1
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
2
|
Meng Z, Huang X, Qiao M, Song L, Liu Y, Hai D. Lactic Acid Bacteria Surface Proteins in the Mechanisms of Cell Adhesion and Immunoregulation. Food Sci Nutr 2024; 12:10148-10163. [PMID: 39723039 PMCID: PMC11666997 DOI: 10.1002/fsn3.4517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/09/2024] [Accepted: 08/29/2024] [Indexed: 12/28/2024] Open
Abstract
This study delves into the role of lactic acid bacteria (LAB) surface proteins in cell adhesion and immunoregulation. Using fluorescence microscopy, we observed distinct adhesion patterns on various cell types. LAB surface proteins demonstrated concentration-dependent inhibition of Salmonella adhesion, with LAB69 exhibiting potent antagonistic effects. Genetic expression analysis revealed nuanced responses in key genes (MD2, TLR4, IL-10, MUC3, MIF) across different cell types, highlighting the diverse immunomodulatory effects of LAB surface proteins. Modulation of pro-inflammatory (TNF-α) and anti-inflammatory (IL-10) cytokines further emphasized the complex interplay. In conclusion, this study underscores the pivotal role of LAB surface proteins in mediating cell adhesion and immunoregulation, providing a foundation for isolating specific immunomodulatory molecules within LAB surface proteins for potential applications in microbial ecological agents.
Collapse
Affiliation(s)
- Ziheng Meng
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Xianqing Huang
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| | - Mingwu Qiao
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| | - Lianjun Song
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| | - Yufei Liu
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Dan Hai
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| |
Collapse
|
3
|
Tewari N, Dey P. Navigating commensal dysbiosis: Gastrointestinal host-pathogen interplay orchestrating opportunistic infections. Microbiol Res 2024; 286:127832. [PMID: 39013300 DOI: 10.1016/j.micres.2024.127832] [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: 04/22/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
The gut commensals, which are usually symbiotic or non-harmful bacteria that live in the gastrointestinal tract, have a positive impact on the health of the host. This review, however, specifically discuss distinct conditions where commensals aid in the development of pathogenic opportunistic infections. We discuss that the categorization of gut bacteria as either pathogens or non-pathogens depends on certain circumstances, which are significantly affected by the tissue microenvironment and the dynamic host-microbe interaction. Under favorable circumstances, commensals have the ability to transform into opportunistic pathobionts by undergoing overgrowth. These conditions include changes in the host's physiology, simultaneous infection with other pathogens, effective utilization of nutrients, interactions between different species of bacteria, the formation of protective biofilms, genetic mutations that enhance pathogenicity, acquisition of genes associated with virulence, and the ability to avoid the host's immune response. These processes allow commensals to both initiate infections themselves and aid other pathogens in populating the host. This review highlights the need of having a detailed and sophisticated knowledge of the two-sided nature of gut commensals. Although commensals mostly promote health, they may also become harmful in certain changes in the environment or the body's functioning. This highlights the need of acknowledging the intricate equilibrium in interactions between hosts and microbes, which is crucial for preserving intestinal homeostasis and averting diseases. Finally, we also emphasize the further need of research to better understand and anticipate the behavior of gut commensals in different situations, since they play a crucial and varied role in human health and disease.
Collapse
Affiliation(s)
- Nisha Tewari
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India.
| |
Collapse
|
4
|
Poloamina VI, Alrammah H, Abate W, Avent ND, Fejer G, Jackson SK. Lysophosphatidylcholine Acetyltransferase 2 ( LPCAT2) Influences the Gene Expression of the Lipopolysaccharide Receptor Complex in Infected RAW264.7 Macrophages, Depending on the E. coli Lipopolysaccharide Serotype. BIOLOGY 2024; 13:314. [PMID: 38785798 PMCID: PMC11117747 DOI: 10.3390/biology13050314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Escherichia coli (E. coli) is a frequent gram-negative bacterium that causes nosocomial infections, affecting more than 100 million patients annually worldwide. Bacterial lipopolysaccharide (LPS) from E. coli binds to toll-like receptor 4 (TLR4) and its co-receptor's cluster of differentiation protein 14 (CD14) and myeloid differentiation factor 2 (MD2), collectively known as the LPS receptor complex. LPCAT2 participates in lipid-raft assembly by phospholipid remodelling. Previous research has proven that LPCAT2 co-localises in lipid rafts with TLR4 and regulates macrophage inflammatory response. However, no published evidence exists of the influence of LPCAT2 on the gene expression of the LPS receptor complex induced by smooth or rough bacterial serotypes. We used RAW264.7-a commonly used experimental murine macrophage model-to study the effects of LPCAT2 on the LPS receptor complex by transiently silencing the LPCAT2 gene, infecting the macrophages with either smooth or rough LPS, and quantifying gene expression. LPCAT2 only significantly affected the gene expression of the LPS receptor complex in macrophages infected with smooth LPS. This study provides novel evidence that the influence of LPCAT2 on macrophage inflammatory response to bacterial infection depends on the LPS serotype, and it supports previous evidence that LPCAT2 regulates inflammatory response by modulating protein translocation to lipid rafts.
Collapse
Affiliation(s)
| | - Hanaa Alrammah
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
- Zoonoses Research Unit, College of Veterinary Medicine, University of Bagdad, Baghdad 10071, Iraq
| | - Wondwossen Abate
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
- College of Medicine and Health, University of Exeter, Exeter EX1 2HZ, UK
| | - Neil D. Avent
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
| | - Gyorgy Fejer
- Faculty of Health, University of Plymouth, Plymouth PL4 8AA, UK
| | | |
Collapse
|
5
|
Didriksen BJ, Eshleman EM, Alenghat T. Epithelial regulation of microbiota-immune cell dynamics. Mucosal Immunol 2024; 17:303-313. [PMID: 38428738 PMCID: PMC11412483 DOI: 10.1016/j.mucimm.2024.02.008] [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: 12/12/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
The mammalian gastrointestinal tract hosts a diverse community of trillions of microorganisms, collectively termed the microbiota, which play a fundamental role in regulating tissue physiology and immunity. Recent studies have sought to dissect the cellular and molecular mechanisms mediating communication between the microbiota and host immune system. Epithelial cells line the intestine and form an initial barrier separating the microbiota from underlying immune cells, and disruption of epithelial function has been associated with various conditions ranging from infection to inflammatory bowel diseases and cancer. From several studies, it is now clear that epithelial cells integrate signals from commensal microbes. Importantly, these non-hematopoietic cells also direct regulatory mechanisms that instruct the recruitment and function of microbiota-sensitive immune cells. In this review, we discuss the central role that has emerged for epithelial cells in orchestrating intestinal immunity and highlight epithelial pathways through which the microbiota can calibrate tissue-intrinsic immune responses.
Collapse
Affiliation(s)
- Bailey J Didriksen
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily M Eshleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
6
|
Khabiri P, Rahimi MR, Rashidi I, Nedaei SE. Impacts of an 8-week regimen of aged garlic extract and aerobic exercise on the levels of Fetuin-A and inflammatory markers in the liver and visceral fat tissue of obese male rats. Clin Nutr ESPEN 2023; 58:79-88. [PMID: 38057040 DOI: 10.1016/j.clnesp.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 08/18/2023] [Accepted: 09/06/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND & AIMS Obesity-induced chronic low-grade systemic inflammation is linked to the development of numerous diseases. Fetuin-A is known to affect inflammation and insulin resistance in obesity conditions. Free fatty acid (FFA)-induced proinflammatory cytokine expression in adipocytes occurs only in the presence of both Fetuin-A and toll-like receptor 4 (TLR4) and removing either of them prevented FFA-induced insulin resistance. Aged garlic extract (AGE) and exercise training have anti-inflammatory effects; however, the impact of AGE on Fetuin-A is unknown. We examined the effects of AGE with or without aerobic training (AT) on Fetuin-A and inflammatory markers. METHODS Forty healthy male Sprague Dawley rats were randomly assigned to normal diet (ND) (n = 8) or high-fat diet (HFD) groups (n = 32) and fed for 9 weeks. After 9 weeks ND group continued normal diet, and the HFD group was randomly assigned to the HFD, HFD + AGE (600 mg/kg, once daily), HFD + AT (5 days/week), and HFD + AGE + AT groups that were continued for 8 weeks (n = 8). The significance of differences among groups was assessed using one-way analysis of variance followed by the post-hoc Tukey test. Statistically significant differences were considered for p < 0.05. RESULTS AGE, AT, and AGE + AT significantly decreased body weight, plasma Fetuin-A, HOMA-IR, mRNA and protein levels of Fetuin-A and NFƙB in the liver and mRNA and Protein levels of Fetuin-A, TLR4 and NFƙB in visceral adipose tissue (VAT) compared to HFD. However, only AGE + AT significantly decreased TLR4 protein levels in the liver. CONCLUSION Although AT and AGE reduce Fetuin-A and inflammatory markers, a combination of the two may be more effective at lowering inflammation.
Collapse
Affiliation(s)
- Parisa Khabiri
- Department of Exercise Physiology, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj, 66177-15175, Iran.
| | - Mohammad Rahman Rahimi
- Department of Exercise Physiology, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj, 66177-15175, Iran.
| | - Iraj Rashidi
- Department of Anatomical Sciences, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, 6715847141, Iran.
| | - Seyed Ershad Nedaei
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, 6715847141, Iran.
| |
Collapse
|
7
|
Dey P, Ray Chaudhuri S. The opportunistic nature of gut commensal microbiota. Crit Rev Microbiol 2023; 49:739-763. [PMID: 36256871 DOI: 10.1080/1040841x.2022.2133987] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 11/03/2022]
Abstract
The abundance of gut commensals has historically been associated with health-promoting effects despite the fact that the definition of good or bad microbiota remains condition-specific. The beneficial or pathogenic nature of microbiota is generally dictated by the dimensions of host-microbiota and microbe-microbe interactions. With the increasing popularity of gut microbiota in human health and disease, emerging evidence suggests opportunistic infections promoted by those gut bacteria that are generally considered beneficial. Therefore, the current review deals with the opportunistic nature of the gut commensals and aims to summarise the concepts behind the occasional commensal-to-pathogenic transformation of the gut microbes. Specifically, relevant clinical and experimental studies have been discussed on the overgrowth and bacteraemia caused by commensals. Three key processes and their underlying mechanisms have been summarised to be responsible for the opportunistic nature of commensals, viz. improved colonisation fitness that is dictated by commensal-pathogen interactions and availability of preferred nutrients; pathoadaptive mutations that can trigger the commensal-to-pathogen transformation; and evasion of host immune response as a survival and proliferation strategy of the microbes. Collectively, this review provides an updated concept summary on the underlying mechanisms of disease causative events driven by gut commensal bacteria.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
8
|
Iyer K, Erkert L, Becker C. Know your neighbors: microbial recognition at the intestinal barrier and its implications for gut homeostasis and inflammatory bowel disease. Front Cell Dev Biol 2023; 11:1228283. [PMID: 37519301 PMCID: PMC10375050 DOI: 10.3389/fcell.2023.1228283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
Intestinal epithelial cells (IECs) perform several physiological and metabolic functions at the epithelial barrier. IECs also play an important role in defining the overall immune functions at the mucosal region. Pattern recognition receptors (PRRs) on the cell surface and in other cellular compartments enable them to sense the presence of microbes and microbial products in the intestinal lumen. IECs are thus at the crossroads of mediating a bidirectional interaction between the microbial population and the immune cells present at the intestinal mucosa. This communication between the microbial population, the IECs and the underlying immune cells has a profound impact on the overall health of the host. In this review, we focus on the various PRRs present in different cellular compartments of IECs and discuss the recent developments in the understanding of their role in microbial recognition. Microbial recognition and signaling at the epithelial barrier have implications in the maintenance of intestinal homeostasis, epithelial barrier function, maintenance of commensals, and the overall tolerogenic function of PRRs in the gut mucosa. We also highlight the role of an aberrant microbial sensing at the epithelial barrier in the pathogenesis of inflammatory bowel disease (IBD) and the development of colorectal cancer.
