;
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68 100 Alexandroupolis, Greece; (D.E.K.); (E.K.); (A.P.); (A.G.)
| | - Despoina E. Kiousi
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68 100 Alexandroupolis, Greece; (D.E.K.); (E.K.); (A.P.); (A.G.)
| | - Georgia D. Brellou
- Laboratory of Pathology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 546 27 Thessaloniki, Greece
| | - Georgios A. Papadopoulos
- Laboratory of Animal Husbandry, School of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Vangelis Economou
- Laboratory of Hygiene of Animal Food Products—Veterinary Public Health, School of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Marili Vasilogianni
- Pathobiology and Population Sciences, Royal Veterinary College, London NW1 0TU, UK;
| | - Elisavet Kanari
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68 100 Alexandroupolis, Greece; (D.E.K.); (E.K.); (A.P.); (A.G.)
| | - Evanthia Petridou
- Laboratory of Microbiology and Infectious Diseases, School of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Ilias Giannenas
- Laboratory of Nutrition, School of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | | | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68 100 Alexandroupolis, Greece; (D.E.K.); (E.K.); (A.P.); (A.G.)
| | - Alex Galanis
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68 100 Alexandroupolis, Greece; (D.E.K.); (E.K.); (A.P.); (A.G.)
| | - Vasilios Tsiouris
- Unit of Avian Medicine, Clinic of Farm Animals, School of Veterinary Medicine, Aristotle University of Thessaloniki, 546 27 Thessaloniki, Greece;
| |
Collapse
|
4
|
Smith CB, Gao A, Bravo P, Alam A. Microbial Metabolite Trimethylamine N-Oxide Promotes Campylobacter jejuni Infection by Escalating Intestinal Inflammation, Epithelial Damage, and Barrier Disruption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588895. [PMID: 38645062 PMCID: PMC11030326 DOI: 10.1101/2024.04.10.588895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The interactions between Campylobacter jejuni , a critical foodborne cause of gastroenteritis, and the intestinal microbiota during infection are not completely understood. The crosstalk between C. jejuni and its host is impacted by the gut microbiota through mechanisms of competitive exclusion, microbial metabolites, or immune response. To investigate the role of gut microbiota on C. jejuni pathogenesis, we examined campylobacteriosis in the IL10KO mouse model, which was characterized by an increase in the relative abundance of intestinal proteobacteria, E. coli , and inflammatory cytokines during C. jejuni infection. We also found a significantly increased abundance of microbial metabolite Trimethylamine N-Oxide (TMAO) in the colonic lumens of IL10KO mice. We further investigated the effects of TMAO on C. jejuni pathogenesis. We determined that C. jejuni senses TMAO as a chemoattractant and the administration of TMAO promotes C. jejuni invasion into Caco-2 monolayers. TMAO also increased the transmigration of C. jejuni across polarized monolayers of Caco-2 cells, decreased TEER, and increased C. jejuni -mediated intestinal barrier damage. Interestingly, TMAO treatment and presence during C. jejuni infection of Caco-2 cells synergistically caused an increased inflammatory cytokine expression, specifically IL-1β and IL-8. These results establish that C. jejuni utilizes microbial metabolite TMAO for increased virulence during infection.
Collapse
|
5
|
Tsang YP, Hao T, Mao Q, Kelly EJ, Unadkat JD. Dysregulation of the mRNA Expression of Human Renal Drug Transporters by Proinflammatory Cytokines in Primary Human Proximal Tubular Epithelial Cells. Pharmaceutics 2024; 16:285. [PMID: 38399338 PMCID: PMC10893102 DOI: 10.3390/pharmaceutics16020285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Proinflammatory cytokines, which are elevated during inflammation or infections, can affect drug pharmacokinetics (PK) due to the altered expression or activity of drug transporters and/or metabolizing enzymes. To date, such studies have focused on the effect of cytokines on the activity and/or mRNA expression of hepatic transporters and drug-metabolizing enzymes. However, many antibiotics and antivirals used to treat infections are cleared by renal transporters, including the basal organic cation transporter 2 (OCT2), organic anion transporters 1 and 3 (OAT1 and 3), the apical multidrug and toxin extrusion proteins 1 and 2-K (MATE1/2-K), and multidrug resistance-associated protein 2 and 4 (MRP2/4). Here, we determined the concentration-dependent effect of interleukin-6 (IL-6), IL-1β, tumor necrosis factor (TNF)-α, and interferon-γ (IFN-γ) on the mRNA expression of human renal transporters in freshly isolated primary human renal proximal tubular epithelial cells (PTECs, n = 3-5). PTECs were exposed to either a cocktail of cytokines, each at 0.01, 0.1, 1, or 10 ng/mL or individually at the same concentrations. Exposure to the cytokine cocktail for 48 h was found to significantly downregulate the mRNA expression, in a concentration-dependent manner, of OCT2, the organic anion transporting polypeptides 4C1 (OATP4C1), OAT4, MATE2-K, P-glycoprotein (P-gp), and MRP2 and upregulate the mRNA expression of the organic cation/carnitine transporter 1 (OCTN1) and MRP3. OAT1 and OAT3 also appeared to be significantly downregulated but only at 0.1 and 10 ng/mL, respectively, without a clear concentration-dependent trend. Among the cytokines, IL-1β appeared to be the most potent at down- and upregulating the mRNA expression of the transporters. Taken together, our results demonstrate for the first time that proinflammatory cytokines transcriptionally dysregulate renal drug transporters in PTECs. Such dysregulation could potentially translate into changes in transporter protein abundance or activity and alter renal transporter-mediated drug PK during inflammation or infections.
Collapse
Affiliation(s)
| | | | | | | | - Jashvant D. Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (Y.P.T.); (T.H.); (E.J.K.)
| |
Collapse
|
6
|
Wang M, Shi J, Yu C, Zhang X, Xu G, Xu Z, Ma Y. Emerging strategy towards mucosal healing in inflammatory bowel disease: what the future holds? Front Immunol 2023; 14:1298186. [PMID: 38155971 PMCID: PMC10752988 DOI: 10.3389/fimmu.2023.1298186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
For decades, the therapeutic goal of conventional treatment among inflammatory bowel disease (IBD) patients is alleviating exacerbations in acute phase, maintaining remission, reducing recurrence, preventing complications, and increasing quality of life. However, the persistent mucosal/submucosal inflammation tends to cause irreversible changes in the intestinal structure, which can barely be redressed by conventional treatment. In the late 1990s, monoclonal biologics, mainly anti-TNF (tumor necrosis factor) drugs, were proven significantly helpful in inhibiting mucosal inflammation and improving prognosis in clinical trials. Meanwhile, mucosal healing (MH), as a key endoscopic and histological measurement closely associated with the severity of symptoms, has been proposed as primary outcome measures. With deeper comprehension of the mucosal microenvironment, stem cell niche, and underlying mucosal repair mechanisms, diverse potential strategies apart from monoclonal antibodies have been arising or undergoing clinical trials. Herein, we elucidate key steps or targets during the course of MH and review some promising treatment strategies capable of promoting MH in IBD.
Collapse
Affiliation(s)
- Min Wang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jingyan Shi
- Medical School, Nanjing University, Nanjing, China
| | - Chao Yu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xinyi Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Gaoxin Xu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ziyan Xu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Ma
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Zhao H, Zhang Y, Hou L, Lu H, Zhang Y, Xing M. Effects of environmentally relevant cypermethrin and sulfamethoxazole on intestinal health, microbiome, and liver metabolism in grass carp. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 265:106760. [PMID: 37977013 DOI: 10.1016/j.aquatox.2023.106760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
The incorrect use of antibiotics and pesticides poses significant risks of biological toxicity. Their simultaneous exposure could jeopardize fish health and hinder sustainable aquaculture. Here, we subjected grass carp to waterborne cypermethrin (0.65 μg/L) or/and sulfamethoxazole (0.30 μg/L) treatments for a duration of 6 weeks. We closely monitored the effects on intestinal function, the intestinal microbiome, and the liver metabolome. The results revealed that exposure to waterborne cypermethrin or/and sulfamethoxazole compromised intestinal barrier function and decreased the expression of intestinal tight junction proteins. Additionally, heightened levels of pro-inflammatory cytokines in the intestines and reduced antioxidant levels indicated systemic inflammation and oxidative stress, with more severe effects observed in the combined exposure group. 16S rRNA sequencing of intestinal tissues suggested Firmicutes play a key role in the intestinal microbiota. GC/MS metabolomics of the liver showed more differential metabolites (56) in the co-exposure group compared to cypermethrin (45) or sulfamethoxazole (32) alone, indicating greater toxicological effects with combined exposure. Our analyses also suggest that ATP-binding cassette transporters could serve as a novel endpoint for assessing the risk of pesticide and antibiotic mixtures in grass carp. In summary, this study underscores the potential ecological risks posed by antibiotics and pesticides to aquatic environments and products. It emphasizes the importance of the gut-liver axis as a comprehensive pathway for assessing the toxicity in fish exposed to environmental contaminants.
Collapse
Affiliation(s)
- Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Yue Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Lulu Hou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Hongmin Lu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Yingxue Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| |
Collapse
|
8
|
Gorgojo JP, Carrica MDC, Baroli CM, Valdez HA, Alvarez Hayes J, Rodriguez ME. Adenylate cyclase toxin of Bordetella parapertussis disrupts the epithelial barrier granting the bacterial access to the intracellular space of epithelial cells. PLoS One 2023; 18:e0291331. [PMID: 38011105 PMCID: PMC10681170 DOI: 10.1371/journal.pone.0291331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/27/2023] [Indexed: 11/29/2023] Open
Abstract
B. parapertussis is one of the etiological agents of whooping cough. Once inhaled, the bacteria bind to the respiratory epithelium and start the infection. Little is known about this first step of host colonization and the role of the human airway epithelial barrier on B. parapertussis infection. We here investigated the outcome of the interaction of B. parapertussis with a polarized monolayer of respiratory epithelial cells. Our results show that B. parapertussis preferentially attaches to the intercellular boundaries, and causes the disruption of the tight junction integrity through the action of adenylate cyclase toxin (CyaA). We further found evidence indicating that this disruption enables the bacterial access to components of the basolateral membrane of epithelial cells to which B. parapertussis efficiently attaches and gains access to the intracellular location, where it can survive and eventually spread back into the extracellular environment. Altogether, these results suggest that the adenylate cyclase toxin enables B. parapertussis to overcome the epithelial barrier and eventually establish a niche of persistence within the respiratory epithelial cells.
Collapse
Affiliation(s)
- Juan Pablo Gorgojo
- CINDEFI (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Mariela del Carmen Carrica
- CINDEFI (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Carlos Manuel Baroli
- CINDEFI (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Hugo Alberto Valdez
- CINDEFI (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Jimena Alvarez Hayes
- CINDEFI (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Maria Eugenia Rodriguez
- CINDEFI (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
9
|
Fan Y, Li P, Zhu D, Zhao C, Jiao J, Ji X, Du X. Effects of ESA_00986 Gene on Adhesion/Invasion and Virulence of Cronobacter sakazakii and Its Molecular Mechanism. Foods 2023; 12:2572. [PMID: 37444309 DOI: 10.3390/foods12132572] [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: 05/31/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Cronobacter sakazakii is an opportunistic Gram-negative pathogen that has been identified as a causative agent of severe foodborne infections with a higher risk of mortality in neonates, premature infants, the elderly, and immunocompromised populations. The specific pathogenesis mechanisms of C. sakazakii, such as adhesion and colonization, remain unclear. Previously, we conducted comparative proteomic studies on the two strains with the stronger and weaker infection ability, respectively, and found an interesting protein, ESA_00986, which was more highly expressed in the strain with the stronger ability. This unknown protein, predicted to be a type of invasitin related to invasion, may be a critical factor contributing to its virulence. This study aimed to elucidate the precise roles of the ESA_00986 gene in C. sakazakii by generating gene knockout mutants and complementary strains. The mutant and complementary strains were assessed for their biofilm formation, mobility, cell adhesion and invasion, and virulence in a rat model. Compared with the wild-type strain, the mutant strain exhibited a decrease in motility, whereas the complementary strain showed comparable motility to the wild-type. The biofilm-forming ability of the mutant was weakened, and the mutant also exhibited attenuated adhesion to/invasion of intestinal epithelial cells (HCT-8, HICE-6) and virulence in a rat model. This indicated that ESA_00986 plays a positive role in adhesion/invasion and virulence. This study proves that the ESA_00986 gene encodes a novel virulence factor and advances our understanding of the pathogenic mechanism of C. sakazakii.
