1
|
Pieszka M, Szczepanik K, Łoniewski I. Utilizing pigs as a model for studying intestinal barrier function. ANNALS OF ANIMAL SCIENCE 2024. [DOI: 10.2478/aoas-2024-0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Intestinal permeability has been extensively studied, particularly in gastrointestinal diseases such as inflammatory bowel disease, food allergy, visceral disease, celiac disease, and Crohn’s disease. These studies have established that changes in intestinal permeability contribute to the pathogenesis of many gastrointestinal and systemic diseases. While numerous works in the 20th century focused on this topic, it remains relevant for several reasons. Despite the development of new research techniques, it is still unclear whether changes in intestinal permeability are the primary mechanism initiating the disease process or if they occur secondary to an ongoing chronic inflammatory process. Investigating the possibility of stabilizing the intestinal barrier, thereby reducing its permeability preemptively to prevent damage and after the damage has occurred, may offer new therapeutic approaches. Increased intestinal permeability is believed to lead to reduced nutrient absorption, resulting in decreased immunity and production of digestive enzymes.
Collapse
Affiliation(s)
- Marek Pieszka
- Department of Animal Nutrition and Feed Sciences , National Research Institute of Animal Production , Balice , Poland
| | - Kinga Szczepanik
- Department of Animal Nutrition and Feed Sciences , National Research Institute of Animal Production , Balice , Poland
| | - Igor Łoniewski
- Sanprobi sp. z o.o. sp. k ., Kurza Stopka 5/C , Szczecin , Poland
- Department of Biochemical Science , Pomeranian Medical University in Szczecin , Szczecin , Poland
| |
Collapse
|
2
|
Tharabenjasin P, Moonwiriyakit A, Sontikun J, Timpratueang K, Kuno S, Aiebchun T, Jongkon N, Mongkolrob R, Pabalan N, Choowongkomon K, Muanprasat C. The barrier-protective effect of β-eudesmol against type 2-inflammatory cytokine-induced tight junction disassembly in airway epithelial cells. PLoS One 2024; 19:e0302851. [PMID: 38687777 PMCID: PMC11060601 DOI: 10.1371/journal.pone.0302851] [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: 09/17/2023] [Accepted: 04/14/2024] [Indexed: 05/02/2024] Open
Abstract
Allergic inflammation, which is the pathogenesis of allergic rhinitis and asthma, is associated with disruption of the airway epithelial barrier due to the effects of type 2 inflammatory cytokines, i.e. interleukin-4 and interleukin-13 (IL-4/13). The anti-allergic inflammatory effect of β-eudesmol (BE) on the tight junction (TJ) of the airway epithelium has not previously been reported. Herein, the barrier protective effect of BE was determined by measurement of transepithelial electrical resistance and by paracellular permeability assay in an IL-4/13-treated 16HBE14o- monolayer. Pre-treatment of BE concentration- and time- dependently inhibited IL-4/13-induced TJ barrier disruption, with the most significant effect observed at 20 μM. Cytotoxicity analyses showed that BE, either alone or in combination with IL-4/13, had no effect on cell viability. Western blot and immunofluorescence analyses showed that BE inhibited IL-4/13-induced mislocalization of TJ components, including occludin and zonula occludens-1 (ZO-1), without affecting the expression of these two proteins. In addition, the mechanism of the TJ-protective effect of BE was mediated by inhibition of IL-4/13-induced STAT6 phosphorylation, in which BE might serve as an antagonist of cytokine receptors. In silico molecular docking analysis demonstrated that BE potentially interacted with the site I pocket of the type 2 IL-4 receptor, likely at Asn-126 and Tyr-127 amino acid residues. It can therefore be concluded that BE is able to prevent IL-4/13-induced TJ disassembly by interfering with cytokine-receptor interaction, leading to suppression of STAT6-induced mislocalization of occludin and ZO-1. BE is a promising candidate for a therapeutic intervention for inflammatory airway epithelial disorders driven by IL-4/13.
Collapse
Affiliation(s)
- Phuntila Tharabenjasin
- Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Klongluang, Pathumthani, Thailand
| | - Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Kanokphorn Timpratueang
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Suhaibee Kuno
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| | - Thitinan Aiebchun
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Nathjanan Jongkon
- Department of Social and Applied Science, College of Industrial Technology, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| | - Rungrawee Mongkolrob
- Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Klongluang, Pathumthani, Thailand
| | - Noel Pabalan
- Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Klongluang, Pathumthani, Thailand
| | | | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakan, Thailand
| |
Collapse
|
3
|
Moonwiriyakit A, Dinsuwannakol S, Sontikun J, Timpratueang K, Muanprasat C, Khemawoot P. Fine particulate matter PM2.5 and its constituent, hexavalent chromium induce acute cytotoxicity in human airway epithelial cells via inflammasome-mediated pyroptosis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104416. [PMID: 38492761 DOI: 10.1016/j.etap.2024.104416] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
PM2.5-induced airway injury contributes to an increased rate of respiratory morbidity. However, the relationship between PM2.5 toxicants and acute cytotoxic effects remains poorly understood. This study aimed to investigate the mechanisms of PM2.5- and its constituent-induced cytotoxicity in human airway epithelial cells. Exposure to PM2.5 resulted in dose-dependent cytotoxicity within 24 h. Among the PM2.5 constituents examined, Cr(VI) at the dose found in PM2.5 exhibited cytotoxic effects. Both PM2.5 and Cr(VI) cause necrosis while also upregulating the expression of proinflammatory cytokine transcripts. Interestingly, exposure to the conditioned PM, obtained from adsorption in the Cr(VI)-reducing agents, FeSO4 and EDTA, showed a decrease in cytotoxicity. Furthermore, PM2.5 mechanistically enhances programmed pyroptosis through the activation of NLRP3/caspase-1/Gasdermin D pathway and increase of IL-1β. These pyroptosis markers were reduced when exposure to conditioned PM. These findings provide a deeper understanding of mechanisms underlying PM2.5 and Cr(VI) in acute airway toxicity.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand.