Collapse
Affiliation(s)
- Krishna Iyer
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, United States
| | - Lena Erkert
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Costa EM, Silva S, Pereira CF, Ribeiro AB, Casanova F, Freixo R, Pintado M, Ramos ÓL. Carboxymethyl Cellulose as a Food Emulsifier: Are Its Days Numbered? Polymers (Basel) 2023; 15:polym15102408. [PMID: 37242982 DOI: 10.3390/polym15102408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Carboxymethyl cellulose use in industry is ubiquitous. Though it is recognized as safe by the EFSA and FDA, newer works have raised concerns related to its safety, as in vivo studies showed evidence of gut dysbiosis associated with CMC's presence. Herein lies the question, is CMC a gut pro-inflammatory compound? As no work addressed this question, we sought to understand whether CMC was pro-inflammatory through the immunomodulation of GI tract epithelial cells. The results showed that while CMC was not cytotoxic up to 25 mg/mL towards Caco-2, HT29-MTX and Hep G2 cells, it had an overall pro-inflammatory behavior. In a Caco-2 monolayer, CMC by itself increased IL-6, IL-8 and TNF-α secretion, with the latter increasing by 1924%, and with these increases being 9.7 times superior to the one obtained for the IL-1β pro-inflammation control. In co-culture models, an increase in secretion in the apical side, particularly for IL-6 (692% increase), was observed, and when RAW 264.7 was added, data showed a more complex scenario as stimulation of pro-inflammatory (IL-6, MCP-1 and TNF-α) and anti-inflammatory (IL-10 and IFN-β) cytokines in the basal side was observed. Considering these results, CMC may exert a pro-inflammatory effect in the intestinal lumen, and despite more studies being required, the incorporation of CMC in foodstuffs must be carefully considered in the future to minimize potential GI tract dysbiosis.
Collapse
Affiliation(s)
- Eduardo M Costa
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Sara Silva
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Carla F Pereira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Alessandra B Ribeiro
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Francisca Casanova
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Ricardo Freixo
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Manuela Pintado
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Óscar L Ramos
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| |
Collapse
|
10
|
Michael D, Kerry-Smith J, Webberley T, Murphy K, Plummer S, Parry L, Marchesi J. Does flow culture impact upon gut-probiotic interactions: A comparison with static culture. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
|
11
|
Investigation of TLR4 Antagonists for Prevention of Intestinal Inflammation. Inflammation 2023; 46:103-114. [PMID: 35867263 PMCID: PMC9971050 DOI: 10.1007/s10753-022-01714-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/08/2022] [Accepted: 07/04/2022] [Indexed: 11/05/2022]
Abstract
Activation of toll-like receptor 4 (TLR4) has been shown to be a major influence on the inflammatory signalling pathways in intestinal mucositis (IM), as demonstrated by TLR4 knock-out mice. Pharmacological TLR4 inhibition has thus been postulated as a potential new therapeutic approach for the treatment of IM but specific TLR4 inhibitors have yet to be investigated. As such, we aimed to determine whether direct TLR4 antagonism prevents inflammation in pre-clinical experimental models of IM. The non-competitive and competitive TLR4 inhibitors, TAK-242 (10 µM) and IAXO-102 (10 µM), respectively, or vehicle were added to human T84, HT-29, and U937 cell lines and mouse colonic explants 1 h before the addition of lipopolysaccharide (LPS) (in vitro: 100 µg/mL; ex vivo: 10 µg/mL), SN-38 (in vitro: 1 µM or 1 nM; ex vivo: 2 µM), and/or tumour necrosis factor-alpha (TNF-α) (5 µg/mL). Supernatant was collected for human IL-8 and mouse IL-6 enzyme-linked immunosorbent assays (ELISAs), as a measure of inflammatory signalling. Cell viability was measured using XTT assays. Explant tissue was used in histopathological and RT-PCR analysis for genes of interest: TLR4, MD2, CD14, MyD88, IL-6, IL-6R, CXCL2, CXCR1, CXCR2. SN-38 increased cytostasis compared to vehicle (P < 0.0001). However, this was not prevented by either antagonist (P > 0.05) in any of the 3 cell lines. Quantitative histological assessment scores showed no differences between vehicle and treatment groups (P > 0.05). There were no differences in in vitro IL-8 (P > 0.05, in all 3 cells lines) and ex vivo IL-6 (P > 0.05) concentrations between vehicle and treatment groups. Transcript expression of all genes was similar across vehicle and treatment groups (P > 0.05). TLR4 antagonism using specific inhibitors TAK-242 and IAXO-102 was not effective at blocking IM in these pre-clinical models of mucositis. This work indicates that specific epithelial inhibition of TLR4 with these compounds is insufficient to manage mucositis-related inflammation. Rather, TLR4 signalling through immune cells may be a more important target to prevent IM.
Collapse
|
12
|
Han Y, Jia R, Zhang J, Zhu Q, Wang X, Ji Q, Zhang W. Hypoxia Attenuates Colonic Innate Immune Response and Inhibits TLR4/NF-κB Signaling Pathway in Lipopolysaccharide-Induced Colonic Epithelial Injury Mice. J Interferon Cytokine Res 2023; 43:43-52. [PMID: 36603105 DOI: 10.1089/jir.2022.0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
High altitude hypoxia can lead to a spectrum of gastrointestinal problems. As the first line of host immune defense, innate immune response in the intestinal mucosa plays a pivotal role in maintaining intestinal homeostasis and protecting against intestinal injury at high altitude. This study aimed to investigate the effect of hypoxia on the colonic mucosal barrier and toll-like receptor 4 (TLR4)-mediated innate immune responses in the colon. The mice were exposed to a hypobaric chamber to simulate a 5,000 m plateau environment for 7 days, and the colonic mucosa changes were recorded. At the same time, the inflammation model was established by lipopolysaccharide (LPS) to explore the effects of hypoxia on the TLR4/nuclear factor kappa B (NF-κB) signaling pathway and its downstream inflammatory factors [tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and interferon (IFN)-γ] in the colon. We found that hypoxic exposure caused weight loss and structural disturbance of the colonic mucosa in mice. Compared with the control group, the protein levels of TLR4 [fold change (FC) = 0.75 versus FC = 0.23], MyD88 (FC = 0.80 versus FC = 0.30), TIR-domain-containing adaptor protein inducing interferon-β (TRIF: FC = 0.89 versus FC = 0.38), and NF-κB p65 (FC = 0.75 versus FC = 0.24) in the colon of mice in the hypobaric hypoxia group were significantly decreased. LPS-induced upregulation of the TLR4/NF-κB signaling and its downstream inflammatory factors was inhibited by hypoxia. Specifically, compared with the LPS group, the protein levels of TLR4 (FC = 1.18, FC = 0.86), MyD88 (FC = 1.20, FC = 0.80), TRIF (FC = 1.20, FC = 0.86), and NF-κB p65 (FC = 1.29, FC = 0.62) and the mRNA levels of IL-1β (FC = 7.38, FC = 5.06), IL-6 (FC = 16.06, FC = 9.22), and IFN-γ (FC = 2.01, FC = 1.16) were reduced in the hypobaric hypoxia plus LPS group. Our findings imply that hypoxia could lead to marked damage of the colonic mucosa and a reduction of TLR4-mediated colonic innate immune responses, potentially reducing host defense responses to colonic pathogens.
Collapse
Affiliation(s)
- Ying Han
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ruhan Jia
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Jingxuan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Qinfang Zhu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Xiaozhou Wang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Qiaorong Ji
- Department of Pathophysiology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| |
Collapse
|
13
|
Tam JSY, Pei JV, Coller JK, Prestidge CA, Bowen JM. Structural insight and analysis of TLR4 interactions with IAXO-102, TAK-242 and SN-38: an in silico approach. In Silico Pharmacol 2022; 11:1. [PMID: 36438853 PMCID: PMC9681971 DOI: 10.1007/s40203-022-00137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 11/05/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Toll-like receptor 4 (TLR4) has attracted interest due to its role in chemotherapy-induced gastrointestinal inflammation. This structural study aimed to provide in silico rational of the recognition and potential binding of TLR4 ligands IAXO-102, TAK-242, and SN-38 (the toxic metabolite of the chemotherapeutic irinotecan hydrochloride), which could contribute to rationale development of therapeutic anti-inflammation drugs targeting TLR4 in the gastrointestinal tract. Methods In silico docking was performed between the human TLR4-MD-2 complex and ligands (IAXO-102, TAK-242, SN-38) using Autodock Vina, setting the docking grids to cover either the upper or the lower bound of TLR4. The conformation having the lowest binding energy value (kcal/mol) was processed for post-hoc analysis of the best-fit model. Hydrogen bonding was calculated by using ChimeraX. Results Binding energies of IAXO-102, TAK-242 and SN-38 at the upper bound of TLR4-MD-2 ranged between - 3.8 and - 3.1, - 6.9 and - 6.3, and - 9.0 and - 7.0, respectively. Binding energies of IAXO-102, TAK-242 and SN-38 at the lower bound ranged between - 3.9 and - 3.5, - 6.5 and - 5.8, and - 8.2 and - 6.8, respectively. Hydrogen bonding at the upper bound of TLR4/MD-2 with IAXO-102, TAK-242 and SN-38 was to aspartic acid 70, cysteine 133 and serine 120, respectively. Hydrogen bonding at the lower bound of TLR4-MD-2 with IAXO-102, TAK-242 and SN-38 was to serine 528, glycine 480 and glutamine 510, respectively. Conclusion The in silico rational presented here supports further investigation of the binding activity of IAXO-102 and TAK-242 for their potential application in the prevention of gastrointestinal inflammation caused by SN-38.