Collapse
Affiliation(s)
- Yufei Fan
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Ping Li
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Dongdong Zhu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Chumin Zhao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jingbo Jiao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xuemeng Ji
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xinjun Du
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
10
|
Hossain MI, Nasrin S, Das R, Palit P, Sultana AA, Sobi RA, Khan SH, Dash S, Chisti MJ, Ahmed T, Faruque ASG. Symptomatic and Asymptomatic Campylobacter Infections and Child Growth in South Asia: Analyzing Data from the Global Enteric Multicenter Study. Am J Trop Med Hyg 2023; 108:1204-1211. [PMID: 37127268 PMCID: PMC10540126 DOI: 10.4269/ajtmh.22-0347] [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: 05/22/2022] [Accepted: 03/14/2023] [Indexed: 05/03/2023] Open
Abstract
Campylobacter is a major cause of food-borne gastrointestinal illnesses worldwide, predominantly affecting children under 5 years of age. This study examined potential associations of symptomatic (with diarrhea) and asymptomatic (without diarrhea) Campylobacter infections with child growth among children under 5 years of age in South Asia. The Global Enteric Multicenter Study was conducted from 2007 to 2011 with a case-control design. Children were followed for 60 days after enrollment. Stool culture was performed to isolate Campylobacter spp. Among the 22,567 enrolled children, 9,439 were symptomatic, with 786 (8.28%) testing positive for Campylobacter. Conversely, 13,128 asymptomatic healthy controls were included, with 1,057 (8.05%) testing positive for Campylobacter. Growth faltering was observed in the symptomatic group, particularly among children aged 0-11 months (-0.19 height-for-age z score [HAZ]; 95% CI: -0.36, -0.03; P = 0.018) and 24-59 months (-0.16 HAZ; 95% CI: -0.28, -0.04; P = 0.010). However, in the asymptomatic group, growth faltering was observed only in the 24- to 59-month age group, in terms of HAZ (-0.15 HAZ; 95% CI: -0.24, -0.05; P = 0.002) and weight-for-height z score (-0.16; 95% CI: -0.26, -0.06; P = 0.001). These findings underscore the importance of immediate and enhanced introduction of preventive modalities to reduce the burden of Campylobacter infections and reduce their long-term sequelae.
Collapse
Affiliation(s)
- Md Iqbal Hossain
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | - Sabiha Nasrin
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Rina Das
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
- Gangarosa Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Parag Palit
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
- University of Virginia School of Medicine, Charlottesville, Virginia
| | - Al-Afroza Sultana
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Rukaeya Amin Sobi
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Soroar Hossain Khan
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sampa Dash
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mohammod Jobayer Chisti
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, Washington
| | - Abu Syed Golam Faruque
- Nutrition and Clinical Services Division, International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
11
|
Rogers AP, Mileto SJ, Lyras D. Impact of enteric bacterial infections at and beyond the epithelial barrier. Nat Rev Microbiol 2023; 21:260-274. [PMID: 36175770 DOI: 10.1038/s41579-022-00794-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 11/09/2022]
Abstract
The mucosal lining of the gut has co-evolved with a diverse microbiota over millions of years, leading to the development of specialized mechanisms to actively limit the invasion of pathogens. However, some enteric microorganisms have adapted against these measures, developing ways to hijack or overcome epithelial micro-integrity mechanisms. This breach of the gut barrier not only enables the leakage of host factors out of circulation but can also initiate a cascade of detrimental systemic events as microbiota, pathogens and their affiliated secretions passively leak into extra-intestinal sites. Under normal circumstances, gut damage is rapidly repaired by intestinal stem cells. However, with substantial and deep perturbation to the gut lining and the systemic dissemination of gut contents, we now know that some enteric infections can cause the impairment of host regenerative processes. Although these local and systemic aspects of enteric disease are often studied in isolation, they heavily impact one another. In this Review, by examining the journey of enteric infections from initial establishment to systemic sequelae and how, or if, the host can successfully repair damage, we will tie together these complex interactions to provide a holistic overview of the impact of enteric infections at and beyond the epithelial barrier.
Collapse
Affiliation(s)
- Ashleigh P Rogers
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.,Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Steven J Mileto
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.,Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia. .,Department of Microbiology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
12
|
Otte ML, Lama Tamang R, Papapanagiotou J, Ahmad R, Dhawan P, Singh AB. Mucosal healing and inflammatory bowel disease: Therapeutic implications and new targets. World J Gastroenterol 2023; 29:1157-1172. [PMID: 36926666 PMCID: PMC10011951 DOI: 10.3748/wjg.v29.i7.1157] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/16/2022] [Accepted: 02/14/2023] [Indexed: 02/21/2023] Open
Abstract
Mucosal healing (MH) is vital in maintaining homeostasis within the gut and protecting against injury and infections. Multiple factors and signaling pathways contribute in a dynamic and coordinated manner to maintain intestinal homeostasis and mucosal regeneration/repair. However, when intestinal homeostasis becomes chronically disturbed and an inflammatory immune response is constitutively active due to impairment of the intestinal epithelial barrier autoimmune disease results, particularly inflammatory bowel disease (IBD). Many proteins and signaling pathways become dysregulated or impaired during these pathological conditions, with the mechanisms of regulation just beginning to be understood. Consequently, there remains a relative lack of broadly effective therapeutics that can restore MH due to the complexity of both the disease and healing processes, so tissue damage in the gastrointestinal tract of patients, even those in clinical remission, persists. With increased understanding of the molecular mechanisms of IBD and MH, tissue damage from autoimmune disease may in the future be ameliorated by developing therapeutics that enhance the body’s own healing response. In this review, we introduce the concept of mucosal healing and its relevance in IBD as well as discuss the mechanisms of IBD and potential strategies for altering these processes and inducing MH.
Collapse
Affiliation(s)
- Megan Lynn Otte
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Raju Lama Tamang
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Julia Papapanagiotou
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
13
|
Montanari M, Guescini M, Gundogdu O, Luchetti F, Lanuti P, Ciacci C, Burattini S, Campana R, Ortolani C, Papa S, Canonico B. Extracellular Vesicles from Campylobacter jejuni CDT-Treated Caco-2 Cells Inhibit Proliferation of Tumour Intestinal Caco-2 Cells and Myeloid U937 Cells: Detailing the Global Cell Response for Potential Application in Anti-Tumour Strategies. Int J Mol Sci 2022; 24:ijms24010487. [PMID: 36613943 PMCID: PMC9820799 DOI: 10.3390/ijms24010487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Cytolethal distending toxin (CDT) is produced by a range of Gram-negative pathogenic bacteria such as Campylobacter jejuni. CDT represents an important virulence factor that is a heterotrimeric complex composed of CdtA, CdtB, and CdtC. CdtA and CdtC constitute regulatory subunits whilst CdtB acts as the catalytic subunit exhibiting phosphatase and DNase activities, resulting in cell cycle arrest and cell death. Extracellular vesicle (EV) secretion is an evolutionarily conserved process that is present throughout all kingdoms. Mammalian EVs play important roles in regular cell-to-cell communications but can also spread pathogen- and host-derived molecules during infections to alter immune responses. Here, we demonstrate that CDT targets the endo-lysosomal compartment, partially evading lysosomal degradation and exploiting unconventional secretion (EV release), which is largely involved in bacterial infections. CDT-like effects are transferred by Caco-2 cells to uninfected heterologous U937 and homologous Caco-2 cells. The journey of EVs derived from CDT-treated Caco-2 cells is associated with both intestinal and myeloid tumour cells. EV release represents the primary route of CDT dissemination, revealing an active toxin as part of the cargo. We demonstrated that bacterial toxins could represent suitable tools in cancer therapy, highlighting both the benefits and limitations. The global cell response involves a moderate induction of apoptosis and autophagic features may play a protective role against toxin-induced cell death. EVs from CDT-treated Caco-2 cells represent reliable CDT carriers, potentially suitable in colorectal cancer treatments. Our data present a potential bacterial-related biotherapeutic supporting a multidrug anticancer protocol.
Collapse
Affiliation(s)
- Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Caterina Ciacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Raffaella Campana
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Claudio Ortolani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence:
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| |
Collapse
|
14
|
Cheng C, Yin Y, Bian G. Effects of whole maize high-grain diet feeding on colonic fermentation and bacterial community in weaned lambs. Front Microbiol 2022; 13:1018284. [PMID: 36569065 PMCID: PMC9772272 DOI: 10.3389/fmicb.2022.1018284] [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: 08/13/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
High-grain diet is commonly used in intensive production to boost yield in short term, which may cause adverse effects such as rumen and colonic acidosis in ruminants. Maize is one of the key components of high-grain diet, and different processing methods of maize affect the digestive absorption and gastrointestinal development of ruminants. To investigate the effects of maize form in high-grain diets on colonic fermentation and bacterial community of weaned lambs, twenty-two 2.5-month-old healthy Hu lambs were fed separately a maize meal low-grain diet (19.2% grain; CON), a maize meal high-grain diet (50.4% grain; CM), and a whole maize high-grain diet (50.4% grain; CG). After 7 weeks of feeding, the total volatile fatty acid concentration (P = 0.035) were significantly higher in lambs from CM than that from CON. The sequencing results of colonic content microbial composition revealed that the relative abundance of genera Parasutterella (P = 0.028), Comamonas (P = 0.031), Butyricicoccus (P = 0.049), and Olsenella (P = 0.010) were higher in CM than those in CON; compared with CM, the CG diet had the higher relative abundance of genera Bacteroides (P = 0.024) and Angelakisella (P = 0.020), while the lower relative abundance of genera Olsenella (P = 0.031) and Paraprevotella (P = 0.006). For colonic mucosal microbiota, the relative abundance of genera Duncaniella (P = 0.024), Succiniclasticum (P = 0.044), and Comamonas (P = 0.012) were significantly higher in CM than those in CON. In comparison, the relative abundance of genera Alistipes (P = 0.020) and Campylobacter (P = 0.017) were significantly lower. And the relative abundance of genera Colidextribacter (P = 0.005), Duncaniella (P = 0.032), Christensenella (P = 0.042), and Lawsonibacter (P = 0.018) were increased in the CG than those in the CM. Furthermore, the CG downregulated the relative abundance of genes encoding infectious-disease-parasitic (P = 0.049), cancer-specific-types (P = 0.049), and neurodegenerative-disease (P = 0.037) in colonic microbiota than those in the CM. Overall, these results indicated that maize with different grain sizes might influence the colonic health of weaned lambs by altering the composition of the colonic bacterial community.
Collapse
Affiliation(s)
- Chao Cheng
- College of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, China
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuyang Yin
- Huzhou Academy of Agricultural Sciences, Huzhou, China
| | - Gaorui Bian
- College of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, China
| |
Collapse
|
15
|
Carserides C, Smith K, Zinicola M, Kumar A, Swedrowska M, Scala C, Cameron G, Riches Z, Iannelli F, Pozzi G, Hold GL, Forbes B, Kelly C, Hijazi K. Comprehensive Study of Antiretroviral Drug Permeability at the Cervicovaginal Mucosa via an In Vitro Model. Pharmaceutics 2022; 14:pharmaceutics14091938. [PMID: 36145684 PMCID: PMC9504208 DOI: 10.3390/pharmaceutics14091938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Modulation of drug transporter activity at mucosal sites of HIV-1 transmission may be exploited to optimize retention of therapeutic antiretroviral drug concentrations at target submucosal CD4+ T cells. Previously, we showed that darunavir was a substrate for the P-glycoprotein efflux drug transporter in colorectal mucosa. Equivalent studies in the cervicovaginal epithelium have not been reported. Here, we describe the development of a physiologically relevant model to investigate the permeability of antiretroviral drugs across the vaginal epithelium. Barrier properties of the HEC-1A human endometrial epithelial cell line were determined, in a dual chamber model, by measurement of transepithelial electrical resistance, immunofluorescent staining of tight junctions and bi-directional paracellular permeability of mannitol. We then applied this model to investigate the permeability of tenofovir, darunavir and dapivirine. Efflux ratios indicated that the permeability of each drug was transporter-independent in this model. Reduction of pH to physiological levels in the apical compartment increased absorptive transfer of darunavir, an effect that was reversed by inhibition of MRP efflux transport via MK571. Thus, low pH may increase the transfer of darunavir across the epithelial barrier via increased MRP transporter activity. In a previous in vivo study in the macaque model, we demonstrated increased MRP2 expression following intravaginal stimulation with darunavir which may further increase drug uptake. Stimulation with inflammatory modulators had no effect on drug permeability across HEC-1A barrier epithelium but, in the VK2/E6E7 vaginal cell line, increased expression of both efflux and uptake drug transporters which may influence darunavir disposition.