Collapse
Affiliation(s)
- Janine S. Y. Tam
- Discipline of Physiology, Department of Physiology, School of Biomedicine, University of Adelaide, Adelaide, South Australia 5005 Australia
| | - Jinxin V. Pei
- Research School of Biology, College of Science, Australian National University, Canberra, Australian Capital Territory Australia
| | - Janet K. Coller
- Discipline of Pharmacology, School of Biomedicine, University of Adelaide, Adelaide, South Australia Australia
| | - Clive A. Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia Australia
| | - Joanne M. Bowen
- Discipline of Physiology, Department of Physiology, School of Biomedicine, University of Adelaide, Adelaide, South Australia 5005 Australia
| |
Collapse
|
14
|
Narabayashi H, Koma C, Nakata K, Ikegami M, Nakanishi Y, Ogihara J, Tsuda M, Hosono A, Hanazawa S, Takahashi K. Gut microbiota-dependent adaptor molecule recruits DNA methyltransferase to the TLR4 gene in colonic epithelial cells to suppress inflammatory reactions. Front Mol Biosci 2022; 9:1005136. [PMID: 36339704 PMCID: PMC9634067 DOI: 10.3389/fmolb.2022.1005136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
The intestine is inhabited by a large number of commensal bacteria that are immunologically non-self, potentially causing inflammation. However, in a healthy intestine, inflammation is strictly controlled at low levels to maintain homeostasis. We previously reported that the gut microbiota induce DNA methylation of the gene encoding Toll-like receptor (TLR) 4, a pattern recognition receptor that recognizes lipopolysaccharides of gram-negative bacteria, in colonic epithelial cells, suggesting its role in controlling intestinal inflammation. However, there remains a question of how gut microbiota cause methylation of only specific genes including TLR4, despite the fact that DNA methyltransferase (DNMT) is common to all genes targeted for methylation. Here, we identified RBM14 as an adaptor molecule that recruits DNMT to the TLR4 gene. RBM14 was shown to bind DNMT3 and be expressed at significantly higher levels in an intestinal epithelial cell (IEC) line with hypermethylated TLR4 gene than in an IEC line with hypomethylated TLR4 gene. In addition, RBM14 interacted with DNA regions of the TLR4 gene, and knockdown of RBM14 suppressed DNA methylation of the TLR4 gene in IECs. Furthermore, RBM14 expression was higher in colonic epithelial cells of conventional mice than in those of germ-free mice. Collectively, these results indicate that the gut microbiota induce methylation of the TLR4 gene in colonic epithelial cells by upregulating RBM14, which can recruit DNMT3 to the gene. The regulation of adaptor molecules such as RBM14, which bind to specific target genes and recruit DNMT, can explain, at least in part, how gut microbiota contribute to the maintenance of intestinal homeostasis through epigenetic control of specific gene expression in IECs.
Collapse
Affiliation(s)
- Hikari Narabayashi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Chiharu Koma
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Kazuaki Nakata
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Mion Ikegami
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Yusuke Nakanishi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Jun Ogihara
- Department of Chemistry and Life Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Masato Tsuda
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Akira Hosono
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Shigemasa Hanazawa
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Kyoko Takahashi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
- *Correspondence: Kyoko Takahashi,
| |
Collapse
|
15
|
McGrath CJ, Laveckis E, Bell A, Crost E, Juge N, Schüller S. Development of a novel human intestinal model to elucidate the effect of anaerobic commensals on Escherichia coli infection. Dis Model Mech 2022; 15:275170. [PMID: 35302159 PMCID: PMC9066490 DOI: 10.1242/dmm.049365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/10/2022] [Indexed: 01/01/2023] Open
Abstract
The gut microbiota plays a crucial role in protecting against enteric infection. However, the underlying mechanisms are largely unknown owing to a lack of suitable experimental models. Although most gut commensals are anaerobic, intestinal epithelial cells require oxygen for survival. In addition, most intestinal cell lines do not produce mucus, which provides a habitat for the microbiota. Here, we have developed a microaerobic, mucus-producing vertical diffusion chamber (VDC) model and determined the influence of Limosilactobacillus reuteri and Ruminococcus gnavus on enteropathogenic Escherichia coli (EPEC) infection. Optimization of the culture medium enabled bacterial growth in the presence of mucus-producing T84/LS174T cells. Whereas L. reuteri diminished EPEC growth and adhesion to T84/LS174T and mucus-deficient T84 epithelia, R. gnavus only demonstrated a protective effect in the presence of LS174T cells. Reduced EPEC adherence was not associated with altered type III secretion pore formation. In addition, co-culture with L. reuteri and R. gnavus dampened EPEC-induced interleukin 8 secretion. The microaerobic mucin-producing VDC system will facilitate investigations into the mechanisms underpinning colonization resistance and aid the development of microbiota-based anti-infection strategies. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Conor J. McGrath
- Department of Clinical Medicine, Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK
| | - Edgaras Laveckis
- Department of Clinical Medicine, Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK
| | - Andrew Bell
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich NR4 7UQ, UK
| | - Emmanuelle Crost
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich NR4 7UQ, UK
| | - Nathalie Juge
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich NR4 7UQ, UK
| | - Stephanie Schüller
- Department of Clinical Medicine, Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK,Author for correspondence ()
| |
Collapse
|
16
|
Lofft Z, Taibi A, Massara P, Tokar T, Paetau‐Robinson I, Khoo C, Comelli EM. Cranberry proanthocyanidin and its microbial metabolite 3,4‐dihydroxyphenylacetic acid, but not 3‐(4‐hydroxyphenyl)‐propionic acid, partially reverse pro‐inflammatory microRNA responses in human intestinal epithelial cells. Mol Nutr Food Res 2022; 66:e2100853. [DOI: 10.1002/mnfr.202100853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/13/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Zoe Lofft
- Department of Nutritional Sciences University of Toronto ON Canada
| | - Amel Taibi
- Department of Nutritional Sciences University of Toronto ON Canada
| | - Paraskevi Massara
- Department of Nutritional Sciences University of Toronto ON Canada
- Translational Medicine Program Hospital for Sick Children Toronto Canada
| | - Tomas Tokar
- Krembil Research Institute University Health Network Toronto ON M5T 0S8 Canada
| | | | - Christina Khoo
- Ocean Spray Cranberries, Inc. Lakeville‐Middleboro MA USA
| | - Elena M. Comelli
- Department of Nutritional Sciences University of Toronto ON Canada
- Joannah and Brian Lawson Centre for Child Nutrition University of Toronto ON Canada
| |
Collapse
|
17
|
Notararigo S, Varela E, Otal A, Antolín M, Guarner F, López P. Anti-Inflammatory Effect of an O-2-Substituted (1-3)-β-D-Glucan Produced by Pediococcus parvulus 2.6 in a Caco-2 PMA-THP-1 Co-Culture Model. Int J Mol Sci 2022; 23:ijms23031527. [PMID: 35163449 PMCID: PMC8835822 DOI: 10.3390/ijms23031527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 01/01/2023] Open
Abstract
Bacterial β-glucans are exopolysaccharides (EPSs), which can protect bacteria or cooperate in biofilm formation or in bacterial cell adhesion. Pediococcus parvulus 2.6 is a lactic acid bacterium that produces an O-2-substituted (1-3)-β-D-glucan. The structural similarity of this EPS to active compounds such as laminarin, together with its ability to modulate the immune system and to adhere in vitro to human enterocytes, led us to investigate, in comparison with laminarin, its potential as an immunomodulator of in vitro co-cultured Caco-2 and PMA-THP-1 cells. O-2-substituted (1-3)-β-D-glucan synthesized by the GTF glycosyl transferase of Pediococcus parvulus 2.6 or that by Lactococcus lactis NZ9000[pGTF] were purified and used in this study. The XTT tests revealed that all β-glucans were non-toxic for both cell lines and activated PMA-THP-1 cells’ metabolisms. The O-2-substituted (1-3)-β-D-glucan modulated production and expression of IL-8 and the IL-10 in Caco-2 and PMA-THP-1 cells. Laminarin also modulated cytokine production by diminishing TNF-α in Caco-2 cells and IL-8 in PMA-THP-1. All these features could be considered with the aim to produce function foods, supplemented with laminarin or with another novel β-glucan-producing strain, in order to ameliorate an individual’s immune system response toward pathogens or to control mild side effects in remission patients affected by inflammatory bowel diseases.
Collapse
Affiliation(s)
- Sara Notararigo
- Molecular Biology of Gram-Positive Bacteria, Margarita Salas Center for Biological Research (CIB-Margarita Salas-CSIC), Department of Microbial and Plant Biotechnology, Ramiro de Maeztu 9, 28040 Madrid, Spain;
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- Foundation Health Research Institute of Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Encarnación Varela
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- CIBERehd, Instituto Carlos III, 28029 Madrid, Spain
| | - Anna Otal
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
| | - María Antolín
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- CIBERehd, Instituto Carlos III, 28029 Madrid, Spain
| | - Francisco Guarner
- Digestive System Research Unit, Institut de RecercaValld’Hebron (VHIR), University Hospital Valld’Hebron, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.V.); (A.O.); (M.A.); (F.G.)
- CIBERehd, Instituto Carlos III, 28029 Madrid, Spain
| | - Paloma López
- Molecular Biology of Gram-Positive Bacteria, Margarita Salas Center for Biological Research (CIB-Margarita Salas-CSIC), Department of Microbial and Plant Biotechnology, Ramiro de Maeztu 9, 28040 Madrid, Spain;
- Correspondence: ; Tel.: +34-91-837-31-12; Fax: +34-91-538-04-32
| |
Collapse
|
18
|
Kotlyarov S. Involvement of the Innate Immune System in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2022; 23:985. [PMID: 35055174 PMCID: PMC8778852 DOI: 10.3390/ijms23020985] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 01/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common, socially significant disease characterized by progressive airflow limitation due to chronic inflammation in the bronchi. Although the causes of COPD are considered to be known, the pathogenesis of the disease continues to be a relevant topic of study. Mechanisms of the innate immune system are involved in various links in the pathogenesis of COPD, leading to persistence of chronic inflammation in the bronchi, their bacterial colonization and disruption of lung structure and function. Bronchial epithelial cells, neutrophils, macrophages and other cells are involved in the development and progression of the disease, demonstrating multiple compromised immune mechanisms.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
19
|
Tian Z, Li Z, Guo T, Li H, Mu Y. Atorvastatin suppresses lipopolysaccharide-induced inflammation in human coronary artery endothelial cells. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-979020200001181092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Zhen Tian
- Northeast Agricultural University, China; Harbin Medical University, China
| | | | - Tian Guo
- Harbin Medical University, China
| | - He Li
- Harbin Medical University, China
| | | |
Collapse
|
20
|
Zongo AWS, Zogona D, Youssef M, Ye S, Zhan F, Li J, Li B. Senegalia macrostachya seed polysaccharides attenuate inflammation-induced intestinal epithelial barrier dysfunction in a Caco-2 and RAW264.7 macrophage co-culture model by inhibiting the NF-κB/MLCK pathway. Food Funct 2022; 13:11676-11689. [DOI: 10.1039/d2fo02377f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Senegalia macrostachya seed polysaccharides improved the Caco-2 cell monolayer integrity from the inflammatory insult. SMSP2 treatment lowered the inflammatory cytokine release, increased TJ proteins, and downregulated the NF-κB/MLCK pathway.
Collapse
Affiliation(s)
- Abel Wend-Soo Zongo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
- Center for Research in Biological Sciences, Food and Nutrition, Department of Biochemistry and Microbiology, University Joseph Ki-Zerbo, BP 7021 Ouagadougou 03, Burkina Faso
| | - Daniel Zogona
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
- Center for Research in Biological Sciences, Food and Nutrition, Department of Biochemistry and Microbiology, University Joseph Ki-Zerbo, BP 7021 Ouagadougou 03, Burkina Faso
| | - Mahmoud Youssef
- Food Science and Technology Department, Faculty of Agriculture, Al-Azhar University, Cairo, Egypt
| | - Shuxin Ye
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Fuchao Zhan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| |
Collapse
|
21
|
Harikrishnan R, Devi G, Van Doan H, Balamurugan P, Arockiaraj J, Balasundaram C. Hepatic antioxidant activity, immunomodulation, and pro-anti-inflammatory cytokines manipulation of κ-carrageenan (κ-CGN) in cobia, Rachycentron canadum against Lactococcus garvieae. FISH & SHELLFISH IMMUNOLOGY 2021; 119:128-144. [PMID: 34562582 DOI: 10.1016/j.fsi.2021.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 06/13/2023]
Abstract
The effects of dietary k-Carrageenan (k-CGN) at 10, 20, and 30 g kg-1 on growth rate, hemato-biochemical indices, innate-adaptive parameters and modification of pro- and/or anti-inflammatory cytokines and chemokines pathway in cobia, Rachycentron canadum against Lactococcus garvieae is reported. The weight gain (WG) increased substantially (P < 0.05) in all k-CGN treated groups; the specific growth rate (SGR) was significant in healthy uninfected normal (HuN) and L. garvieae challenged (LaC) groups fed with 20 g kg-1k-CGN diet on 45 and 60 days. The white blood cell (WBC) counts, total protein (TP) level, total anti-oxidant (T-AOC), catalase (CAT), and glutathione (GSH) activities increased significantly when fed with 20 g and 30 g kg-1k-CG diets on 45th and 60th day. The immunological parameters such as phagocytic (PC) index and the activity of phagocytic (PC), respiratory burst (RB), superoxide dismutase (SOD), alternate complement pathway (ACH50), and lysozyme (LZM) were significantly enhanced with all k-CG diets in 45 and 60 days of treatment. No cumulative mortality (CM) in HuN group fed by control or any k-CGN diets. CM was 5% in LaC group fed with 20 g kg-1k-CGN diet whereas in LaC groups fed with 10 g and 30 g kg-1k-CGN diets the CM was 10%. The interleukin 1 beta (IL-1β) and tumor necrosis factor alpha (TNFα) pro-inflammatory cytokines mRNA transcripts were one-fold high (P < 0.05) in both HuN and LaC group fed all k-CGN enriched diets on 45 and 60 days. Similarly, IL-18 and TLR2 mRNA was one-fold high expression in both groups fed the 20 g and 30 g kg-1k-CGN enriched diets on 45 or 60 days. Interferon gamma (IFNγ) and interferon regulatory factor 3/7 (IRF3/IRF7) mRNA transcripts did not change with any diet. IL-6, IL-10, and IL-11 mRNA were one-fold high expressions in both groups fed the 20 g and 30 g kg-1k-CGN enriched diets on 45 and 60 days. However, the expression of CC1, CC3, and CCR9 pro-inflammatory chemokines mRNA did not vary with any control or k-CGN enriched diets. The results indicate that diet enriched with k-CGN at 20 g kg-1 significantly influences the growth, antioxidant and innate-adaptive immune performance, and pro-anti-inflammatory cytokines and chemokines regulation in cobia against L. garvieae.
Collapse
Affiliation(s)
- Ramasamy Harikrishnan
- Department of Zoology, Pachaiyappa's College for Men, Kanchipuram, 631 501, Tamil Nadu, India
| | - Gunapathy Devi
- Department of Zoology, Nehru Memorial College, Puthanampatti, 621 007, Tamil Nadu, India
| | - Hien Van Doan
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, 50200, Thailand; Science and Technology Research Institute, Chiang Mai University, 239 Huay Keaw Rd., Suthep, Muang, Chiang Mai, 50200, Thailand.
| | - Paramaraj Balamurugan
- Department of Biotechnology, St. Michael College of Engineering and Technology, Kalayarkoil, 630 551, Tamil Nadu, India
| | - Jesu Arockiaraj
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, 603 203, Chennai, Tamil Nadu, India; Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203, Chennai, Tamil Nadu, India
| | - Chellam Balasundaram
- Department of Herbal and Environmental Science, Tamil University, Thanjavur, 613 005, Tamil Nadu, India
| |
Collapse
|
22
|
Cuesta CM, Pascual M, Pérez-Moraga R, Rodríguez-Navarro I, García-García F, Ureña-Peralta JR, Guerri C. TLR4 Deficiency Affects the Microbiome and Reduces Intestinal Dysfunctions and Inflammation in Chronic Alcohol-Fed Mice. Int J Mol Sci 2021; 22:ijms222312830. [PMID: 34884634 PMCID: PMC8657603 DOI: 10.3390/ijms222312830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/21/2022] Open
Abstract
Chronic alcohol abuse causes an inflammatory response in the intestinal tract with damage to the integrity of the mucosa and epithelium, as well as dysbiosis in the gut microbiome. However, the role of gut bacteria in ethanol effects and how these microorganisms interact with the immune system are not well understood. The aim of the present study was to evaluate if TLR4 alters the ethanol-induced intestinal inflammatory response, and whether the response of this receptor affects the gut microbiota profile. We analyzed the 16S rRNA sequence of the fecal samples from wild-type (WT) and TLR4-knockout (TLR4-KO) mice with and without ethanol intake for 3 months. The results demonstrated that chronic ethanol consumption reduces microbiota diversity and causes dysbiosis in WT mice. Likewise, ethanol upregulates several inflammatory genes (IL-1β, iNOS, TNF-α) and miRNAs (miR-155-5p, miR-146a-5p) and alters structural and permeability genes (INTL1, CDH1, CFTR) in the colon of WT mice. Our results further demonstrated that TLR4-KO mice exhibit a different microbiota that can protect against the ethanol-induced activation of the immune system and colon integrity dysfunctions. In short, our results reveal that TLR4 is a key factor for determining the gut microbiota, which can participate in dysbiosis and the inflammatory response induced by alcohol consumption.
Collapse
Affiliation(s)
- Carlos M. Cuesta
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
| | - María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 15 Avda. Blasco Ibanez, 46010 Valencia, Spain
| | - Raúl Pérez-Moraga
- Bioinformatics and Biostatistics Unit, Prince Felipe Research Center, 46012 Valencia, Spain; (R.P.-M.); (F.G.-G.)
| | - Irene Rodríguez-Navarro
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Prince Felipe Research Center, 46012 Valencia, Spain; (R.P.-M.); (F.G.-G.)
| | - Juan R. Ureña-Peralta
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
- Correspondence: (J.R.U.-P.); (C.G.)
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Prince Felipe Research Center, 46012 Valencia, Spain; (C.M.C.); (M.P.); (I.R.-N.)
- Correspondence: (J.R.U.-P.); (C.G.)
| |
Collapse
|
23
|
Gorecki AM, Anyaegbu CC, Anderton RS. TLR2 and TLR4 in Parkinson's disease pathogenesis: the environment takes a toll on the gut. Transl Neurodegener 2021; 10:47. [PMID: 34814947 PMCID: PMC8609261 DOI: 10.1186/s40035-021-00271-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/29/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is an incurable, devastating disorder that is characterized by pathological protein aggregation and neurodegeneration in the substantia nigra. In recent years, growing evidence has implicated the gut environment and the gut-brain axis in the pathogenesis and progression of PD, especially in a subset of people who exhibit prodromal gastrointestinal dysfunction. Specifically, perturbations of gut homeostasis are hypothesized to contribute to α-synuclein aggregation in enteric neurons, which may spread to the brain over decades and eventually result in the characteristic central nervous system manifestations of PD, including neurodegeneration and motor impairments. However, the mechanisms linking gut disturbances and α-synuclein aggregation are still unclear. A plethora of research indicates that toll-like receptors (TLRs), especially TLR2 and TLR4, are critical mediators of gut homeostasis. Alongside their established role in innate immunity throughout the body, studies are increasingly demonstrating that TLR2 and TLR4 signalling shapes the development and function of the gut and the enteric nervous system. Notably, TLR2 and TLR4 are dysregulated in patients with PD, and may thus be central to early gut dysfunction in PD. To better understand the putative contribution of intestinal TLR2 and TLR4 dysfunction to early α-synuclein aggregation and PD, we critically discuss the role of TLR2 and TLR4 in normal gut function as well as evidence for altered TLR2 and TLR4 signalling in PD, by reviewing clinical, animal model and in vitro research. Growing evidence on the immunological aetiology of α-synuclein aggregation is also discussed, with a focus on the interactions of α-synuclein with TLR2 and TLR4. We propose a conceptual model of PD pathogenesis in which microbial dysbiosis alters the permeability of the intestinal barrier as well as TLR2 and TLR4 signalling, ultimately leading to a positive feedback loop of chronic gut dysfunction promoting α-synuclein aggregation in enteric and vagal neurons. In turn, α-synuclein aggregates may then migrate to the brain via peripheral nerves, such as the vagal nerve, to contribute to neuroinflammation and neurodegeneration typically associated with PD.
Collapse
Affiliation(s)
- Anastazja M Gorecki
- School of Biological Science, University of Western Australia, Crawley, WA, Australia.
- Neurodegenerative Disorders Research Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.
| | - Chidozie C Anyaegbu
- Curtin Health Innovation Research Institute, Ralph and Patricia Sarich Neuroscience Research Institute, Curtin University, Nedlands, WA, Australia
| | - Ryan S Anderton
- Faculty of Medicine, Nursing and Midwifery and Faculty of Health Sciences, University of Notre Dame Australia, Fremantle, WA, Australia
- School of Nursing, Midwifery, Health Sciences and Physiotherapy, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
24
|
Dogra SK, Cheong Kwong C, Wang D, Sakwinska O, Colombo Mottaz S, Sprenger N. Nurturing the Early Life Gut Microbiome and Immune Maturation for Long Term Health. Microorganisms 2021; 9:2110. [PMID: 34683431 PMCID: PMC8537230 DOI: 10.3390/microorganisms9102110] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
Early life is characterized by developmental milestones such as holding up the head, turning over, sitting up and walking that are typically achieved sequentially in specific time windows. Similarly, the early gut microbiome maturation can be characterized by specific temporal microorganism acquisition, colonization and selection with differential functional features over time. This orchestrated microbial sequence occurs from birth during the first years of age before the microbiome reaches an adult-like composition and function between 3 and 5 years of age. Increasingly, these different steps of microbiome development are recognized as crucial windows of opportunity for long term health, primarily linked to appropriate immune and metabolic development. For instance, microbiome disruptors such as preterm and Cesarean-section birth, malnutrition and antibiotic use are associated with increased risk to negatively affect long-term immune and metabolic health. Different age discriminant microbiome taxa and functionalities are used to describe age-appropriate microbiome development, and advanced modelling techniques enable an understanding and visualization of an optimal microbiome maturation trajectory. Specific microbiome features can be related to later health conditions, however, whether such features have a causal relationship is the topic of intense research. Early life nutrition is an important microbiome modulator, and 'Mother Nature' provides the model with breast milk as the sole source of nutrition for the early postnatal period, while dietary choices during the prenatal and weaning period are to a large extent guided by tradition and culture. Increasing evidence suggests prenatal maternal diet and infant and child nutrition impact the infant microbiome trajectory and immune competence development. The lack of a universal feeding reference for such phases represents a knowledge gap, but also a great opportunity to provide adequate nutritional guidance to maintain an age-appropriate microbiome for long term health. Here, we provide a narrative review and perspective on our current understanding of age-appropriate microbiome maturation, its relation to long term health and how nutrition shapes and influences this relationship.