Collapse
Affiliation(s)
- Constandinos Carserides
- Centre for Host Microbiome Interactions, King’s College London, London SE1 9NH, UK
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK
| | - Kieron Smith
- School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, UK
| | - Marta Zinicola
- Centre for Host Microbiome Interactions, King’s College London, London SE1 9NH, UK
| | - Abhinav Kumar
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK
| | - Magda Swedrowska
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK
| | - Carlo Scala
- Centre for Host Microbiome Interactions, King’s College London, London SE1 9NH, UK
| | - Gary Cameron
- School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, UK
| | - Zoe Riches
- School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, UK
| | - Francesco Iannelli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Georgina L. Hold
- School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, UK
| | - Ben Forbes
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK
| | - Charles Kelly
- Centre for Host Microbiome Interactions, King’s College London, London SE1 9NH, UK
| | - Karolin Hijazi
- School of Medicine Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZR, UK
- Correspondence: ; Tel.: +44-(0)-1224-555153
| |
Collapse
|
16
|
von Buchholz JS, Ruhnau D, Hess C, Aschenbach JR, Hess M, Awad WA. Paracellular intestinal permeability of chickens induced by DON and/or C. jejuni is associated with alterations in tight junction mRNA expression. Microb Pathog 2022; 168:105509. [PMID: 35367310 DOI: 10.1016/j.micpath.2022.105509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022]
Abstract
Toxins, antigens, and harmful pathogens continuously challenge the intestinal mucosa. Therefore, regulation of the intestinal barrier is crucial for the maintenance of mucosal homeostasis and gut health. Intercellular complexes, namely, tight junctions (TJs), regulate paracellular permeability. TJs are mainly composed of claudins (CLDN), occludin (OCLN), tight junction associated MARVEL-domain proteins (TAMPS), the scaffolding zonula occludens (ZO) proteins and junction-adhesion molecules (JAMs). Different studies have shown that a Campylobacter infection can lead to a phenomenon so-called "leaky gut", including the translocation of luminal bacteria to the underlying tissue and internal organs. Based on the effects of C. jejuni on the chicken gut, we hypothesize that impacts on TJ proteins play a crucial role in the destructive effects of the intestinal barrier. Likewise, the mycotoxin deoxynivalenol (DON) can also alter gut permeability in chickens. Albeit DON and C. jejuni are widely distributed, no data are available on their effect on the tight junctions' barrier in the broiler intestine and consequences for permeability. Therefore, the aim of this study was to analyze the interaction between DON and C. jejuni on the gut barrier by linking permeability with gene expression of TJ proteins and to determine the relationships between the measurements. Following oral infection of birds with C. jejuni NCTC 12744 at 14 days of age, we demonstrate that the co-exposure with DON has considerable consequences on gut permeability as well as on gut TJ mRNA expression. Co-exposure of DON and C. jejuni enhanced the negative effect on paracellular permeability of the intestine, which was also noticed for the bacteria or the mycotoxin alone by the Ussing chamber technique at certain time points in both jejunum and caecum. Furthermore, the increased paracellular permeability was associated with significant changes in TJ mRNA expression in the small and large intestine. The actual study demonstrates that co-exposure of broiler chickens to DON and C. jejuni resulted in a decreased barrier function via up-regulation of pore-forming tight junctions (CLDN7 and CLDN10), as well as the cytosolic TJ protein occludin (OCLN) that can shift to various paracellular locations and are therefore able to alter the epithelial permeability. These findings indicate that the co-exposure of broiler chickens to DON and C. jejuni affects the paracellular permeability of the gut by altering the tight junction proteins. Furthermore, analysing of correlations between TJs revealed that the mRNA expression levels of most tight junctions were correlated with each other in both jejunum and caecum. Finally, the findings indicate that the molecular composition of tight junctions can be used as a marker for gut health and integrity.
Collapse
Affiliation(s)
- J Sophia von Buchholz
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Daniel Ruhnau
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Claudia Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Jörg R Aschenbach
- Department of Veterinary Medicine, Institute of Veterinary Physiology, Freie Universität Berlin, Berlin, Germany
| | - Michael Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Wageha A Awad
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
17
|
Cosert KM, Kim S, Jalilian I, Chang M, Gates BL, Pinkerton KE, Van Winkle LS, Raghunathan VK, Leonard BC, Thomasy SM. Metallic Engineered Nanomaterials and Ocular Toxicity: A Current Perspective. Pharmaceutics 2022; 14:pharmaceutics14050981. [PMID: 35631569 PMCID: PMC9145553 DOI: 10.3390/pharmaceutics14050981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
The ocular surface, comprised of the transparent cornea, conjunctiva, and protective tear film, forms a protective barrier defending deeper structures of the eye from particulate matter and mechanical trauma. This barrier is routinely exposed to a multitude of naturally occurring and engineered nanomaterials (ENM). Metallic ENMs are particularly ubiquitous in commercial products with a high risk of ocular exposure, such as cosmetics and sunscreens. Additionally, there are several therapeutic uses for metallic ENMs owing to their attractive magnetic, antimicrobial, and functionalization properties. The increasing commercial and therapeutic applications of metallic ENMs come with a high risk of ocular exposure with poorly understood consequences to the health of the eye. While the toxicity of metallic ENMs exposure has been rigorously studied in other tissues and organs, further studies are necessary to understand the potential for adverse effects and inform product usage for individuals whose ocular health may be compromised by injury, disease, or surgical intervention. This review provides an update of current literature on the ocular toxicity of metallic ENMs in vitro and in vivo, as well as the risks and benefits of therapeutic applications of metallic ENMs in ophthalmology.
Collapse
Affiliation(s)
- Krista M. Cosert
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Soohyun Kim
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Iman Jalilian
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Maggie Chang
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Brooke L. Gates
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California Davis, Davis, CA 95616, USA; (K.E.P.); (L.S.V.W.)
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Laura S. Van Winkle
- Center for Health and the Environment, University of California Davis, Davis, CA 95616, USA; (K.E.P.); (L.S.V.W.)
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Vijay Krishna Raghunathan
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX 77004, USA;
- The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX 77004, USA
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
| | - Sara M. Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (K.M.C.); (S.K.); (I.J.); (M.C.); (B.L.G.); (B.C.L.)
- Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-530-752-0926
| |
Collapse
|
18
|
Wang Y, Hong C, Wu Z, Li S, Xia Y, Liang Y, He X, Xiao X, Tang W. Resveratrol in Intestinal Health and Disease: Focusing on Intestinal Barrier. Front Nutr 2022; 9:848400. [PMID: 35369090 PMCID: PMC8966610 DOI: 10.3389/fnut.2022.848400] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
The integrity of intestinal barrier determines intestinal homeostasis, which could be affected by various factors, like physical, chemical, and biological stimuli. Therefore, it is of considerable interest and importance to maintain intestinal barrier function. Fortunately, many plant polyphenols, including resveratrol, could affect the health of intestinal barrier. Resveratrol has many biological functions, such as antioxidant, anti-inflammation, anti-tumor, and anti-cardiovascular diseases. Accumulating studies have shown that resveratrol affects intestinal tight junction, microbial composition, and inflammation. In this review, we summarize the effects of resveratrol on intestinal barriers as well as the potential mechanisms (e.g., inhibiting the growth of pathogenic bacteria and fungi, regulating the expression of tight junction proteins, and increasing anti-inflammatory T cells while reducing pro-inflammatory T cells), and highlight the applications of resveratrol in ameliorating various intestinal diseases.
Collapse
Affiliation(s)
- Youxia Wang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Changming Hong
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zebiao Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shuwei Li
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtech Feed Co., Ltd., Chengdu, China
| | - Yaoyao Xia
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yuying Liang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaohua He
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xinyu Xiao
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtech Feed Co., Ltd., Chengdu, China
- *Correspondence: Wenjie Tang
| |
Collapse
|
19
|
Preventing Bacterial Translocation in Patients with Leaky Gut Syndrome: Nutrition and Pharmacological Treatment Options. Int J Mol Sci 2022; 23:ijms23063204. [PMID: 35328624 PMCID: PMC8949204 DOI: 10.3390/ijms23063204] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
Leaky gut syndrome is a medical condition characterized by intestinal hyperpermeability. Since the intestinal barrier is one of the essential components maintaining homeostasis along the gastrointestinal tract, loss of its integrity due to changes in bacterial composition, decreased expression levels of tight junction proteins, and increased concentration of pro-inflammatory cytokines may lead to intestinal hyperpermeability followed by the development of gastrointestinal and non-gastrointestinal diseases. Translocation of microorganisms and their toxic metabolites beyond the gastrointestinal tract is one of the fallouts of the leaky gut syndrome. The presence of intestinal bacteria in sterile tissues and distant organs may cause damage due to chronic inflammation and progression of disorders, including inflammatory bowel diseases, liver cirrhosis, and acute pancreatitis. Currently, there are no medical guidelines for the treatment or prevention of bacterial translocation in patients with the leaky gut syndrome; however, several studies suggest that dietary intervention can improve barrier function and restrict bacteria invasion. This review contains current literature data concerning the influence of diet, dietary supplements, probiotics, and drugs on intestinal permeability and bacterial translocation.
Collapse
|
20
|
Latek U, Chłopecka M, Karlik W, Mendel M. Phytogenic Compounds for Enhancing Intestinal Barrier Function in Poultry-A Review. PLANTA MEDICA 2022; 88:218-236. [PMID: 34331305 DOI: 10.1055/a-1524-0358] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
After the European Union ban of antibiotic growth promoters, works on different methods of improving gut health have intensified. The poultry industry is struggling with problems that were previously controlled by antibiotic growth promoters, therefore the search for optimal solutions continues. Simultaneously, there is also increasing social pressure to minimize the use of antibiotics and replace them with alternative feed additives. A variety of available alternatives is considered safe by consumers, among which phytogenics play a significant role. However, there are still some limitations that need to be considered. The most questionable are the issues related to bioavailability, metabolism of plant derivatives in birds, and the difficulty of standardizing commercial products. There is still a need for more evidence-based recommendations for the use of phytogenics in livestock. On the other hand, a positive influence of phytogenic compounds on the health of poultry has been previously described by many researchers and practical application of these compounds has auspicious perspectives in poultry production. Supplementation with phytogenic feed additives has been shown to protect birds from various environmental threats leading to impaired intestinal barrier function. Phytogenic feed additives have the potential to improve the overall structure of intestinal mucosa as well as gut barrier function on a molecular level. Recognition of the phytogenics' effect on the components of the intestinal barrier may enable the selection of the most suitable ones to alleviate negative effects of different agents. This review aims to summarize current knowledge of the influence of various phytogenic constituents on the intestinal barrier and health of poultry.
Collapse
Affiliation(s)
- Urszula Latek
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Magdalena Chłopecka
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Wojciech Karlik
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Marta Mendel
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| |
Collapse
|
21
|
Necrotic enteritis in chickens: a review of pathogenesis, immune responses and prevention, focusing on probiotics and vaccination. Anim Health Res Rev 2022; 22:147-162. [DOI: 10.1017/s146625232100013x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AbstractNecrotic enteritis (NE), caused by Clostridium perfringens (CP), is one of the most common of poultry diseases, causing huge economic losses to the poultry industry. This review provides an overview of the pathogenesis of NE in chickens and of the interaction of CP with the host immune system. The roles of management, nutrition, probiotics, and vaccination in reducing the incidence and severity of NE in poultry flocks are also discussed.
Collapse
|
22
|
Helmy YA, Kassem II, Rajashekara G. Immuno-modulatory effect of probiotic E. coli Nissle 1917 in polarized human colonic cells against Campylobacter jejuni infection. Gut Microbes 2022; 13:1-16. [PMID: 33382951 PMCID: PMC7781529 DOI: 10.1080/19490976.2020.1857514] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni is among the leading causes of bacterial foodborne illness. Poultry is the major reservoir and source of human campylobacteriosis. Currently, there is no effective and practical method to decrease C. jejuni colonization in chickens or to reduce human infections. Additionally, antibiotic-resistant infections pose a serious public health concern; therefore, antibiotic-alternative approaches are needed to reduce transmission of C. jejuni including resistant bacteria from chickens to humans. Here, we evaluated the effect of E. coli Nissle 1917 (EcN) on innate responses of polarized HT-29 cells and consequently on C. jejuni 81176 infections in HT-29 cells. Pre-treatment of HT-29 cells with EcN for 4 h had a significant effect on the invasion of different C. jejuni strains (2 h post-infection) (P < .05) and no intracellular C. jejuni (24 h post-infection) were recovered. To further understand how EcN mediates its impact on C. jejuni's survival inside the cells, we used Human Antibacterial RT2 ProfilerTM PCR arrays to profile gene expression in HT-29 cells after treatment with EcN with or without C. jejuni 81-176 infection. Our results suggest that pre-treatment of the HT-29 cells with EcN induced the anti-inflammatory cytokines and activated the anti-apoptotic Akt signaling which likely to protect the cells against the proinflammatory and apoptosis responses induced by C. jejuni. EcN also positively affected the expression of genes involved in cellular maintenance, growth, development, and proliferation. Further, EcN modulated the expression of genes involved in protective innate immunity, such as TLRs, ERK1/2, p38 MAPK, Ap1, JNK, IL1B, IL17A, and NF-κB signaling.