Collapse
Affiliation(s)
| | | | | | | | | | - Norbert Sprenger
- Nestlé Institute of Health Sciences, Société des Produits Nestlé S.A., 1000 Lausanne 26, Switzerland; (S.K.D.); (K.C.C.); (D.W.); (O.S.); (S.C.M.)
| |
Collapse
|
25
|
Yu LM, Mao LQ, Wu CY, Ye W, Wang X. Chlorogenic acid improves intestinal barrier function by downregulating CD14 to inhibit the NF-κB signaling pathway. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
26
|
Wen Y, Xiao H, Liu Y, Yang Y, Wang Y, Xu S, Huang S, Hou S, Liang J. Polysaccharides from Dendrobium officinale ameliorate colitis-induced lung injury via inhibiting inflammation and oxidative stress. Chem Biol Interact 2021; 347:109615. [PMID: 34363819 DOI: 10.1016/j.cbi.2021.109615] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 01/13/2023]
Abstract
It has been reported that Dendrobium officinale polysaccharides (DOPS) could alleviate colitis in animal model and suppress the activation of NLRP3 inflammasome and β-arrestin1 in vitro. However, it remains unclear whether DOPS has effect on protecting against colitis-induced pulmonary injury. The purpose of this study was to explore the protective effect and mechanism of DOPS on colitis-induced lung injury. A dextran sodium sulfate (DSS)-induced mice colitis model and lipopolysaccharide (LPS)-stimulated BEAS-2B cells model were applied in this study. The results showed that DOPS treatment restored histopathological changes, reduced inflammatory cells infiltration, pro-inflammatory cytokines levels, reactive oxygen species (ROS) formation and MDA generation, and increased anti-oxidative enzymes activities including SOD and GSH-Px in colitis mice. Further investigation showed that DOPS significantly inhibited the protein expression of TLR4, and apparently up-regulated proteins expressions of nuclear-Nrf2, HO-1 and NQO-1 in lung tissues of colitis mice and in BEAS-2B cells. These results indicated that DOPS significantly inhibited inflammation and oxidative stress to alleviate colitis-induced secondary lung injury, and its mechanisms are closely related to the inhibition of TLR4 signaling pathway and the activation of Nrf2 signaling pathway. DOPS may be a promising drug for alleviating colitis-induced lung injury.
Collapse
Affiliation(s)
- Yifan Wen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Hongyu Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Ying Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Yiqi Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, PR China
| | - Yumin Wang
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, School of Chemistry, South China Normal University, Guangzhou, Guangdong, 510006, PR China
| | - Shijie Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China
| | - Song Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| | - Jian Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, PR China.
| |
Collapse
|
27
|
Le Noci V, Bernardo G, Bianchi F, Tagliabue E, Sommariva M, Sfondrini L. Toll Like Receptors as Sensors of the Tumor Microbial Dysbiosis: Implications in Cancer Progression. Front Cell Dev Biol 2021; 9:732192. [PMID: 34604233 PMCID: PMC8485072 DOI: 10.3389/fcell.2021.732192] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/23/2021] [Indexed: 01/02/2023] Open
Abstract
The microbiota is a complex ecosystem of active microorganisms resident in the body of mammals. Although the majority of these microorganisms resides in the distal gastrointestinal tract, high-throughput DNA sequencing technology has made possible to understand that several other tissues of the human body host their own microbiota, even those once considered sterile, such as lung tissue. These bacterial communities have important functions in maintaining a healthy body state, preserving symbiosis with the host immune system, which generates protective responses against pathogens and regulatory pathways that sustain the tolerance to commensal microbes. Toll-like receptors (TLRs) are critical in sensing the microbiota, maintaining the tolerance or triggering an immune response through the direct recognition of ligands derived from commensal microbiota or pathogenic microbes. Lately, it has been highlighted that the resident microbiota influences the initiation and development of cancer and its response to therapies and that specific changes in the number and distribution of taxa correlate with the existence of cancers in various tissues. However, the knowledge of functional activity and the meaning of microbiome changes remain limited. This review summarizes the current findings on the function of TLRs as sensors of the microbiota and highlighted their modulation as a reflection of tumor-associated changes in commensal microbiota. The data available to date suggest that commensal "onco-microbes" might be able to break the tolerance of TLRs and become complicit in cancer by sustaining its growth.
Collapse
Affiliation(s)
- Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Francesca Bianchi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
28
|
Chevalier G, Laveissière A, Desachy G, Barnich N, Sivignon A, Maresca M, Nicoletti C, Di Pasquale E, Martinez-Medina M, Simpson KW, Yajnik V, Sokol H, Plassais J, Strozzi F, Cervino A, Morra R, Bonny C. Blockage of bacterial FimH prevents mucosal inflammation associated with Crohn's disease. MICROBIOME 2021; 9:176. [PMID: 34425887 PMCID: PMC8383459 DOI: 10.1186/s40168-021-01135-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/01/2021] [Indexed: 05/14/2023]
Abstract
BACKGROUND An Escherichia coli (E. coli) pathotype with invasive properties, first reported by Darfeuille-Michaud and termed adherent-invasive E. coli (AIEC), was shown to be prevalent in up to half the individuals with Crohn's Disease (CD), suggesting that these bacteria could be involved in the pathophysiology of CD. Among the genes related to AIEC pathogenicity, fim has the potential to generate an inflammatory reaction from the intestinal epithelial cells and macrophages, as it interacts with TLR4, inducing the production of inflammatory cytokines independently of LPS. Therefore, targeting the bacterial adhesion of FimH-expressing bacteria seems a promising therapeutic approach, consisting of disarming bacteria without killing them, representing a selective strategy to suppress a potentially critical trigger of intestinal inflammation, without disturbing the intestinal microbiota. RESULTS We analyzed the metagenomic composition of the gut microbiome of 358 patients with CD from two different cohorts and characterized the presence of FimH-expressing bacteria. To assess the pathogenic role of FimH, we used human intestinal explants and tested a specific FimH blocker to prevent bacterial adhesion and associated inflammation. We observed a significant and disease activity-dependent enrichment of Enterobacteriaceae in the gut microbiome of patients with CD. Bacterial FimH expression was functionally confirmed in ileal biopsies from 65% of the patients with CD. Using human intestinal explants, we further show that FimH is essential for adhesion and to trigger inflammation. Finally, a specific FimH-blocker, TAK-018, inhibits bacterial adhesion to the intestinal epithelium and prevents inflammation, thus preserving mucosal integrity. CONCLUSIONS We propose that TAK-018, which is safe and well tolerated in humans, is a promising candidate for the treatment of CD and in particular in preventing its recurrence. Video abstract.
Collapse
Affiliation(s)
| | | | | | - Nicolas Barnich
- Université Clermont Auvergne, Inserm U1071, M2iSH, USC-INRA 2018, F-63000, Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, Inserm U1071, M2iSH, USC-INRA 2018, F-63000, Clermont-Ferrand, France
| | - Marc Maresca
- Aix Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Cendrine Nicoletti
- Aix Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Eric Di Pasquale
- Aix-Marseille Université, CNRS, INP, Institut de Neurophysiopathologie, Marseille, France
| | | | | | - Vijay Yajnik
- GI Therapeutic Area Unit, Takeda Pharmaceuticals, Cambridge, MA, 02139, USA
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, 75012, Paris, France
- INRA, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, France
| | | | | | | | - Rachel Morra
- Enterome, 94-96 Avenue Ledru-Rollin, 75011, Paris, France
| | | |
Collapse
|
29
|
Sheikh A, Taube J, Greathouse KL. Contribution of the Microbiota and their Secretory Products to Inflammation and Colorectal Cancer Pathogenesis: The Role of Toll-like Receptors. Carcinogenesis 2021; 42:1133-1142. [PMID: 34218275 DOI: 10.1093/carcin/bgab060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/08/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022] Open
Abstract
Alterations in diversity and function of the gut microbiome are associated with concomitant changes in immune response, including chronic inflammation. Chronic inflammation is a major risk factor for colorectal cancer (CRC). An important component of the inflammatory response system are the toll-like receptors (TLRs). TLRs are capable of sensing microbial components, including nucleic acids, lipopolysaccharides, and peptidoglycans, as well as bacterial outer membrane vesicles (OMV). OMVs can be decorated with or carry as cargo these TLR activating factors. These microbial factors can either promote tolerance or activate signaling pathways leading to chronic inflammation. Herein we discuss the role of the microbiome and the OMVs that originate from intestinal bacteria in promoting chronic inflammation and the development of colitis-associated CRC. We also discuss the contribution of TLRs in mediating the microbiome-inflammation axis and subsequent cancer development. Understanding the role of the microbiome and its secretory factors in TLR response may lead to the development of better cancer therapeutics.
Collapse
Affiliation(s)
- Aadil Sheikh
- Department of Biology, College of Arts and Sciences, Baylor University
| | - Joseph Taube
- Department of Biology, College of Arts and Sciences, Baylor University
| | - K Leigh Greathouse
- Department of Biology, College of Arts and Sciences, Baylor University.,Human Science and Design, Robbins College of Health and Human Sciences, Baylor University
| |
Collapse
|
30
|
Vissenaekens H, Criel H, Grootaert C, Raes K, Smagghe G, Van Camp J. Flavonoids and cellular stress: a complex interplay affecting human health. Crit Rev Food Sci Nutr 2021; 62:8535-8566. [PMID: 34098806 DOI: 10.1080/10408398.2021.1929822] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Flavonoid consumption has beneficial effects on human health, however, clinical evidence remains often inconclusive due to high interindividual variability. Although this high interindividual variability has been consistently observed in flavonoid research, the potential underlying reasons are still poorly studied. Especially the knowledge on the impact of health status on flavonoid responsiveness is limited and merits more investigation. Here, we aim to highlight the bidirectional interplay between flavonoids and cellular stress. First, the state-of-the-art concerning inflammatory stress and mitochondrial dysfunction is reviewed and a comprehensive overview of recent in vitro studies investigating the impact of flavonoids on cellular stress, induced by tumor necrosis factor α, lipopolysaccharide and mitochondrial stressors, is given. Second, we critically discuss the influence of cellular stress on flavonoid uptake, accumulation, metabolism and cell responses, which has, to our knowledge, never been extensively reviewed before. Next, we advocate the innovative insight that stratification of the general population based on health status can reveal subpopulations that benefit more from flavonoid consumption. Finally, suggestions are given for the development of future cell models that simulate the physiological micro-environment, including interindividual variability, since more mechanistic research is needed to establish scientific-based personalized food recommendations for specific subpopulations.
Collapse
Affiliation(s)
- Hanne Vissenaekens
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.,Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Hanne Criel
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Charlotte Grootaert
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Katleen Raes
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - John Van Camp
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
31
|
Effect of probiotic Lactobacillus rhamnosus by-products on gingival epithelial cells challenged with Porphyromonas gingivalis. Arch Oral Biol 2021; 128:105174. [PMID: 34058722 DOI: 10.1016/j.archoralbio.2021.105174] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Probiotics are usually given as living cells, but their effects may be also achieved by postbiotics. We hypothesized that probiotics products (spent media and lysate) altered the response induced by P. gingivalis in gingival epithelial cells (GECS). METHODS Immortalized human OBA-9 GECs (∼2,5 × 105cells/well) were challenged with P. gingivalis ATCC33277, and co-infected with L. rhamnosus Lr-32 for 4 h. L. rhamnosus Lr-32 spent medium or cells lysate was added to GECs co-infected with P. gingivalis. Another set of OBA-9 GECs were first exposed to P. gingivalis ATCC 33277 and then to the living probiotic or probiotic products. Transcription of genes encoding inflammatory mediators (IL-1β, TNF-α, IL-6, and CXCL-8) and receptors (TLR2 and TLR4) were evaluated by RT-qPCR. P. gingivalis growth under L. rhamnosus Lr-32 postbiotics was also evaluated. RESULTS L. rhamnosus Lr-32 spent media decreased cell viability, while living cells and cell lysates did not. L. rhamnosus Lr-32 lysate, but not spent media, upregulated transcription of inflammatory mediators (IL-1β, TNF-α, IL-6, and CXCL-8) in GECs infected with P. gingivalis. Transcription of TRL2 was upregulated in all experimental groups compared to control, whereas TLR4 was upregulated by the probiotic or its postbiotics in P. gingivalis infected cells. Spent media and lysates reduced the growth of P. gingivalis. CONCLUSION L. rhamnosus Lr-32 cell components rather than live probiotic enhanced the expression of inflammatory mediators in P. gingivalis infected gingival epithelial cells. The increased potential of Lr-32 cell lysates to promote immune response to the periodontopathogen may favor pathogen elimination but may also lead to additional deleterious effects of the exacerbated inflammation.
Collapse
|
32
|
Rønning SB, Voldvik V, Bergum SK, Aaby K, Borge GIA. Ellagic acid and urolithin A modulate the immune response in LPS-stimulated U937 monocytic cells and THP-1 differentiated macrophages. Food Funct 2021; 11:7946-7959. [PMID: 32832941 DOI: 10.1039/c9fo03008e] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dietary polyphenols are subjected, following ingestion, to an extensive metabolism, and the molecules that act at the cellular and tissue level will be, most likely, metabolites rather than native polyphenols. The mechanisms behind the positive effects exerted by polyphenols are not yet completely elucidated, since most in vitro studies use unmetabolised polyphenols rather than the metabolites present in the body. The aim of this study was to investigate and compare the potential effect of phenolic metabolites on the immune response using U937 monocyte and THP-1 macrophage cell cultures. Of the 16 metabolites tested, urolithins (Uro), and Uro A, in particular were the most potent, showing a modest increase in basal NF-κB activity and a reduction in lipopolysaccaride (LPS)-induced NF-κB activity, gene expression and secretion of pro-inflammatory cytokines. Protocatechuic acid and its sulfate/glucuronide metabolites reduced LPS-induced NF-κB activity, but not IL-6 and TNF-α cytokine secretion. Interestingly, both ellagic acid and its metabolite Uro A had immunomodulating effects, although they regulated the immune response differently, and both reduced LPS-induced NF-κB activity in U937 cells. However, while Uro A dramatically reduced IL-6 and IL-10 mRNA expression, no effect could be observed with ellagic acid. In THP-1 cells, treatment with ellagic acid dramatically reduced the expression of Toll-like receptor 4, while Uro A had no effect. The dual role observed for Uro A, showing both a modest increase in basal NF-κB activity and a reduction in LPS-induced NF-κB activity, as well as a reduction in LPS-induced pro-inflammatory cytokine secretion, makes this metabolite particularly interesting for further studies in animals and humans.
Collapse
|
33
|
Marescotti D, Lo Sasso G, Guerrera D, Renggli K, Ruiz Castro PA, Piault R, Jaquet V, Moine F, Luettich K, Frentzel S, Peitsch MC, Hoeng J. Development of an Advanced Multicellular Intestinal Model for Assessing Immunomodulatory Properties of Anti-Inflammatory Compounds. Front Pharmacol 2021; 12:639716. [PMID: 33935729 PMCID: PMC8085553 DOI: 10.3389/fphar.2021.639716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Intestinal inflammation is the collective term for immune system-mediated diseases of unknown, multifactorial etiology, with often complex interactions between genetic and environmental factors. To mechanistically investigate the effect of treatment with compounds possessing immunomodulating properties in the context of intestinal inflammation, we developed an immunocompetent in vitro triculture intestinal model consisting of a differentiated intestinal epithelial layer (Caco-2/HT29-MTX) and immunocompetent cells (differentiated THP-1). The triculture mimicked a healthy intestine with stable barrier integrity. Lipopolysaccharide treatment triggered a controlled and reversible inflammatory state, resulting in significant impairment of barrier integrity and release of pro-inflammatory cytokines and chemokines, which are known hallmarks of intestinal inflammation. Treatment with known anti-inflammatory reference compounds (TPCA-1 and budenoside) prevented the induction of an inflammatory state; the decreasing triculture responses to this treatment measured by cytokine release, transepithelial electric resistance (TEER), and epithelial layer permeability proved the suitability of the intestinal model for anti-inflammatory drug screening. Finally, selected tobacco alkaloids (nicotine and anatabine (R/S and S forms)) were tested in the in vitro triculture for their potential anti-inflammatory properties. Indeed, naturally occurring alkaloids, such as tobacco-derived alkaloids, have shown substantial anti-inflammatory effects in several in vitro and in vivo models of inflammation, gaining increasing interest. Similar to the anti-inflammatory reference compounds, one of the tobacco alkaloids under investigation partially prevented the decrease in the TEER and increase in permeability and reduced the release of pro-inflammatory cytokines and chemokines. Taken together, these data confirm that our in vitro model is suitable for screening potential anti-inflammatory compounds in the context of intestinal inflammation.
Collapse
Affiliation(s)
| | | | - Diego Guerrera
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Kasper Renggli
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | | | - Romain Piault
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Vincent Jaquet
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Fabian Moine
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Karsta Luettich
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Stefan Frentzel
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | | | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| |
Collapse
|
34
|
Bagherzadeh A, Vaziri H, Nasimi FS, Ahmadian S, Feyzi A, Farhadi M, Yahyavi F, Hashemi B, Rahbarghazi R, Mahdipour M. Bacterial Lipase Neutralized Toxicity of Lipopolysaccharide on Chicken Embryo Cardiac Tissue. Cardiovasc Toxicol 2021; 21:582-591. [PMID: 33856644 DOI: 10.1007/s12012-021-09651-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/07/2021] [Indexed: 11/30/2022]
Abstract
It has been shown that near all organs, especially the cardiovascular system, are affected by bacterial lipopolysaccharide via the activation of Toll-like receptor signaling pathways. Here, we tried to find the blunting effect of bacterial lipase on lipopolysaccharide (LPS)-induced cardiac tissue toxicity in chicken embryos. 7-day fertilized chicken eggs were divided randomly into different groups as follows; Control, Normal Saline, LPS (0.1, 0.5 and 1 mg/kbw), and LPS (0.1, 0.5 and 1 mg/kbw) plus 5 mg/ml Lipase. On day 17, the hearts were sampled. The expression of genes such as GATA4, NKX2.5, EGFR, TRIF, and NF-ƙB was monitored using real-time PCR analysis. Using western blotting, we measured NF-ƙB protein level. Total antioxidant capacity, glutathione peroxidase, and Catalase activity were also studied. Microvascular density and anterior wall thickness were monitored in histological samples using H&E staining. High dose of LPS (1 mg/kbw) increased the expression of TRIF but not NF-ƙB compared to the control group (p < 0.05). We found a statistically significant reduction in groups that received LPS + Lipase compared to the control and LPS groups (p < 0.05). Western blotting revealed that the injection of Lipase could reduce LPS-induced NF-ƙB compared to the control group (p < 0.05). The expression of GATA4, NKx2.5, and EGFR was not altered in the LPS group, while the simultaneous application of LPS and Lipase significantly reduced GATA4, NKx2.5, and EGFR levels below the control (p < 0.05). We found non-significant differences in glutathione peroxidase, and Catalase activity in all groups (p > 0.05), while total antioxidant capacity was increased in groups that received LPS + Lipase. Anterior wall thickness was diminished in LPS groups and the use of both lipase and LPS returned near-to-control values (p < 0.05). Despite a slight increase in microvascular density, we found statistically non-significant differences in all groups (p > 0.05). Bacterial lipase reduces detrimental effects of LPS on chicken embryo heart induced via Toll-like receptor signaling pathway.
Collapse
Affiliation(s)
| | - Hamidreza Vaziri
- Department of Biology, Faculty of Science, Guilan University, Rasht, Iran
| | | | - Shahin Ahmadian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Feyzi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Mehrdad Farhadi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Yahyavi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Hashemi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
35
|
Sharma N, Akhade AS, Ismaeel S, Qadri A. Serum-borne lipids amplify TLR-activated inflammatory responses. J Leukoc Biol 2021; 109:821-831. [PMID: 32717772 DOI: 10.1002/jlb.3ab0720-241rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
TLRs recognize conserved pathogen associated molecular patterns and generate innate immune responses. Several circulating and cell membrane associated proteins have been shown to collaborate with TLRs in sensing microbial ligands and promoting inflammatory responses. Here, we show that serum and serum-borne lipids including lysophosphatidylcholine (LPC) amplify inflammatory responses from intestinal epithelial cells and mononuclear phagocytes primed with microbial TLR ligands. Treatment with the inhibitors of G protein-coupled receptor (GPCR) signaling, suramin, or pertussis toxin (PT), the inhibitor of JNK-MAPK, or knockdown of LPC response-regulating GPCR, G2A, decreases the augmentation brought about by serum or LPC in TLR-induced inflammatory response. In vivo administration of PT or anti-G2A antibody reduces TLR2-activated cytokine secretion. The ability of host lipids to costimulate TLR-generated cellular responses represents a novel pathway for the amplification of innate immunity and inflammation.
Collapse
Affiliation(s)
- Naveen Sharma
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
- Department of Immunology, The University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | - Ajay Suresh Akhade
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
- Institute for Systems Biology, Seattle, Washington, USA
| | - Sana Ismaeel
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
36
|
Zhang P, Nguyen J, Abdulla F, Nelson AT, Beckman JD, Vercellotti GM, Belcher JD. Soluble MD-2 and Heme in Sickle Cell Disease Plasma Promote Pro-Inflammatory Signaling in Endothelial Cells. Front Immunol 2021; 12:632709. [PMID: 33841413 PMCID: PMC8033004 DOI: 10.3389/fimmu.2021.632709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/10/2021] [Indexed: 12/22/2022] Open
Abstract
Recent evidence indicates that hemolysis in sickle cell disease (SCD) promotes inflammation via innate immune signaling through toll-like receptor 4 (TLR4). Free heme released by hemolyzed red blood cells can bind to myeloid differentiation factor-2 (MD-2) and activate TLR4 pro-inflammatory signaling on endothelium to promote vaso-occlusion and acute chest syndrome in murine models of SCD. MD-2 is co-expressed with TLR4 on cell membranes, but in inflammatory conditions, soluble MD-2 (sMD-2) is elevated in plasma. sMD-2 levels were significantly increased in human and murine sickle (SS) plasma as compared to normal (AA) plasma. Human umbilical vein endothelial cells (HUVEC) and human lung microvascular endothelial cells incubated with human SS plasma had significant increases in pro-inflammatory IL-8, IL-6, and soluble VCAM-1 secretion compared to endothelial cells incubated with AA plasma. The increase in HUVEC IL-8 secretion was blocked by depletion of sMD-2 from SS plasma and enhanced by the addition of sMD-2 to AA plasma. The TLR4 signaling inhibitor, TAK-242, inhibited HUVEC IL-8 secretion in response to SS plasma by 85%. Heme-agarose pull-down assays and UV/Vis spectroscopy demonstrated that heme binds to sMD-2. Hemopexin, a high affinity heme-binding protein, inhibited HUVEC IL-8 secretion induced by SS plasma or SS and AA plasma supplemented with sMD-2. These data suggest that sMD-2 bound to heme might play an important role in pro-inflammatory signaling by endothelium in SCD.