Collapse
Affiliation(s)
- Yosra A. Helmy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA
| | - Issmat I. Kassem
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA,College of Agricultural and Environmental Sciences, Center for Food Safety, University of Georgia, Griffin, Georgia, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA,Corresponding author Gireesh Rajashekara Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH44691, USA
| |
Collapse
|
23
|
Daskou M, Mu W, Sharma M, Vasilopoulos H, Heymans R, Ritou E, Rezek V, Hamid P, Kossyvakis A, Sen Roy S, Grijalva V, Chattopadhyay A, Kitchen SG, Fogelman AM, Reddy ST, Kelesidis T. ApoA-I mimetics reduce systemic and gut inflammation in chronic treated HIV. PLoS Pathog 2022; 18:e1010160. [PMID: 34995311 PMCID: PMC8740974 DOI: 10.1371/journal.ppat.1010160] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022] Open
Abstract
Novel therapeutic strategies are needed to attenuate increased systemic and gut inflammation that contribute to morbidity and mortality in chronic HIV infection despite potent antiretroviral therapy (ART). The goal of this study is to use preclinical models of chronic treated HIV to determine whether the antioxidant and anti-inflammatory apoA-I mimetic peptides 6F and 4F attenuate systemic and gut inflammation in chronic HIV. We used two humanized murine models of HIV infection and gut explants from 10 uninfected and 10 HIV infected persons on potent ART, to determine the in vivo and ex vivo impact of apoA-I mimetics on systemic and intestinal inflammation in HIV. When compared to HIV infected humanized mice treated with ART alone, mice on oral apoA-I mimetic peptide 6F with ART had consistently reduced plasma and gut tissue cytokines (TNF-α, IL-6) and chemokines (CX3CL1) that are products of ADAM17 sheddase activity. Oral 6F attenuated gut protein levels of ADAM17 that were increased in HIV-1 infected mice on potent ART compared to uninfected mice. Adding oxidized lipoproteins and endotoxin (LPS) ex vivo to gut explants from HIV infected persons increased levels of ADAM17 in myeloid and intestinal cells, which increased TNF-α and CX3CL1. Both 4F and 6F attenuated these changes. Our preclinical data suggest that apoA-I mimetic peptides provide a novel therapeutic strategy that can target increased protein levels of ADAM17 and its sheddase activity that contribute to intestinal and systemic inflammation in treated HIV. The large repertoire of inflammatory mediators involved in ADAM17 sheddase activity places it as a pivotal orchestrator of several inflammatory pathways associated with morbidity in chronic treated HIV that make it an attractive therapeutic target.
Collapse
Affiliation(s)
- Maria Daskou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - William Mu
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Madhav Sharma
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hariclea Vasilopoulos
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rachel Heymans
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Eleni Ritou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Valerie Rezek
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Philip Hamid
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Athanasios Kossyvakis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Shubhendu Sen Roy
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Victor Grijalva
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Arnab Chattopadhyay
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Scott G. Kitchen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Alan M. Fogelman
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Srinivasa T. Reddy
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, Los Angeles, California, United States of America
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
24
|
OUP accepted manuscript. Pathog Dis 2022; 80:6521441. [DOI: 10.1093/femspd/ftac003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/30/2021] [Accepted: 02/01/2022] [Indexed: 11/15/2022] Open
|
25
|
Kochar A, Farooq S. Human immune deficiency virus conundrum: An everlasting challenge! Oman J Ophthalmol 2022; 15:13-19. [PMID: 35388268 PMCID: PMC8979399 DOI: 10.4103/ojo.ojo_125_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/08/2021] [Accepted: 07/18/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Human immune deficiency virus (HIV) causes a wide spectrum of diseases worldwide and has the capability to affect every organ system in the body. Ocular manifestations have been reported in up to 70% of individuals infected with HIV and the ocular manifestations reflect systemic disease and maybe the first sign of disseminated infection. PURPOSE To study the pattern of ocular manifestations related to HIV in seropositive patients with the objective of identifying its spectrum in highly active antiretroviral therapy (HAART) era and to emphasize on the role of ophthalmologists in fighting the battle against HIV. MATERIALS AND METHODS A cross sectional study was undertaken on 300 HIV positive patients. Data were collected, tabulated and analyzed using MSTAT software. Chi-square test was applied and P < 0.05 was considered significant. RESULTS Ocular lesions were observed in 50.33% of patients. Conjunctival microvasculopathy being the most common finding seen in 27 (9%) patients, followed by retinal microvasculopathy and trichomegaly in 20 (6.66%) patients each. Herpes zoster ophthalmicus and cytomegalovirus retinitis were observed in four (1.33%) and three (1%) patients respectively. In the study 78.33% patients were on ART. CONCLUSION HAART has reduced serious ocular opportunistic infections and ocular malignancies, but HAART mediated visually disabling immune recovery uveitis has emerged as a clinical challenge for ophthalmologists. Improved communication between the two broad specialties of HIV medicine and ophthalmology will definitely go a long way in the battle against this dreadful disease.
Collapse
Affiliation(s)
- Anju Kochar
- Department of Ophthalmology, SP Medical College, Bikaner, Rajasthan, India
| | - Shaheen Farooq
- Department of Ophthalmology, SP Medical College, Bikaner, Rajasthan, India
| |
Collapse
|
26
|
Theodoros K, Sharma M, Anton P, Hugo C, Ellen O, Hultgren NW, Ritou E, Williams DS, Orian S S, Srinivasa T R. The ApoA-I mimetic peptide 4F attenuates in vitro replication of SARS-CoV-2, associated apoptosis, oxidative stress and inflammation in epithelial cells. Virulence 2021; 12:2214-2227. [PMID: 34494942 PMCID: PMC8437485 DOI: 10.1080/21505594.2021.1964329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/12/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
An oral antiviral against SARS-CoV-2 that also attenuates inflammatory instigators of severe COVID-19 is not available to date. Herein, we show that the apoA-I mimetic peptide 4 F inhibits Spike mediated viral entry and has antiviral activity against SARS-CoV-2 in human lung epithelial Calu3 and Vero-E6 cells. In SARS-CoV-2 infected Calu3 cells, 4 F upregulated inducers of the interferon pathway such as MX-1 and Heme oxygenase 1 (HO-1) and downregulated mitochondrial reactive oxygen species (mito-ROS) and CD147, a host protein that mediates viral entry. 4 F also reduced associated cellular apoptosis and secretion of IL-6 in both SARS-CoV-2 infected Vero-E6 and Calu3 cells. Thus, 4 F attenuates in vitro SARS-CoV-2 replication, associated apoptosis in epithelial cells and secretion of IL-6, a major cytokine related to COVID-19 morbidity. Given established safety of 4 F in humans, clinical studies are warranted to establish 4 F as therapy for COVID-19.
Collapse
Affiliation(s)
- Kelesidis Theodoros
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Madhav Sharma
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Petcherski Anton
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Cristelle Hugo
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - O’Connor Ellen
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
| | - Nan W Hultgren
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Eleni Ritou
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David S Williams
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Shirihai Orian S
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Reddy Srinivasa T
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
27
|
Evidence of MHC class I and II influencing viral and helminth infection via the microbiome in a non-human primate. PLoS Pathog 2021; 17:e1009675. [PMID: 34748618 PMCID: PMC8601626 DOI: 10.1371/journal.ppat.1009675] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/18/2021] [Accepted: 10/05/2021] [Indexed: 01/04/2023] Open
Abstract
Until recently, the study of major histocompability complex (MHC) mediated immunity has focused on the direct link between MHC diversity and susceptibility to parasite infection. However, MHC genes can also influence host health indirectly through the sculpting of the bacterial community that in turn shape immune responses. We investigated the links between MHC class I and II gene diversity gut microbiome diversity and micro- (adenovirus, AdV) and macro- (helminth) parasite infection probabilities in a wild population of non-human primates, mouse lemurs of Madagascar. This setup encompasses a plethora of underlying interactions between parasites, microbes and adaptive immunity in natural populations. Both MHC classes explained shifts in microbiome composition and the effect was driven by a few select microbial taxa. Among them were three taxa (Odoribacter, Campylobacter and Prevotellaceae-UCG-001) which were in turn linked to AdV and helminth infection status, correlative evidence of the indirect effect of the MHC via the microbiome. Our study provides support for the coupled role of MHC diversity and microbial flora as contributing factors of parasite infection. The selective pressure of the major histocompatibility complex (MHC) on microbial communities, and the potential role of this interaction in driving parasite resistance has been largely neglected. Using a natural population of the primate Microcebus griseorufus, we provide correlative evidence of two outstanding findings: that MHCI and MHCII diversity shapes the composition of the gut microbiota; and that select taxa associated with MHC diversity predicted adenovirus and helminth infection status. Our study highlights the importance of incorporating the microbiome when investigating parasite-mediated MHC selection.
Collapse
|
28
|
Rath A, Rautenschlein S, Rzeznitzeck J, Breves G, Hewicker-Trautwein M, Waldmann KH, von Altrock A. Impact of Campylobacter spp. on the Integrity of the Porcine Gut. Animals (Basel) 2021; 11:ani11092742. [PMID: 34573708 PMCID: PMC8467837 DOI: 10.3390/ani11092742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Campylobacter (C.) is the most common food-borne zoonosis in humans, which mainly manifests with watery to bloody diarrhoea. While C. jejuni is responsible for most cases of infection, C. coli is less frequently encountered. The object of the study was to prove the clinical impact of mono- and co-colonisation of C. coli and C. jejuni on weaned piglets in an infection model and to investigate the impact on transepithelial transport processes in the jejunum and caecum. At an age of eight weeks, eight pigs were infected with C. coli (ST-5777), 10 pigs with C. jejuni (ST-122), eight pigs with both strains, and 11 piglets served as control. During the four-week observation period, no clinical signs were observed. During dissection, both strains could be isolated from the jejunum and the caecum, but no alteration of the tissue could be determined histopathologically. Mono-infection with C. jejuni showed an impact on transepithelial ion transport processes of the caecum. An increase in the short circuit current (Isc) was observed in the Ussing chamber resulting from carbachol- and forskolin-mediated Cl- secretion. Therefore, we speculate that caecal colonisation of C. jejuni might affect the transport mechanisms of the intestinal mucosa without detectable inflammatory reaction.
Collapse
Affiliation(s)
- Alexandra Rath
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany; (K.-H.W.); (A.v.A.)
- Correspondence:
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (S.R.); (J.R.)
| | - Janina Rzeznitzeck
- Clinic for Poultry, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (S.R.); (J.R.)
| | - Gerhard Breves
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany;
| | - Marion Hewicker-Trautwein
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany;
| | - Karl-Heinz Waldmann
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany; (K.-H.W.); (A.v.A.)
| | - Alexandra von Altrock
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany; (K.-H.W.); (A.v.A.)
| |
Collapse
|
29
|
Thanabalan A, Kiarie EG. Influence of Feeding Omega-3 Polyunsaturated Fatty Acids to Broiler Breeders on Indices of Immunocompetence, Gastrointestinal, and Skeletal Development in Broiler Chickens. Front Vet Sci 2021; 8:653152. [PMID: 34262961 PMCID: PMC8273488 DOI: 10.3389/fvets.2021.653152] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/01/2021] [Indexed: 01/24/2023] Open
Abstract
Modern broiler chickens are associated with rapid growth rates and superior feed efficiency. However, they are also susceptible to physiological and metabolic disorders (e.g., skin lesions, lameness, sudden death, enteric diseases, myopathies) that exert substantial economic losses to producers. This is further exacerbated by consumer pressure and mandated cessation of production practices such as indiscriminate use of antimicrobial growth promoters. Manipulation of broiler breeder (BB) nutrition and management can influence chick quality, robustness, and resilience to stressors in the production environment. The present review examines the role of feeding BB functional polyunsaturated omega-3 fatty acids (n-3 PUFA) and subsequent impact on the indices of immunocompetence, skeletal, and gastrointestinal (GIT) development in broiler chickens. Research in mammalian and avian models led evidence that perinatal feeding of long chain n-3 PUFA such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) engender transgenerational effects through regulation of a variety of biological processes including development of vital organs such as skeleton, brain and GIT. It is shown that feeding poultry breeders n-3 PUFA decreases inflammatory states and enriches hatching eggs with n-3 PUFA and immunoglobulins. Further evidence also shows that after 15 days of incubation, chicken embryos preferentially utilize long chain n-3 PUFA-critical for optimal cell, tissues, and organ development. Enrichment of n-3 PUFA in newly hatchling tissues reduce proinflammatory eicosanoids with consequences of enhanced bone mineralization. Dietary n-3 PUFA also modulates breeder GIT microbiota with consequences of microbial colonization and succession in chicks. As well, research shows that feeding poultry breeders n-3 PUFA bolsters progeny immunocompetence through enhanced passive immunity and antibody titres against routine vaccination. In conclusion, it appears that chicks may benefit from the incorporation of n-3 PUFA in the breeder diets; however, little attention is paid to fatty acids composition in breeder nutrition. We also highlight gaps in knowledge and future research perspectives.