Collapse
Affiliation(s)
- Ping Zhang
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Julia Nguyen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Fuad Abdulla
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Alexander T Nelson
- University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Joan D Beckman
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - John D Belcher
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
37
|
Trevisi P, Luise D, Correa F, Messori S, Mazzoni M, Lallès JP, Bosi P. Maternal antibiotic treatment affects offspring gastric sensing for umami taste and ghrelin regulation in the pig. J Anim Sci Biotechnol 2021; 12:31. [PMID: 33731211 PMCID: PMC7972225 DOI: 10.1186/s40104-021-00557-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/13/2021] [Indexed: 11/18/2022] Open
Abstract
Background Scarce is knowledge on the process regulating the development of acid secretion, orexigenic signaling, and chemosensing in the stomach of young pigs. Changes of early microbial encounters by suckling pigs can interact with the gut maturation, by the induction of different molecular signaling. Our goal was to assess if the age of offspring and the maternal environment, influenced by sow antibiotic treatment peripartum, could affect gastric morphology and the expression of genes involved in the control of hydrochloric secretion, feed intake, taste, and inflammation in offspring stomach. Methods 84 pigs from sows fed a diet with amoxicillin (on –d10 to +d21 from farrowing, ANT) or without (CON) were sacrificed at d14, d21, d28 (weaning) or d42. Samples of oxyntic (OXY), pyloric (PY) and cardiac mucosae close to OXY were collected and parietal and enteroendocrine cells (EECs) were counted. Relative gene expression of a set of 11 key genes (ATP4A, SSTR2, GAST, GHRL, MBOAT4, PCSK1, GNAT1, TAS1R1, TAS1R3, IL8 and TNF) was assessed by qRT-PCR. In addition, 40 offspring obtained from the same ANT and CON sows were offered a normal or a fat-enriched diet for 4 weeks between 140 and 169 d of age, and then OXY and PY were sampled. Results The number of parietal and EECs increased with age (P < 0.001). ATP4A increased with age (within suckling, P = 0.043, post-weaning vs. suckling, P < 0.001), SSTR2 increased only after weaning (P < 0.001). In OXY, GHRL increased during suckling (P = 0.012), and post-weaning as a trend (P = 0.088). MBOAT4 tended to increase during suckling (P = 0.062). TAS1R1 increased from suckling to post-weaning period (P =0.001) and was lower in ANT offspring (P = 0.013). GNAT1 in PY was higher in ANT offspring (P = 0.041). Antibiotic treatment of sows peripartum increased expression of GHRL and MBOAT4 in OXY of growing-finishing offspring aged 5 months. Conclusions Data show that sensing for umami taste and ghrelin regulation can be affected by maternal environment, but the development of acid secretion, orexigenic signaling and taste perception in the stomach are mostly developmentally controlled. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-021-00557-3.
Collapse
Affiliation(s)
- P Trevisi
- Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - D Luise
- Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - F Correa
- Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - S Messori
- Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy.,Present Address: World Organisation for Animal Health (OIE), Scientific Secretariat for the STAR-IDAZ International Research Consortium on animal health, 12 rue de Prony, 75017, Paris, France
| | - M Mazzoni
- Department of Veterinary Sciences, University of Bologna, Via Tolara di Sopra, Ozzano nell'Emilia (BO), Bologna, Italy
| | - J P Lallès
- INRAE, Human Nutrition Division, Site of Theix, 63122, Saint-Genès-Champanelle, France
| | - P Bosi
- Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy.
| |
Collapse
|
38
|
Lorente-Picón M, Laguna A. New Avenues for Parkinson's Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021; 11:433. [PMID: 33804226 PMCID: PMC7998286 DOI: 10.3390/biom11030433] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder that currently affects 1% of the population over the age of 60 years, and for which no disease-modifying treatments exist. Neurodegeneration and neuropathology in different brain areas are manifested as both motor and non-motor symptoms in patients. Recent interest in the gut-brain axis has led to increasing research into the gut microbiota changes in PD patients and their impact on disease pathophysiology. As evidence is piling up on the effects of gut microbiota in disease development and progression, another front of action has opened up in relation to the potential usage of microbiota-based therapeutic strategies in treating gastrointestinal alterations and possibly also motor symptoms in PD. This review provides status on the different strategies that are in the front line (i.e., antibiotics; probiotics; prebiotics; synbiotics; dietary interventions; fecal microbiota transplantation, live biotherapeutic products), and discusses the opportunities and challenges the field of microbiome research in PD is facing.
Collapse
Affiliation(s)
- Marina Lorente-Picón
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ariadna Laguna
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
39
|
Chang AM, Kantrong N, Darveau RP. Maintaining homeostatic control of periodontal epithelial tissue. Periodontol 2000 2021; 86:188-200. [PMID: 33690934 DOI: 10.1111/prd.12369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Years of coevolution with resident microbes has made them an essential component of health. Yet, little is known about oral commensal bacteria's contribution to and role in the maintenance of oral health and homeostasis. Commensal bacteria are speculated to play a host protective role in the maintenance of health. In this review, we describe and provide examples of the coordinate regulation that occurs between oral commensal bacteria and the host innate immune response to modulate and maintain oral homeostasis.
Collapse
Affiliation(s)
- Ana M Chang
- Department of Periodontics, University of Washington, Seattle, Washington, USA
| | - Nutthapong Kantrong
- Department of Periodontics, University of Washington, Seattle, Washington, USA.,Oral Biology Research Unit, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Richard P Darveau
- Department of Periodontics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
40
|
Association between disease-related malnutrition and innate immunity gene expression in critically ill patients at intensive care unit admission. Cent Eur J Immunol 2021; 45:414-424. [PMID: 33658890 PMCID: PMC7882404 DOI: 10.5114/ceji.2020.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 11/17/2022] Open
Abstract
The aim of the study was to analyse the relationship between nutritional disorders and the expression of innate antibacterial response genes in patients admitted to the intensive care unit (ICU). In 46 patients with severe malnutrition and life-threatening surgical complications, nutritional status tests were performed on the basis of the NRS 2002 (Nutritional Risk Screening) scale, cytokine, albumin, C-reactive protein concentrations, anthropometric tests, and body composition analysis. Concurrently, the expression of Toll-like receptor 2, NOD1, TRAF6, and HMGB1 genes was determined in peripheral blood leukocytes at the mRNA level using real-time polymerase chain reaction. It was found that both the nutritional status and the gene expression changed depending on the group of patients studied (including the group of survivors vs. non-survivors). Significant correlations were found between the results of routine tests used in the diagnostics of malnutrition (including NRS 2002, resistance, reactance, phase angle, excess of extracellular water) and the expression of the studied genes. Moreover, the expression of TRAF6 and HMGB1 genes correlated with the Acute Physiology and Chronic Health Evaluation II scale and the age of the patients. The results of the research suggest that the expression of innate antibacterial response genes may be a new diagnostic tool complementing the assessment of nutritional disorders in surgical patients admitted to the ICU. These tests may be helpful in providing more accurate diagnostics of the genetic effects of malnutrition and in the monitoring of patients for whom nutritional treatment is planned to support the functions of the immune system, thereby increasing the effectiveness of this type of treatment in the ICU.
Collapse
|
41
|
Vyas P, Tulsawani R, Vohora D. Methods for the Screening of New Chemical Entities for Deciphering Neuroinflammatory and Associated Pathways in Seizures: An In Vitro Perspective. NEUROMETHODS 2021:29-53. [DOI: 10.1007/978-1-0716-1254-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Tam JSY, Coller JK, Hughes PA, Prestidge CA, Bowen JM. Toll-like receptor 4 (TLR4) antagonists as potential therapeutics for intestinal inflammation. Indian J Gastroenterol 2021; 40:5-21. [PMID: 33666891 PMCID: PMC7934812 DOI: 10.1007/s12664-020-01114-y] [Citation(s) in RCA: 33] [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] [Received: 08/13/2020] [Accepted: 10/27/2020] [Indexed: 02/04/2023]
Abstract
Gastrointestinal inflammation is a hallmark of highly prevalent disorders, including cancer treatment-induced mucositis and ulcerative colitis. These disorders cause debilitating symptoms, have a significant impact on quality of life, and are poorly managed. The activation of toll-like receptor 4 (TLR4) has been proposed to have a major influence on the inflammatory signalling pathways of the intestinal tract. Inhibition of TLR4 has been postulated as an effective way to treat intestinal inflammation. However, there are a limited number of studies looking into the potential of TLR4 antagonism as a therapeutic approach for intestinal inflammation. This review surveyed available literature and reported on the in vitro, ex vivo and in vivo effects of TLR4 antagonism on different models of intestinal inflammation. Of the studies reviewed, evidence suggests that there is indeed potential for TLR4 antagonists to treat inflammation, although only a limited number of studies have investigated treating intestinal inflammation with TLR4 antagonists directly. These results warrant further research into the effect of TLR4 antagonists in the intestinal tract.
Collapse
Affiliation(s)
- Janine S. Y. Tam
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5005 Australia
| | - Janet K. Coller
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, South Australia Australia
| | - Patrick A. Hughes
- Centre for Nutrition and Gastrointestinal Diseases, Adelaide Medical School, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Clive A. Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia Australia ,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Melbourne, Australia
| | - Joanne M. Bowen
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5005 Australia
| |
Collapse
|
43
|
Semin I, Ninnemann J, Bondareva M, Gimaev I, Kruglov AA. Interplay Between Microbiota, Toll-Like Receptors and Cytokines for the Maintenance of Epithelial Barrier Integrity. Front Med (Lausanne) 2021; 8:644333. [PMID: 34124086 PMCID: PMC8194074 DOI: 10.3389/fmed.2021.644333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal tract is densely populated by microbiota consisting of various commensal microorganisms that are instrumental for the healthy state of the living organism. Such commensals generate various molecules that can be recognized by the Toll-like receptors of the immune system leading to the inflammation marked by strong upregulation of various proinflammatory cytokines, such as TNF, IL-6, and IL-1β. To prevent excessive inflammation, a single layer of constantly renewing, highly proliferating epithelial cells (IEC) provides proper segregation of such microorganisms from the body cavities. There are various triggers which facilitate the disturbance of the epithelial barrier which often leads to inflammation. However, the nature and duration of the stress may determine the state of the epithelial cells and their responses to cytokines. Here we discuss the role of the microbiota-TLR-cytokine axis in the maintenance of the epithelial tissue integrity. In particular, we highlight discrepancies in the function of TLR and cytokines in IEC barrier during acute or chronic inflammation and we suggest that intervention strategies should be applied based on the type of inflammation.
Collapse
Affiliation(s)
- Iaroslav Semin
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Justus Ninnemann
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Marina Bondareva
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ilia Gimaev
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey A. Kruglov
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- *Correspondence: Andrey A. Kruglov
| |
Collapse
|
44
|
Sobhani I, Rotkopf H, Khazaie K. Bacteria-related changes in host DNA methylation and the risk for CRC. Gut Microbes 2020; 12:1800898. [PMID: 32931352 PMCID: PMC7575230 DOI: 10.1080/19490976.2020.1800898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 02/03/2023] Open
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer deaths in men and women combined. Colon-tumor growth is multistage and the result of the accumulation of spontaneous mutations and epigenetic events that silence tumor-suppressor genes and activate oncogenes. Environmental factors are primary contributors to these somatic gene alterations, which account for the increase in incidence of CRC in western countries. In recent decades, gut microbiota and their metabolites have been recognized as essential contributing factors to CRC, and now serve as biomarkers for the diagnosis and prognosis of CRC. In the present review, we highlight holistic approaches to understanding how gut microbiota contributes to CRC. We particularly focus herein on bacteria-related changes in host DNA methylation and the risk for CRC.
Collapse
Affiliation(s)
- Iradj Sobhani
- Head of the Department of Gastroenterology, Consultant in GI Oncology, Hopital Henri Mondor, APHP. Créteil-France; Head of the Research Team EC2M3, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Hugo Rotkopf
- Department of Gastroenterology Hospital Henri Mondor, APHP. Créteil-France; Member of Research Team EC2M3, Université Paris-Est Créteil (UPEC). Créteil, France
| | | |
Collapse
|
45
|
Holani R, Babbar A, Blyth GAD, Lopes F, Jijon H, McKay DM, Hollenberg MD, Cobo ER. Cathelicidin-mediated lipopolysaccharide signaling via intracellular TLR4 in colonic epithelial cells evokes CXCL8 production. Gut Microbes 2020; 12:1785802. [PMID: 32658599 PMCID: PMC7524372 DOI: 10.1080/19490976.2020.1785802] [Citation(s) in RCA: 20] [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] [Indexed: 02/07/2023] Open
Abstract
We hypothesized that the antimicrobial peptide cathelicidin has a physiological role in regulating gut inflammatory homeostasis. We determined that cathelicidin synergizes with LPS to facilitate its internalization and signaling via endosomic TLR4 in colonic epithelium, evoking synthesis of the human neutrophil chemoattractant, CXCL8 (or murine homolog, CXCL1). Interaction of cathelicidin with LPS in the control of CXCL8/CXCL1 synthesis was assessed in human colon epithelial cells, murine colonoids and cathelicidin-null mice (Camp-/- ). Mechanistically, human cathelicidin (LL-37), as an extracellular complex with LPS, interacted with lipid raft-associated GM1 gangliosides to internalize and activate intracellular TLR4. Two signaling pathways converged on CXCL8/CXCL1 production: (1) a p38MAPK-dependent pathway regulated by Src-EGFR kinases; and, (2) a p38MAPK-independent, NF-κB-dependent pathway, regulated by MEK1/2-MAPK. Increased cathelicidin-dependent CXCL8 secretion in the colonic mucosa activated human blood-derived neutrophils. These cathelicidin effects occurred in vitro at concentrations well below those needed for microbicidal function. The important immunomodulatory role of cathelicidins was evident in cathelicidin-null/Camp-/- mice, which had diminished colonic CXCL1 secretion, decreased neutrophil recruitment-activation and reduced bacterial clearance when challenged with the colitis-inducing murine pathogen, Citrobacter rodentium. We conclude that in addition to its known microbicidal action, cathelicidin has a unique pathogen-sensing role, facilitating LPS-mediated intestinal responses, including the production of CXCL8/CXCL1 that would contribute to an integrated tissue response to recruit neutrophils during colitis.
Collapse
Affiliation(s)
- Ravi Holani
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Anshu Babbar
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Graham A. D. Blyth
- Microbiology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Fernando Lopes
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Humberto Jijon
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Derek M. McKay
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Morley D. Hollenberg
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Eduardo R. Cobo
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada,CONTACT Eduardo R. Cobo Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, ABT2N 4N1, Canada
| |
Collapse
|
46
|
Camacho JH, Rusinky Pinilla L, Salazar Peña D, Sanabria Dueñas S, Rojas Carvajal D, Burbano Castillo N, Ruiz Peña R, Palacino Saenz I, Martínez Quesada S, García Salazar A, Abdala Galvis N. Microbiota intestinal en pediatría. REPERTORIO DE MEDICINA Y CIRUGÍA 2020. [DOI: 10.31260/repertmedcir.01217372.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
La microbiota intestinal es el conjunto de millones de microrganismos vivos ubicados en el tracto gastrointestinal. Es indispensable en múltiples funciones del organismo, regulación de la inmunidad, en aspectos nutricionales y procesos de inflamación sistémica entre otros. La disbiosis es la alteración del equilibrio de la microbiota normal, debido a cambios en la composición, funcionamiento, orden o su distribución; esto puede predisponer al individuo a la adquisición de enfermedades gastrointestinales, alérgicas y metabólicas, entre otras. El objetivo del presente artículo es realizar una revisión narrativa de la literatura sobre los conceptos claves de la microbiota intestinal, sus asociaciones fisiopatológicas con desórdenes gastrointestinales, alérgicos y metabólicos en pediatría.
Collapse
|
47
|
Beneficial Effect of a Fermented Wheat Germ Extract in Intestinal Epithelial Cells in case of Lipopolysaccharide-Evoked Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1482482. [PMID: 32849997 PMCID: PMC7436289 DOI: 10.1155/2020/1482482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/29/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022]
Abstract
In this study, the protective effect of a fermented wheat germ extract (FWGE) against LPS-induced inflammation and oxidative stress in IPEC-J2 porcine intestinal epithelial cells was studied. Enterocytes were treated with LPS derived from Salmonella enterica ser. Typhimurium and Escherichia coli O55:B5, O111:B4, and O127:B8 strains. Intracellular ROS level and extracellular H2O2 level were followed up by two fluorescent assays (DCFH-DA and Amplex Red). The effect of FWGE on the intestinal barrier integrity was determined by transepithelial electric resistance measurements and using a FD4 fluorescent tracer dye. IL-6 concentration of supernatants was also measured by the ELISA method. Our data revealed that FWGE had a significant lowering effect on the inflammatory response especially related to oxidative stress. Treatment with FWGE (1-2%) significantly decreased the level of intracellular ROS compared to LPS-treated cells. Furthermore, LPS-triggered partial disruption of epithelial integrity was reduced after FWGE application.
Collapse
|
48
|
Oliveira LP, Guimarães VHD, Oliveira JR, Guimarães ALS, de Paula AMB, Bader M, Santos RASD, Santos SHS. Genetic deletion of the angiotensin-(1-7) receptor Mas leads to alterations in gut villi length modulating TLR4/PI3K/AKT and produces microbiome dysbiosis. Neuropeptides 2020; 82:102056. [PMID: 32505463 DOI: 10.1016/j.npep.2020.102056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/17/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023]
Abstract
Renin-Angiotensin System (RAS) is an important peptide cascade involved in physiological processes. RAS homeostasis disruption produces several cardiovascular and metabolic disorders, such as arterial hypertension, atherosclerosis, acute myocardial infarct, obesity, diabetes, metabolic syndrome and increases gastrointestinal tract (GIT) cell proliferation. Angiotensin (Ang)-(1-7) peptide is the main RAS counter-regulatory axis effector. It is formed from ACE2 enzyme and acts mainly through Mas receptor (MasR). In this context, the aim of the present study was to evaluate alterations in small intestine morphology and intestinal microbiota composition in MasR knockout C57BL/6 mice. We analyzed glucose tolerance; insulin sensitivity and blood collected for biochemical parameters as well as small intestine tissues samples for immunohistochemistry. mRNA and bacteria gDNA expression evaluation. mRNA expression was evaluated by qRT-PCR for TLR4, PI3K and AKT. The main results showed that Mas-R-knockout mice presented lower body weight. MasR-knockout mice also presented increased fasted blood glucose and total cholesterol with reduced HDL, lower glucose tolerance and impaired insulin sensitivity. Increased intestinal mucosa length, increased intestinal villi, reduced Lieberkühn crypt depth. The increased expression of cell proliferation markers Ki-67 and Cyclin D1 and increased TLR4, PI3K and AKT expressions were observed with augmented Bacteroidetes and decreased amount of Firmicutes. That results suggests that MasR deletion generated changes in intestinal microbiota, possibly due to a lower neutral amino acids absorption followed by a compensatory increase in intestinal villi length associated with disbiosis and LPS overproduction that ultimately lead to proliferation and cell inflammation.
Collapse
Affiliation(s)
- Luis Paulo Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Victor Hugo Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Janaina Ribeiro Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - André Luiz Sena Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Alfredo Maurício Batista de Paula
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Robson Augusto Souza Dos Santos
- Institute of Biological Sciences (ICB), Physiology Department, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences (ICA), Food Engineering, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
49
|
Bruning EE, Coller JK, Wardill HR, Bowen JM. Site-specific contribution of Toll-like receptor 4 to intestinal homeostasis and inflammatory disease. J Cell Physiol 2020; 236:877-888. [PMID: 32730645 DOI: 10.1002/jcp.29976] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Toll-like receptor 4 (TLR4) is a highly conserved protein of innate immunity, responsible for the regulation and maintenance of homeostasis, as well as immune recognition of external and internal ligands. TLR4 is expressed on a variety of cell types throughout the gastrointestinal tract, including on epithelial and immune cell populations. In a healthy state, epithelial cell expression of TLR4 greatly assists in homeostasis by shaping the host microbiome, promoting immunoglobulin A production, and regulating follicle-associated epithelium permeability. In contrast, immune cell expression of TLR4 in healthy states is primarily centred on the maturation of dendritic cells in response to stimuli, as well as adequately priming the adaptive immune system to fight infection and promote immune memory. Hence, in a healthy state, there is a clear distinction in the site-specific roles of TLR4 expression. Similarly, recent research has indicated the importance of site-specific TLR4 expression in inflammation and disease, particularly the impact of epithelial-specific TLR4 on disease progression. However, the majority of evidence still remains ambiguous for cell-specific observations, with many studies failing to provide the distinction of epithelial versus immune cell expression of TLR4, preventing specific mechanistic insight and greatly impacting the translation of results. The following review provides a critical overview of the current understanding of site-specific TLR4 activity and its contribution to intestinal/immune homeostasis and inflammatory diseases.
Collapse
Affiliation(s)
- Elise E Bruning
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Janet K Coller
- Discipline of Pharmacology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Hannah R Wardill
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia.,Department of Paediatric Oncology/Haematology, The University of Groningen (University Medical Centre Groningen), Groningen, The Netherlands
| | - Joanne M Bowen
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
50
|
Abstract
Dysfunction of the gut-blood barrier plays an important role in many diseases, such as inflammatory bowel disease, hemorrhagic shock (HS), or burn injury. However, little is known about gut barrier dysfunction after hemodynamically instable polytrauma (PT). Therefore, we aimed to evaluate the effects of PT and HS on remote intestinal damage and barrier dysfunction, especially regarding the role of zonula occludens protein 1 (ZO-1) as an important tight junction protein.Male C57BL/6 mice were subjected to either PT (thorax trauma, closed head injury, soft tissue injury, and distal femoral fracture), 60 min of pressure-controlled HS (30 ± 5 mmHg), or PT+HS, or sham procedures.Animals of all trauma groups showed an increase in abdominal girth and dilation of the intestine during the experimental period, which was largest in the PT+HS group. Increased blood-tissue permeability to albumin (assessed by Evans blue dye) was found in the HS group. Experimental groups showed a slight increase in plasma concentration of intestinal fatty acid binding protein and some intestinal damage was histologically detectable. Of note, PT+HS animals revealed significantly reduced expression of ZO-1 in intestinal epithelial cells. In an in-vitro model, stimulation of human colon epithelial cells with peptidoglycan, but not with lipopolysaccharide, resulted in elevated secretion of pro-inflammatory cytokines, reflecting inflammatory activity of the intestinal epithelium.Taken together, PT and HS lead to increased permeability of the gut-blood barrier. Bacterial components may lead to production of inflammatory and chemotactic mediators by gut epithelial cells, underlining the role of the gut as an immunologically active organ.
Collapse
|