Collapse
Affiliation(s)
| | - Elijah G Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
30
|
Lobo de Sá FD, Heimesaat MM, Bereswill S, Nattramilarasu PK, Schulzke JD, Bücker R. Resveratrol Prevents Campylobacter jejuni-Induced Leaky gut by Restoring Occludin and Claudin-5 in the Paracellular Leak Pathway. Front Pharmacol 2021; 12:640572. [PMID: 33935732 PMCID: PMC8082453 DOI: 10.3389/fphar.2021.640572] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/19/2021] [Indexed: 12/22/2022] Open
Abstract
Campylobacter jejuni is a bacterial human pathogen causing gastroenteritis and sequelae like irritable bowel syndrome. Epidemiologists count the human campylobacteriosis by C. jejuni as the most common foodborne zoonosis and bacterial diarrheal disease worldwide. Based on bioinformatics predictions for potential protective compounds in campylobacteriosis, the question was raised whether the plant-based polyphenol resveratrol is sufficient to attenuate intestinal epithelial damage induced by C. jejuni. We investigated this by performing experimental infection studies in an epithelial cell culture and the secondary abiotic IL-10-/- mouse model. In C. jejuni-infected human colonic HT-29/B6 cell monolayers, transepithelial electrical resistance (TER) was decreased and the paracellular marker flux of fluorescein (332 Da) increased. Concomitantly, the tight junction (TJ) proteins occludin and claudin-5 were re-distributed off the tight junction domain. This was accompanied by an increased induction of epithelial apoptosis, both changes contributing to compromised barrier function and the opening of the leak pathway induced by C. jejuni. In parallel, the recovery experiments with the application of resveratrol revealed a functional improvement of the disturbed epithelial barrier in both models in vitro and in vivo. During treatment with resveratrol, TJ localization of occludin and claudin-5 was fully restored in the paracellular domain of HT-29/B6 cells. Moreover, resveratrol decreased the rate of epithelial apoptosis. These resveratrol-induced molecular and cellular effects would therefore be expected to improve epithelial barrier function, thereby minimizing the so-called leaky gut phenomenon. In conclusion, the induction of the leak pathway by C. jejuni and the restoration of barrier function by resveratrol demonstrates its effectiveness as a potential preventive or therapeutic method of mitigating the leaky gut associated with campylobacteriosis.
Collapse
Affiliation(s)
- F. D. Lobo de Sá
- Medical Department, Division of Gastroenterology, Infectious Diseases, Rheumatology, Nutritional Medicine/Clinical Physiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - M. M. Heimesaat
- Institute of Microbiology, Infectious Diseases, and Immunology, Gastrointestinal Microbiology Research Group, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - S. Bereswill
- Institute of Microbiology, Infectious Diseases, and Immunology, Gastrointestinal Microbiology Research Group, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - P. K. Nattramilarasu
- Medical Department, Division of Gastroenterology, Infectious Diseases, Rheumatology, Nutritional Medicine/Clinical Physiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - J. D. Schulzke
- Medical Department, Division of Gastroenterology, Infectious Diseases, Rheumatology, Nutritional Medicine/Clinical Physiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - R. Bücker
- Medical Department, Division of Gastroenterology, Infectious Diseases, Rheumatology, Nutritional Medicine/Clinical Physiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
31
|
Khemgaew R, Omachi M, Takesada T, Vetchapitak T, Sato H, Taniguchi T, Misawa N. Transcellular penetration of Treponema phagedenis isolated from papillomatous digital dermatitis in polarized normal human epidermal keratinocytes in vitro. J Vet Med Sci 2021; 83:889-897. [PMID: 33853987 PMCID: PMC8267188 DOI: 10.1292/jvms.21-0034] [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] [Indexed: 11/22/2022] Open
Abstract
Papillomatous digital dermatitis (PDD) is a polymicrobial infection causing lameness in
dairy cattle. Culture-independent analysis has shown that Treponema
phagedenis is present consistently and predominantly in the lesions. However,
the pathogenesis of PDD, especially the tissue penetration pathway, has not been examined.
In the present study, we investigated whether T. phagedenis strains
isolated from PDD produce proteolytic enzyme (s) for disruption of the epithelial cell
barrier and have the ability to translocate in polarized normal human epidermal
keratinocytes (NHEK) in vitro. Ten strains of T.
phagedenis isolated from lesions did not show proteolytic activity on modified
skim milk agar, although a human strain of T. denticola used as a control
showed such activity. The integrity of tight junctions was monitored by measurement of
transepithelial electrical resistance (TER). The TER values after inoculation of the
T. phagedenis strains examined did not change during the experimental
period; however, apical to basolateral translocation of T. phagedenis was
confirmed after 24 hr by microscopy and Treponema-specific PCR. We
further confirmed that translocation of T. phagedenis was accelerated by
co-inoculation with live T. denticola, but not with heat-killed
organisms. Furthermore, tight junction ZO-1 protein was not lost intensity after
inoculation with T. phagedenis and the organism was observed in NHEK
cells using a florescence microscope. These results suggest that T.
phagedenis strains may translocate via a transcellular route in
vitro and that the invasion is accelerated by other bacteria, such as
T. denticola, producing proteolytic activity.
Collapse
Affiliation(s)
- Rathanon Khemgaew
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, 5200 Kihara-Kiyotakecho, Miyazaki 889-1692, Japan.,Laboratory of Veterinary Public Health, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Mari Omachi
- Laboratory of Veterinary Public Health, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Tomoe Takesada
- Laboratory of Veterinary Public Health, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Torrung Vetchapitak
- Center for Animal Disease Control, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Hiroyuki Sato
- Laboratory of Veterinary Clinical Radiology, Department of Veterinary Medical Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Takako Taniguchi
- Center for Animal Disease Control, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| | - Naoaki Misawa
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, 5200 Kihara-Kiyotakecho, Miyazaki 889-1692, Japan.,Laboratory of Veterinary Public Health, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan.,Center for Animal Disease Control, University of Miyazaki, 1-1 Gakuenkibanadai-nishi, Miyazaki 889-2192, Japan
| |
Collapse
|
32
|
Chok KC, Ng KY, Koh RY, Chye SM. Role of the gut microbiome in Alzheimer's disease. Rev Neurosci 2021; 32:767-789. [PMID: 33725748 DOI: 10.1515/revneuro-2020-0122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting millions of individuals each year and this number is expected to significantly increase. The complicated microorganisms residing in human gut are closely associated with our health. Emerging evidence has suggested possible involvement of human gut microbiome in AD. Symbiotic gut microbiomes are known to maintain brain health by modulating host's barriers integrity, metabolic system, immune system, nervous system and endocrine system. However, in the event of gut dysbiosis and barriers disruption, gut pathobionts disrupt homeostasis of the metabolic system, immune system, nervous system, and endocrine system, resulting in deterioration of neurological functions and subsequently promoting development of AD. Multiple therapeutic approaches, such as fecal microbiome transplant, antibiotics, prebiotics, probiotics, symbiotic, and diet are discussed as potential treatment options for AD by manipulating the gut microbiome to reverse pathological alteration in the systems above.
Collapse
Affiliation(s)
- Kian Chung Chok
- School of Health Science, International Medical University, 57000Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500Selangor, Malaysia
| | - Rhun Yian Koh
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Mu W, Sharma M, Heymans R, Ritou E, Rezek V, Hamid P, Kossyvakis A, Sen Roy S, Grijalva V, Chattopadhyay A, Papesh J, Meriwether D, Kitchen SG, Fogelman AM, Reddy ST, Kelesidis T. Apolipoprotein A-I mimetics attenuate macrophage activation in chronic treated HIV. AIDS 2021; 35:543-553. [PMID: 33306550 PMCID: PMC8010648 DOI: 10.1097/qad.0000000000002785] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Despite antiretroviral therapy (ART), there is an unmet need for therapies to mitigate immune activation in HIV infection. The goal of this study is to determine whether the apoA-I mimetics 6F and 4F attenuate macrophage activation in chronic HIV. DESIGN Preclinical assessment of the in-vivo impact of Tg6F and the ex-vivo impact of apoA-I mimetics on biomarkers of immune activation and gut barrier dysfunction in treated HIV. METHODS We used two humanized murine models of HIV infection to determine the impact of oral Tg6F with ART (HIV+ART+Tg6F+) on innate immune activation (plasma human sCD14, sCD163) and gut barrier dysfunction [murine I-FABP, endotoxin (LPS), LPS-binding protein (LBP), murine sCD14]. We also used gut explants from 10 uninfected and 10 HIV-infected men on potent ART and no morbidity, to determine the impact of ex-vivo treatment with 4F for 72 h on secretion of sCD14, sCD163, and I-FABP from gut explants. RESULTS When compared with mice treated with ART alone (HIV+ART+), HIV+ART+Tg6F+ mice attenuated macrophage activation (h-sCD14, h-sCD163), gut barrier dysfunction (m-IFABP, LPS, LBP, and m-sCD14), plasma and gut tissue oxidized lipoproteins. The results were consistent with independent mouse models and ART regimens. Both 4F and 6F attenuated shedding of I-FABP and sCD14 from gut explants from HIV-infected and uninfected participants. CONCLUSION Given that gut barrier dysfunction and macrophage activation are contributors to comorbidities like cardiovascular disease in HIV, apoA-I mimetics should be tested as therapy for morbidity in chronic treated HIV.
Collapse
Affiliation(s)
- William Mu
- Division of Infectious Diseases
- Division of Hematology and Oncology
| | | | | | | | | | - Philip Hamid
- Division of Infectious Diseases
- Division of Hematology and Oncology
| | | | | | - Victor Grijalva
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Arnab Chattopadhyay
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Jeremy Papesh
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - David Meriwether
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | | | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
- Department of Molecular and Medical Pharmacology
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | |
Collapse
|
34
|
Lobo de Sá FD, Schulzke JD, Bücker R. Diarrheal Mechanisms and the Role of Intestinal Barrier Dysfunction in Campylobacter Infections. Curr Top Microbiol Immunol 2021; 431:203-231. [PMID: 33620653 DOI: 10.1007/978-3-030-65481-8_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Campylobacter enteritis is the most common cause of foodborne bacterial diarrhea in humans. Although various studies have been performed to clarify the pathomechanism in Campylobacter infection, the mechanism itself and bacterial virulence factors are yet not completely understood. The purpose of this chapter is to (i) give an overview on Campylobacter-induced diarrheal mechanisms, (ii) illustrate underlying barrier defects, (iii) explain the role of the mucosal immune response and (iv) weigh preventive and therapeutic approaches. Our present knowledge of pathogenetic and diarrheal mechanisms of Campylobacter jejuni is explained in the first part of this chapter. In the second part, the molecular basis for the Campylobacter-induced barrier dysfunction is compared with that of other species in the Campylobacter genus. The bacteria are capable of overcoming the intestinal epithelial barrier. The invasion into the intestinal mucosa is the initial step of the infection, followed by a second step, the epithelial barrier impairment. The extent of the impairment depends on various factors, including tight junction dysregulation and epithelial apoptosis. The disturbed intestinal epithelium leads to a loss of water and solutes, the leak flux type of diarrhea, and facilitates the uptake of harmful antigens, the leaky gut phenomenon. The barrier dysfunction is accompanied by increased pro-inflammatory cytokine secretion, which is partially responsible for the dysfunction. Moreover, cytokines also mediate ion channel dysregulation (e.g., epithelial sodium channel, ENaC), leading to another diarrheal mechanism, which is sodium malabsorption. Future perspectives of Campylobacter research are the clarification of molecular pathomechanisms and the characterization of therapeutic and preventive compounds to combat and prevent Campylobacter infections.
Collapse
Affiliation(s)
- Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
35
|
Shen Y, Zou J, Chen M, Zhang Z, Liu C, Jiang S, Qian D, Duan JA. Protective effects of Lizhong decoction on ulcerative colitis in mice by suppressing inflammation and ameliorating gut barrier. JOURNAL OF ETHNOPHARMACOLOGY 2020; 259:112919. [PMID: 32360800 DOI: 10.1016/j.jep.2020.112919] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/21/2020] [Accepted: 04/25/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lizhong Decoction (LZD) is a classical prescription firstly recorded in "Shanghan Lun". It has been used to clinically treat ulcerative colitis (UC) for thousands of years. However, its mechanism is not clear up to now. AIM OF THE STUDY The goal of this study was to assess the amelioration of LZD on dextran sodium sulfate (DSS)-induced colitis in mice and further clarify its mechanism. MATERIALS AND METHODS The ulcerative colitis model induced by DSS was successfully established and applied to evaluate the intervention effect after oral administration of LZD. Furthermore, the expression of key targets in inflammatory signaling pathways and intestinal tight junction proteins were investigated by enzyme-linked immunosorbent assay (ELISA) and quantitative real time polymerase chain reaction (qPCR) analysis. RESULTS The results showed that all doses of LZD could notably improve DSS-induced colon lesions, reduce histological scores, prolong colon length and increase body weight. Colonic inflammation in UC mice was significantly alleviated by inhibiting the activities of myeloperoxidase (MPO) and superoxide dismutase (SOD), reducing the yield of nitric oxide (NO) and inflammatory cytokines such as interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6), and along with promoting the production of anti-inflammatory cytokines such as interleukin-4 (IL-4) and interleukin-10 (IL-10) after LZD treatment. Furthermore, LZD remarkably down-regulated the level of toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) mRNA and up-regulated the expression of tight junction proteins (zonula occluden-1, occludin and claudin-1) in UC mice. CONCLUSION In summary, this study indicated that LZD could notably improve UC symptoms by suppressing inflammation and ameliorating gut barrier, which provided scientific basis for its clinical application in the future.
Collapse
Affiliation(s)
- Yumeng Shen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Junfeng Zou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Mengjun Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zhimiao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chen Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Dawei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
36
|
Azimi T, Nasser A, Shariati A, Shiadeh SMJ, Safari H, Alizade-Sani M, Taghipour A, Dehghan A. The Possible Role of Pathogenic and Non-Pathogenic Bacteria in Initiation and Exacerbation of Celiac Disease; A Comprehensive Review. Curr Pharm Biotechnol 2020; 21:452-466. [PMID: 31858910 DOI: 10.2174/1389201021666191219160729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/29/2019] [Accepted: 11/22/2019] [Indexed: 02/08/2023]
Abstract
Celiac Disease (CD) is an immune-mediated enteropathy, generally of the proximal intestine, that occurs in genetically susceptible individuals triggered by the ingestion of gluten. The incidence and frequency of CD are increasing, and it is predicted that CD affects approximately 1% of the people worldwide. The common clinical manifestations of CD are divided in two sections, including classic and non-classic symptoms that can be created in childhood and adulthood. The relationship between pathogenic and non-pathogenic bacteria with CD is complex and multidirectional. In previous published studies, results demonstrated the triggering impact of bacteria, viruses, and parasites on initiation and development of Inflammatory Bowel Disease (IBD) and Irritable Bowel Syndrome (IBS). Different studies revealed the inducing effect of pathogenic and non-pathogenic bacteria on CD. However, increasing evidence proposes that some of these microorganisms can also play several positive roles in CD process. Although information of the pathogenesis of the CD is quickly expanding, the possible role of bacteria needs further examination. In conclusion, with respect to the possible correlation between different bacteria in CD, the current review-based study aims to discuss the possible relationship between CD and pathogenic and non-pathogenic bacteria and to show various and significant aspects of mechanisms involved in the CD process.
Collapse
Affiliation(s)
- Taher Azimi
- Pediatric Infections Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Nasser
- Clinical Microbiology Research Center, Ilam University of Medical Science, Ilam, Iran.,Department of Medical Microbiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Aref Shariati
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Seyedeh M J Shiadeh
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Safari
- Health Promotion Research Center, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Alizade-Sani
- Students Research Committee, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Taghipour
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amin Dehghan
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Budge S, Barnett M, Hutchings P, Parker A, Tyrrel S, Hassard F, Garbutt C, Moges M, Woldemedhin F, Jemal M. Risk factors and transmission pathways associated with infant Campylobacter spp. prevalence and malnutrition: A formative study in rural Ethiopia. PLoS One 2020; 15:e0232541. [PMID: 32384130 PMCID: PMC7209302 DOI: 10.1371/journal.pone.0232541] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Early infection from enteropathogens is recognised as both a cause and effect of infant malnutrition. Specifically, evidence demonstrates associations between growth shortfalls and Campylobacter infection, endemic across low-income settings, with poultry a major source. Whilst improvements in water, sanitation and hygiene (WASH) should reduce pathogen transmission, interventions show inconsistent effects on infant health. This cross-sectional, formative study aimed to understand relationships between infant Campylobacter prevalence, malnutrition and associated risk factors, including domestic animal husbandry practices, in rural Ethiopia. Thirty-five households were visited in Sidama zone, Southern Nations, Nationalities and Peoples' region. Infant and poultry faeces and domestic floor surfaces (total = 102) were analysed for presumptive Campylobacter spp. using selective culture. Infant anthropometry and diarrhoeal prevalence, WASH facilities and animal husbandry data were collected. Of the infants, 14.3% were wasted, 31.4% stunted and 31.4% had recent diarrhoea. Presumptive Campylobacter spp. was isolated from 48.6% of infant, 68.6% of poultry and 65.6% of floor surface samples. Compared to non-wasted infants, wasted infants had an increased odds ratio (OR) of 1.41 for a Campylobacter-positive stool and 1.81 for diarrhoea. Positive infant stools showed a significant relationship with wasting (p = 0.026) but not stunting. Significant risk factors for a positive stool included keeping animals inside (p = 0.027, OR 3.5), owning cattle (p = 0.018, OR 6.5) and positive poultry faeces (p<0.001, OR 1.34). Positive floor samples showed a significant correlation with positive infant (p = 0.023), and positive poultry (p = 0.013, OR 2.68) stools. Ownership of improved WASH facilities was not correlated with lower odds of positive stools. This formative study shows a high prevalence of infants positive for Campylobacter in households with free-range animals. Findings reaffirm contaminated floors as an important pathway to infant pathogen ingestion and suggest that simply upgrading household WASH facilities will not reduce infection without addressing the burden of contamination from animals, alongside adequate separation in the home.
Collapse
Affiliation(s)
| | - Megan Barnett
- British Geological Survey, Environmental Science Centre, Keyworth, Nottingham, United Kingdom
| | | | | | - Sean Tyrrel
- Cranfield University, Cranfield, United Kingdom
| | | | | | - Mathewos Moges
- Department of Environmental Health, Hawassa University College of Medicine and Health Sciences, Hawassa, Ethiopia
| | | | | |
Collapse
|
38
|
Alzheimer M, Svensson SL, König F, Schweinlin M, Metzger M, Walles H, Sharma CM. A three-dimensional intestinal tissue model reveals factors and small regulatory RNAs important for colonization with Campylobacter jejuni. PLoS Pathog 2020; 16:e1008304. [PMID: 32069333 PMCID: PMC7048300 DOI: 10.1371/journal.ppat.1008304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/28/2020] [Accepted: 01/02/2020] [Indexed: 02/06/2023] Open
Abstract
The Gram-negative Epsilonproteobacterium Campylobacter jejuni is currently the most prevalent bacterial foodborne pathogen. Like for many other human pathogens, infection studies with C. jejuni mainly employ artificial animal or cell culture models that can be limited in their ability to reflect the in-vivo environment within the human host. Here, we report the development and application of a human three-dimensional (3D) infection model based on tissue engineering to study host-pathogen interactions. Our intestinal 3D tissue model is built on a decellularized extracellular matrix scaffold, which is reseeded with human Caco-2 cells. Dynamic culture conditions enable the formation of a polarized mucosal epithelial barrier reminiscent of the 3D microarchitecture of the human small intestine. Infection with C. jejuni demonstrates that the 3D tissue model can reveal isolate-dependent colonization and barrier disruption phenotypes accompanied by perturbed localization of cell-cell junctions. Pathogenesis-related phenotypes of C. jejuni mutant strains in the 3D model deviated from those obtained with 2D-monolayers, but recapitulated phenotypes previously observed in animal models. Moreover, we demonstrate the involvement of a small regulatory RNA pair, CJnc180/190, during infections and observe different phenotypes of CJnc180/190 mutant strains in 2D vs. 3D infection models. Hereby, the CJnc190 sRNA exerts its pathogenic influence, at least in part, via repression of PtmG, which is involved in flagellin modification. Our results suggest that the Caco-2 cell-based 3D tissue model is a valuable and biologically relevant tool between in-vitro and in-vivo infection models to study virulence of C. jejuni and other gastrointestinal pathogens. Enteric pathogens have evolved numerous strategies to successfully colonize and persist in the human gastrointestinal tract. However, especially for the research of virulence mechanisms of human pathogens, often only limited infection models are available. Here, we have applied and further advanced a tissue-engineered human intestinal tissue model based on an extracellular matrix scaffold reseeded with human cells that can faithfully mimic pathogenesis-determining processes of the zoonotic pathogen Campylobacter jejuni. Our three-dimensional (3D) intestinal infection model allows for the assessment of epithelial barrier function during infection as well as for the quantification of bacterial adherence, internalization, and transmigration. Investigation of C. jejuni mutant strains in our 3D tissue model revealed isolate-specific infection phenotypes, in-vivo relevant infection outcomes, and uncovered the involvement of a small RNA pair during C. jejuni pathogenesis. Overall, our results demonstrate the power of tissue-engineered models for studying host-pathogen interactions, and our model will also be helpful to investigate other gastrointestinal pathogens.
Collapse
Affiliation(s)
- Mona Alzheimer
- Chair of Molecular Infection Biology II, Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Sarah L. Svensson
- Chair of Molecular Infection Biology II, Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Fabian König
- Chair of Molecular Infection Biology II, Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Matthias Schweinlin
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Marco Metzger
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
- Fraunhofer-Institute for Silicate Research, Translational Centre Regenerative Therapies, Würzburg, Germany
| | - Heike Walles
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke University, Magdeburg, Germany
- * E-mail: (HW); (CMS)
| | - Cynthia M. Sharma
- Chair of Molecular Infection Biology II, Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany
- * E-mail: (HW); (CMS)
| |
Collapse
|
39
|
Xi D, Alter T, Einspanier R, Sharbati S, Gölz G. Campylobacter jejuni genes Cj1492c and Cj1507c are involved in host cell adhesion and invasion. Gut Pathog 2020; 12:8. [PMID: 32064001 PMCID: PMC7011364 DOI: 10.1186/s13099-020-00347-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Background Campylobacter jejuni (C. jejuni) has been assigned as an important food-borne pathogen for human health but many pathogenicity factors of C. jejuni and human host cell responses related to the infection have not yet been adequately clarified. This study aimed to determine further C. jejuni pathogenicity factors and virulence genes based on a random mutagenesis approach. A transposon mutant library of C. jejuni NCTC 11168 was constructed and the ability of individual mutants to adhere to and invade human intestinal epithelial cells was evaluated compared to the wild type. We identified two mutants of C. jejuni possessing altered phenotypes with transposon insertions in the genes Cj1492c and Cj1507c. Cj1492c is annotated as a two-component sensor and Cj1507c is described as a regulatory protein. However, functions of both mutated genes are not clarified so far. Results In comparison to the wild type, Cj::1492c and Cj::1507c showed around 70-80% relative motility and Cj::1492c had around 3-times enhanced adhesion and invasion rates whereas Cj::1507c had significantly impaired adhesive and invasive capability. Moreover, Cj::1492c had a longer lag phase and slower growth rate while Cj::1507c showed similar growth compared to the wild type. Between 5 and 24 h post infection, more than 60% of the intracellular wild type C. jejuni were eliminated in HT-29/B6 cells, however, significantly fewer mutants were able to survive intracellularly. Nevertheless, no difference in host cell viability and induction of the pro-inflammatory chemokine IL-8 were determined between both mutants and the wild type. Conclusion We conclude that genes regulated by Cj1507c have an impact on efficient adhesion, invasion and intracellular survival of C. jejuni in HT-29/B6 cells. Furthermore, potential signal sensing by Cj1492c seems to lead to limiting attachment and hence internalisation of C. jejuni. However, as the intracellular survival capacities are reduced, we suggest that signal sensing by Cj1492c impacts several processes related to pathogenicity of C. jejuni.
Collapse
Affiliation(s)
- De Xi
- 1Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Thomas Alter
- 2Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Ralf Einspanier
- 1Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Soroush Sharbati
- 1Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Greta Gölz
- 2Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
40
|
Bian X, Garber JM, Cooper KK, Huynh S, Jones J, Mills MK, Rafala D, Nasrin D, Kotloff KL, Parker CT, Tennant SM, Miller WG, Szymanski CM. Campylobacter Abundance in Breastfed Infants and Identification of a New Species in the Global Enterics Multicenter Study. mSphere 2020; 5:e00735-19. [PMID: 31941810 PMCID: PMC6968651 DOI: 10.1128/msphere.00735-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Campylobacter jejuni is a leading cause of bacterial diarrhea worldwide and is associated with high rates of mortality and growth stunting in children inhabiting low- to middle-resource countries. To better understand the impact of breastfeeding on Campylobacter infection in infants in sub-Saharan Africa and South Asia, we examined fecal microbial compositions, bacterial isolates, and their carbohydrate metabolic pathways in Campylobacter-positive infants <1 year of age from the Global Enterics Multicenter Study. Exclusively breastfed infants with diarrhea exhibited high Campylobacter abundances, and this negatively correlated with bacterial carbohydrate metabolism. Although C. jejuni and Campylobacter coli are prevalent among these infants, the second most abundant Campylobacter species was a new species, which we named "Candidatus Campylobacter infans." Asymptomatic Campylobacter carriers also possess significantly different proportions of specific gut microbes compared to diarrheal cases. These findings provide insight into Campylobacter infections in infants in sub-Saharan Africa and South Asia and help inform strategies aimed at eliminating campylobacteriosis in these areas.IMPORTANCECampylobacter is the primary cause of bacterial diarrhea in the United States and can lead to the development of the postinfectious autoimmune neuropathy known as Guillain-Barré syndrome. Also, drug-resistant campylobacters are becoming a serious concern both locally and abroad. In low- and middle-income countries (LMICs), infection with Campylobacter is linked to high rates of morbidity, growth stunting, and mortality in children, and breastfeeding is important for infant nutrition, development, and protection against infectious diseases. In this study, we examined the relationship between breastfeeding and Campylobacter infection and demonstrate the increased selection for C. jejuni and C. coli strains unable to metabolize fucose. We also identify a new Campylobacter species coinfecting these infants with a high prevalence in five of the seven countries in sub-Saharan Africa and South Asia examined. These findings indicate that more detailed studies are needed in LMICs to understand the Campylobacter infection process in order to devise a strategy for eliminating this pathogenic microbe.
Collapse
Affiliation(s)
- Xiaoming Bian
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Jolene M Garber
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Kerry K Cooper
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| | - Steven Huynh
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | - Jennifer Jones
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael K Mills
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Daniel Rafala
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Dilruba Nasrin
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Craig T Parker
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | - Sharon M Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - William G Miller
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Albany, California, USA
| | - Christine M Szymanski
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
41
|
Hoffman S, Aviv Cohen N, Carroll IM, Tulchinsky H, Borovok I, Dotan I, Maharshak N. Faecal Proteases from Pouchitis Patients Activate Protease Activating Receptor-2 to Disrupt the Epithelial Barrier. J Crohns Colitis 2019; 13:1558-1568. [PMID: 31056700 DOI: 10.1093/ecco-jcc/jjz086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS The pathogenesis of pouch inflammation may involve epithelial barrier disruption. We investigated whether faecal proteolytic activity is increased during pouchitis and results in epithelial barrier dysfunction through protease activating receptor [PAR] activation, and assessed whether the intestinal microbiome may be the source of the proteases. METHODS Faecal samples were measured for protease activity using a fluorescein isothiocyanate [FITC]-casein florescence assay. Caco-2 cell monolayers were exposed to faecal supernatants to assess permeability to FITC-dextran. Tight junction protein integrity and PAR activation were assessed by immunoblot and immunofluorescence. A truncated PAR2 protein in Caco-2 cells was achieved by stable transfection using CRISPR/Cas9 plasmid. PAR2 activation in pouch biopsies was examined using antibodies directed to the N-terminus of the protein. Microbial composition was analysed based on 16S rRNA gene sequence analysis. RESULTS Ten pouchitis patients, six normal pouch [NP] patients and nine healthy controls [HC] were recruited. The pouchitis patients exhibited a 5.19- and 5.35-fold higher faecal protease [FP] activity [p ≤ 0.05] compared to the NP and HC participants, respectively. The genus Haemophilus was positively associated with FP activity [R = 0.718, false discovery rate < 0.1]. Faecal supernatants from pouchitis patients activated PAR2 on Caco-2 monolayers, disrupted tight junction proteins and increased epithelial permeability. PAR2 truncation in Caco-2 abrogated faecal protease-mediated permeability. Pouch biopsies obtained from pouchitis patients, but not from NP patients, displayed PAR2 activation. CONCLUSIONS Protease-producing bacteria may increase faecal proteolytic activity that results in pouch inflammation through disruption of tight junction proteins and increased epithelial permeability in a PAR2-dependent manner. This mechanism may initiate or propagate pouch inflammation.
Collapse
Affiliation(s)
- Sarit Hoffman
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Nathaniel Aviv Cohen
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,IBD Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Ian M Carroll
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hagit Tulchinsky
- Division of Surgery Colorectal Unit, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Ilya Borovok
- Department of Molecular and Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Nitsan Maharshak
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,IBD Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
42
|
Lobo de Sá FD, Butkevych E, Nattramilarasu PK, Fromm A, Mousavi S, Moos V, Golz JC, Stingl K, Kittler S, Seinige D, Kehrenberg C, Heimesaat MM, Bereswill S, Schulzke JD, Bücker R. Curcumin Mitigates Immune-Induced Epithelial Barrier Dysfunction by Campylobacter jejuni. Int J Mol Sci 2019; 20:ijms20194830. [PMID: 31569415 PMCID: PMC6802366 DOI: 10.3390/ijms20194830] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 12/26/2022] Open
Abstract
Campylobacter jejuni (C. jejuni) is the most common cause of foodborne gastroenteritis worldwide. The bacteria induce diarrhea and inflammation by invading the intestinal epithelium. Curcumin is a natural polyphenol from turmeric rhizome of Curcuma longa, a medical plant, and is commonly used in curry powder. The aim of this study was the investigation of the protective effects of curcumin against immune-induced epithelial barrier dysfunction in C. jejuni infection. The indirect C. jejuni-induced barrier defects and its protection by curcumin were analyzed in co-cultures with HT-29/B6-GR/MR epithelial cells together with differentiated THP-1 immune cells. Electrophysiological measurements revealed a reduction in transepithelial electrical resistance (TER) in infected co-cultures. An increase in fluorescein (332 Da) permeability in co-cultures as well as in the germ-free IL-10−/− mouse model after C. jejuni infection was shown. Curcumin treatment attenuated the C. jejuni-induced increase in fluorescein permeability in both models. Moreover, apoptosis induction, tight junction redistribution, and an increased inflammatory response—represented by TNF-α, IL-1β, and IL-6 secretion—was observed in co-cultures after infection and reversed by curcumin. In conclusion, curcumin protects against indirect C. jejuni-triggered immune-induced barrier defects and might be a therapeutic and protective agent in patients.
Collapse
Affiliation(s)
- Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Eduard Butkevych
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Praveen Kumar Nattramilarasu
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Anja Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Soraya Mousavi
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany.
| | - Verena Moos
- Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Julia C Golz
- German Federal Institute for Risk Assessment (BfR), Department of Biological Safety, National Reference Laboratory for Campylobacter, 12277 Berlin, Germany.
| | - Kerstin Stingl
- German Federal Institute for Risk Assessment (BfR), Department of Biological Safety, National Reference Laboratory for Campylobacter, 12277 Berlin, Germany.
| | - Sophie Kittler
- University of Veterinary Medicine Hannover, Research Center for Emerging Infections and Zoonoses, 30559 Hannover, Germany.
| | - Diana Seinige
- University of Veterinary Medicine Hannover, Research Center for Emerging Infections and Zoonoses, 30559 Hannover, Germany.
| | - Corinna Kehrenberg
- Institute for Veterinary Food Science, Justus-Liebig-University, 35392 Giessen, Germany.
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany.
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany.
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany.
| |
Collapse
|
43
|
Catechin and Procyanidin B 2 Modulate the Expression of Tight Junction Proteins but Do Not Protect from Inflammation-Induced Changes in Permeability in Human Intestinal Cell Monolayers. Nutrients 2019; 11:nu11102271. [PMID: 31546671 PMCID: PMC6836206 DOI: 10.3390/nu11102271] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022] Open
Abstract
The possibility of counteracting inflammation-related barrier defects with dietary compounds such as (poly)phenols has raised much interest, but information is still scarce. We have investigated here if (+)-catechin (CAT) and procyanidin B2 (PB2), two main dietary polyphenols, protect the barrier function of intestinal cells undergoing inflammatory stress. The cell model adopted consisted of co-cultured Caco-2 and HT29-MTX cells, while inflammatory conditions were mimicked through the incubation of epithelial cells with the conditioned medium of activated macrophages (MCM). The epithelial barrier function was monitored through trans-epithelial electrical resistance (TEER), and ROS production was assessed with dichlorofluorescein, while the expression of tight-junctional proteins and signal transduction pathways were evaluated with Western blot. The results indicated that MCM produced significant oxidative stress, the activation of NF-κB and MAPK pathways, a decrease in occludin and ZO-1 expression, and an increase in claudin-7 (CL-7) expression, while TEER was markedly lowered. Neither CAT nor PB2 prevented oxidative stress, transduction pathways activation, ZO-1 suppression, or TEER decrease. However, PB2 prevented the decrease in occludin expression and both polyphenols produced a huge increase in CL-7 abundance. It is concluded that, under the conditions adopted, CAT and PB2 do not prevent inflammation-dependent impairment of the epithelial barrier function of intestinal cell monolayers. However, the two compounds modify the expression of tight-junctional proteins and, in particular, markedly increase the expression of CL-7. These insights add to a better understanding of the potential biological activity of these major dietary flavan-3-ols at intestinal level.
Collapse
|
44
|
Schmidt AM, Escher U, Mousavi S, Tegtmeyer N, Boehm M, Backert S, Bereswill S, Heimesaat MM. Immunopathological properties of the Campylobacter jejuni flagellins and the adhesin CadF as assessed in a clinical murine infection model. Gut Pathog 2019; 11:24. [PMID: 31131028 PMCID: PMC6525468 DOI: 10.1186/s13099-019-0306-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022] Open
Abstract
Background Campylobacter jejuni infections constitute serious threats to human health with increasing prevalences worldwide. Our knowledge regarding the molecular mechanisms underlying host–pathogen interactions is still limited. Our group has established a clinical C. jejuni infection model based on abiotic IL-10−/− mice mimicking key features of human campylobacteriosis. In order to further validate this model for unraveling pathogen-host interactions mounting in acute disease, we here surveyed the immunopathological features of the important C. jejuni virulence factors FlaA and FlaB and the major adhesin CadF (Campylobacter adhesin to fibronectin), which play a role in bacterial motility, protein secretion and adhesion, respectively. Methods and results Therefore, abiotic IL-10−/− mice were perorally infected with C. jejuni strain 81-176 (WT) or with its isogenic flaA/B (ΔflaA/B) or cadF (ΔcadF) deletion mutants. Cultural analyses revealed that WT and ΔcadF but not ΔflaA/B bacteria stably colonized the stomach, duodenum and ileum, whereas all three strains were present in the colon at comparably high loads on day 6 post-infection. Remarkably, despite high colonic colonization densities, murine infection with the ΔflaA/B strain did not result in overt campylobacteriosis, whereas mice infected with ΔcadF or WT were suffering from acute enterocolitis at day 6 post-infection. These symptoms coincided with pronounced pro-inflammatory immune responses, not only in the intestinal tract, but also in other organs such as the liver and kidneys and were accompanied with systemic inflammatory responses as indicated by increased serum MCP-1 concentrations following C. jejuni ΔcadF or WT, but not ΔflaA/B strain infection. Conclusion For the first time, our observations revealed that the C. jejuni flagellins A/B, but not adhesion mediated by CadF, are essential for inducing murine campylobacteriosis. Furthermore, the secondary abiotic IL-10−/− infection model has been proven suitable not only for detailed investigations of immunological aspects of campylobacteriosis, but also for differential analyses of the roles of distinct C. jejuni virulence factors in induction and progression of disease. Electronic supplementary material The online version of this article (10.1186/s13099-019-0306-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna-Maria Schmidt
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Ulrike Escher
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Soraya Mousavi
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Nicole Tegtmeyer
- 2Institute for Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Manja Boehm
- 2Institute for Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Steffen Backert
- 2Institute for Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Stefan Bereswill
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Markus M Heimesaat
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| |
Collapse
|
45
|
Burge K, Gunasekaran A, Eckert J, Chaaban H. Curcumin and Intestinal Inflammatory Diseases: Molecular Mechanisms of Protection. Int J Mol Sci 2019; 20:ijms20081912. [PMID: 31003422 PMCID: PMC6514688 DOI: 10.3390/ijms20081912] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Intestinal inflammatory diseases, such as Crohn’s disease, ulcerative colitis, and necrotizing enterocolitis, are becoming increasingly prevalent. While knowledge of the pathogenesis of these related diseases is currently incomplete, each of these conditions is thought to involve a dysfunctional, or overstated, host immunological response to both bacteria and dietary antigens, resulting in unchecked intestinal inflammation and, often, alterations in the intestinal microbiome. This inflammation can result in an impaired intestinal barrier allowing for bacterial translocation, potentially resulting in systemic inflammation and, in severe cases, sepsis. Chronic inflammation of this nature, in the case of inflammatory bowel disease, can even spur cancer growth in the longer-term. Recent research has indicated certain natural products with anti-inflammatory properties, such as curcumin, can help tame the inflammation involved in intestinal inflammatory diseases, thus improving intestinal barrier function, and potentially, clinical outcomes. In this review, we explore the potential therapeutic properties of curcumin on intestinal inflammatory diseases, including its antimicrobial and immunomodulatory properties, as well as its potential to alter the intestinal microbiome. Curcumin may play a significant role in intestinal inflammatory disease treatment in the future, particularly as an adjuvant therapy.
Collapse
Affiliation(s)
- Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Aarthi Gunasekaran
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| |
Collapse
|
46
|
Schmidt AM, Escher U, Mousavi S, Boehm M, Backert S, Bereswill S, Heimesaat MM. Protease Activity of Campylobacter jejuni HtrA Modulates Distinct Intestinal and Systemic Immune Responses in Infected Secondary Abiotic IL-10 Deficient Mice. Front Cell Infect Microbiol 2019; 9:79. [PMID: 30984628 PMCID: PMC6449876 DOI: 10.3389/fcimb.2019.00079] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/08/2019] [Indexed: 01/20/2023] Open
Abstract
Even though human Campylobacter jejuni infections are progressively increasing worldwide, the underlying molecular mechanisms of pathogen-host-interactions are still not fully understood. We have recently shown that the secreted serine protease HtrA plays a key role in C. jejuni cellular invasion and transepithelial migration in vitro, and is involved in the onset of intestinal pathology in murine infection models in vivo. In the present study, we investigated whether the protease activity of HtrA had an impact in C. jejuni induced acute enterocolitis. For this purpose, we perorally infected secondary abiotic IL-10-/- mice with wildtype C. jejuni strain NCTC11168 (11168WT) or isogenic bacteria carrying protease-inactive HtrA with a single point mutation at S197A in the active center (11168HtrA-S197A). Irrespective of the applied pathogenic strain, mice harbored similar C. jejuni loads in their feces and exhibited comparably severe macroscopic signs of acute enterocolitis at day 6 postinfection (p.i.). Interestingly, the 11168HtrA-S197A infected mice displayed less pronounced colonic apoptosis and immune cell responses, but enhanced epithelial proliferation as compared to the 11168WT strain infected controls. Furthermore, less distinct microscopic sequelae in 11168HtrA-S197A as compared to parental strain infected mice were accompanied by less distinct colonic secretion of pro-inflammatory cytokines such as MCP-1, IL-6, TNF, and IFN-γ in the former as compared to the latter. Strikingly, the S197A point mutation was additionally associated with less pronounced systemic pro-inflammatory immune responses as assessed in serum samples. In conclusion, HtrA is a remarkable novel virulence determinant of C. jejuni, whose protease activity is not required for intestinal colonization and establishment of disease, but aggravates campylobacteriosis by triggering apoptosis and pro-inflammatory immune responses.
Collapse
Affiliation(s)
- Anna-Maria Schmidt
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Ulrike Escher
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Soraya Mousavi
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Manja Boehm
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
47
|
Vojdani A, Vojdani E. Reaction of antibodies to Campylobacter jejuni and cytolethal distending toxin B with tissues and food antigens. World J Gastroenterol 2019; 25:1050-1066. [PMID: 30862994 PMCID: PMC6406185 DOI: 10.3748/wjg.v25.i9.1050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/16/2019] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The bacteria Campylobacter jejuni (C. jejuni) is commonly associated with Guillane-Barré syndrome (GBS) and irritable bowel syndrome (IBS), but studies have also linked it with Miller Fisher syndrome, reactive arthritis and other disorders, some of which are autoimmune. It is possible that C. jejuni and its toxins may be cross-reactive with some human tissues and food antigens, potentially leading to autoimmune responses.
AIM To measure the immune reactivity of C. jejuni and C. jejuni cytolethal distending toxin (Cdt) antibodies with tissue and food antigens to examine their role in autoimmunities.
METHODS Using enzyme-linked immunosorbent assay (ELISA) methodology, specific antibodies made against C. jejuni and C. jejuni Cdt were applied to a variety of microwell plates coated with 45 tissues and 180 food antigens. The resulting immunoreactivities were compared to reactions with control wells coated with human serum albumin (HSA) which were used as negative controls and with wells coated with C. jejuni lysate or C. jejuni Cdt which served as positive controls.
RESULTS At 3 SD above the mean of control wells coated with HSA or 0.41 OD, the mouse monoclonal antibody made against C. jejuni showed moderate to high reactions with zonulin, somatotropin, acetylcholine receptor, β-amyloid and presenilin. This immune reaction was low with an additional 25 tissue antigens including asialoganglioside, and the same antibody did not react at all with another 15 tissue antigens. Examining the reaction between C. jejuni antibody and 180 food antigens, we found insignificant reactions with 163 foods but low to high immune reactions with 17 food antigens. Similarly, we examined the reaction of C. jejuni Cdt with the same tissues and food antigens. The strongest reactions were observed with zonulin, intrinsic factor and somatotropin. The reaction was moderate with 9 different tissue antigens including thyroid peroxidase, and reaction was low with another 10 different antigens, including neuronal antigens. The reaction of C. jejuni Cdt antibody with an additional 23 tissue antigens was insignificant. Regarding the reaction of C. jejuni Cdt antibody with different food antigens, 160 out of 180 foods showed insignificant reactions, while 20 foods showed reactions ranging from low to high.
CONCLUSION Our findings indicate that C. jejuni and its Cdt may play a role in inflammation and autoimmunities beyond the gut.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab., Inc., Los Angeles, CA 90035, United States
- Cyrex Labs, LLC., Phoenix, AZ 85034, United States
- Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Elroy Vojdani
- Regenera Medical, Los Angeles, CA 90025, United States
| |
Collapse
|
48
|
Harrer A, Bücker R, Boehm M, Zarzecka U, Tegtmeyer N, Sticht H, Schulzke JD, Backert S. Campylobacter jejuni enters gut epithelial cells and impairs intestinal barrier function through cleavage of occludin by serine protease HtrA. Gut Pathog 2019; 11:4. [PMID: 30805031 PMCID: PMC6373145 DOI: 10.1186/s13099-019-0283-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/01/2019] [Indexed: 01/17/2023] Open
Abstract
Campylobacter jejuni secretes HtrA (high temperature requirement protein A), a serine protease that is involved in virulence. Here, we investigated the interaction of HtrA with the host protein occludin, a tight junction strand component. Immunofluorescence studies demonstrated that infection of polarized intestinal Caco-2 cells with C. jejuni strain 81-176 resulted in a redistribution of occludin away from the tight junctions into the cytoplasm, an effect that was also observed in human biopsies during acute campylobacteriosis. Occludin knockout Caco-2 cells were generated by CRISPR/Cas9 technology. Inactivation of this gene affected the polarization of the cells in monolayers and transepithelial electrical resistance (TER) was reduced, compared to wild-type Caco-2 cells. Although tight junctions were still being formed, occludin deficiency resulted in a slight decrease of the tight junction plaque protein ZO-1, which was redistributed off the tight junction into the lateral plasma membrane. Adherence of C. jejuni to Caco-2 cell monolayers was similar between the occludin knockout compared to wild-type cells, but invasion was enhanced, indicating that deletion of occludin allowed larger numbers of bacteria to pass the tight junctions and to reach basal membranes to target the fibronectin receptor followed by cell entry. Finally, we discovered that purified C. jejuni HtrA cleaves recombinant occludin in vitro to release a 37 kDa carboxy-terminal fragment. The same cleavage fragment was observed in Western blots upon infection of polarized Caco-2 cells with wild-type C. jejuni, but not with isogenic ΔhtrA mutants. HtrA cleavage was mapped to the second extracellular loop of occludin, and a putative cleavage site was identified. In conclusion, HtrA functions as a secreted protease targeting the tight junctions, which enables the bacteria by cleaving occludin and subcellular redistribution of other tight junction proteins to transmigrate using a paracellular mechanism and subsequently invade epithelial cells.
Collapse
Affiliation(s)
- Aileen Harrer
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Roland Bücker
- 2Institut für Klinische Physiologie, Med. Klinik m.S. Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Manja Boehm
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Urszula Zarzecka
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany.,4Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Nicole Tegtmeyer
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Heinrich Sticht
- 3Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg D Schulzke
- 2Institut für Klinische Physiologie, Med. Klinik m.S. Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Steffen Backert
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| |
Collapse
|
49
|
The Inflammatory Response to Enterotoxigenic E. coli and Probiotic E. faecium in a Coculture Model of Porcine Intestinal Epithelial and Dendritic Cells. Mediators Inflamm 2018; 2018:9368295. [PMID: 30670931 PMCID: PMC6317115 DOI: 10.1155/2018/9368295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/23/2018] [Indexed: 12/31/2022] Open
Abstract
The gut epithelium constitutes an interface between the intestinal contents and the underlying gut-associated lymphoid tissue (GALT) including dendritic cells (DC). Interactions of intestinal epithelial cells (IEC) and resident DC are characterized by bidirectional crosstalk mediated by various factors, such as transforming growth factor-β (TGF-β) and thymic stromal lymphopoietin (TSLP). In the present study, we aimed (1) to model the interplay of both cell types in a porcine in vitro coculture consisting of IEC (cell line IPEC-J2) and monocyte-derived DC (MoDC) and (2) to assess whether immune responses to bacteria are altered because of the interplay between IPEC-J2 cells and MoDC. With regard to the latter, we focused on the inflammasome pathway. Here, we propose caspase-13 as a promising candidate for the noncanonical inflammasome activation in pigs. We conducted challenge experiments with enterotoxigenic Escherichia coli (ETEC) and probiotic Enterococcus faecium (E. faecium) NCIMB 10415. As potential mediators of IEC/DC interactions, TGF-β and TSLP were selected for analyses. Cocultured MoDC showed attenuated ETEC-induced inflammasome-related and proinflammatory interleukin (IL)-8 reactions compared with MoDC monocultures. Caspase-13 was more strongly expressed in IPEC-J2 cells cocultured with MoDC and upon ETEC incubation. We found that IPEC-J2 cells and MoDC were capable of releasing TSLP. The latter cells secreted greater amounts of TSLP when cocultured with IPEC-J2 cells. TGF-β was not modulated under the present experimental conditions in either cell types. We conclude that, in the presence of IPEC-J2 cells, porcine MoDC exhibited a more tolerogenic phenotype, which might be partially regulated by autocrine TSLP production. Noncanonical inflammasome signaling appeared to be modulated in IPEC-J2 cells. Our results indicate that the reciprocal interplay of the intestinal epithelium and GALT is essential for promoting balanced immune responses.
Collapse
|
50
|
Sima F, Stratakos AC, Ward P, Linton M, Kelly C, Pinkerton L, Stef L, Gundogdu O, Lazar V, Corcionivoschi N. A Novel Natural Antimicrobial Can Reduce the in vitro and in vivo Pathogenicity of T6SS Positive Campylobacter jejuni and Campylobacter coli Chicken Isolates. Front Microbiol 2018; 9:2139. [PMID: 30245680 PMCID: PMC6137164 DOI: 10.3389/fmicb.2018.02139] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/21/2018] [Indexed: 01/17/2023] Open
Abstract
Human campylobacteriosis is considered one of the most common foodborne diseases worldwide with poultry identified as the main source of infection accounting for 50-80% of human cases. Highly virulent Campylobacter spp., positive for the Type VI secretion system (T6SS), which have an increased ability to adhere to and invade the host gastrointestinal epithelium are highly prevalent in poultry. Multidrug resistant strains of bacteria are rapidly evolving and therefore, new antimicrobials to supplement animal feed that are able to control Campylobacter species, are in great need. The work presented herein indicates that a novel phenolic antimicrobial, Auranta 3001, is able to reduce the adhesion and invasion of human intestinal epithelial cells (HCT-8) by two T6SS positive chicken isolates, C. jejuni RC039 (p < 0.05) and C. coli RC013 (p < 0.001). Exposure of C. jejuni RC039 and C. coli RC013 to Auranta 3001 downregulated the expression of hcp and cetB genes, known to be important in the functionality of T6SS. Furthermore, the reduced adhesion and invasion is associated with a significant decrease in bacterial motility of both isolates (p < 0.05-p < 0.001) in vitro. Most importantly our in vivo results show that Auranta 3001 is able to reduce cecum colonization levels from log 8 CFU/ml to log 2 CFU/ml for C. jejuni RC039 and from log 7 CFU/ml to log 2 CFU/ml for C. coli RC013. In conclusion, this novel antimicrobial is able to reduce the pathogenic properties of T6SS campylobacters in vitro and also to decrease colonization in vivo.
Collapse
Affiliation(s)
- Filip Sima
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Auranta, NovaUCD, Dublin, Ireland
| | - Alexandros Ch. Stratakos
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
- Auranta, NovaUCD, Dublin, Ireland
| | | | - Mark Linton
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
| | - Carmel Kelly
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
| | - Laurette Pinkerton
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
| | - Lavinia Stef
- School of Animal Science and Biotechnology, Banat University of Animal Sciences and Veterinary Medicine – King Michael I of Romania, Timisoara, Romania
| | - Ozan Gundogdu
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Veronica Lazar
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
- School of Animal Science and Biotechnology, Banat University of Animal Sciences and Veterinary Medicine – King Michael I of Romania, Timisoara, Romania
| |
Collapse
|