| | - Sasiwimol Dinsuwannakol
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Kanokphorn Timpratueang
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| |
Collapse
|
4
|
Moonwiriyakit A, Yimnual C, Noitem R, Dinsuwannakol S, Sontikun J, Kaewin S, Worakajit N, Soontornniyomkij V, Muanprasat C. GPR120/FFAR4 stimulation attenuates airway remodeling and suppresses IL-4- and IL-13-induced airway epithelial injury via inhibition of STAT6 and Akt. Biomed Pharmacother 2023; 168:115774. [PMID: 37924784 DOI: 10.1016/j.biopha.2023.115774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Airway remodeling is associated with severity and treatment insensitivity in asthma. This study aimed to investigate the effects of G protein-coupled receptor 120 (GPR120) stimulation on alleviating allergic inflammation and remodeling of airway epithelium. RESEARCH DESIGN AND METHODS Ovalbumin (OVA)-challenged BALB/c mice and type-2-cytokine (IL-4 and IL-13)-exposed 16HBE human bronchial epithelial cells were treated with GSK137647A, a selective GPR120 agonist. Markers of allergic inflammation and airway remodeling were determined. RESULTS GSK137647A attenuated inflammation and mucus secretion in airway epithelium of OVA-challenged mice. Stimulation of GPR120 in 16HBE suppressed expression of asthma-associated cytokines and cytokine-induced expression of pathogenic mucin-MUC5AC. These effects were abolished by co-treatment with AH7614, a GPR120 antagonist. Moreover, GPR120 stimulation in 16HBE cells reduced expression of fibrotic markers including fibronectin protein and ACTA2 mRNA and inhibited epithelial barrier leakage induced by type-2 inflammation via rescuing expression of zonula occludens-1 protein. Furthermore, GPR120 stimulation prevented the cytokine-induced airway epithelial remodeling via suppression of STAT6 and Akt phosphorylation. CONCLUSIONS Our findings suggest that GPR120 activation alleviates allergic inflammation and remodeling of airway epithelium partly through inhibition of STAT6 and Akt. GPR120 may represent a novel therapeutic target for diseases associated with remodeling of airway epithelium, including asthma.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chantapol Yimnual
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Rattikarn Noitem
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sasiwimol Dinsuwannakol
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Suchada Kaewin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nichakorn Worakajit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Virawudh Soontornniyomkij
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
5
|
He S, Lin F, Hu X, Pan P. Gut Microbiome-Based Therapeutics in Critically Ill Adult Patients-A Narrative Review. Nutrients 2023; 15:4734. [PMID: 38004128 PMCID: PMC10675331 DOI: 10.3390/nu15224734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
The gut microbiota plays a crucial role in the human microenvironment. Dysbiosis of the gut microbiota is a common pathophysiological phenomenon in critically ill patients. Therefore, utilizing intestinal microbiota to prevent complications and improve the prognosis of critically ill patients is a possible therapeutic direction. The gut microbiome-based therapeutics approach focuses on improving intestinal microbiota homeostasis by modulating its diversity, or treating critical illness by altering the metabolites of intestinal microbiota. There is growing evidence that fecal microbiota transplantation (FMT), selective digestive decontamination (SDD), and microbiota-derived therapies are all effective treatments for critical illness. However, different treatments are appropriate for different conditions, and more evidence is needed to support the selection of optimal gut microbiota-related treatments for different diseases. This narrative review summarizes the curative effects and limitations of microbiome-based therapeutics in different critically ill adult patients, aiming to provide possible directions for gut microbiome-based therapeutics for critically ill patients such as ventilator-associated pneumonia, sepsis, acute respiratory distress syndrome, and COVID-19, etc.
Collapse
Affiliation(s)
- Shiyue He
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; (S.H.); (F.L.)
- FuRong Laboratory, Changsha 410078, China
| | - Fengyu Lin
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; (S.H.); (F.L.)
- FuRong Laboratory, Changsha 410078, China
| | - Xinyue Hu
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; (S.H.); (F.L.)
- FuRong Laboratory, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, China
| | - Pinhua Pan
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; (S.H.); (F.L.)
- FuRong Laboratory, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha 410008, China
| |
Collapse
|
6
|
Prado TP, Jara CP, Dias Bóbbo VC, Carraro RS, Sidarta-Oliveira D, de Mendonça GRA, Velloso LA, Araújo EP. A Free Fatty Acid Synthetic Agonist Accelerates Wound Healing and Improves Scar Quality in Mice. Biol Res Nurs 2023; 25:353-366. [PMID: 36444640 DOI: 10.1177/10998004221142331] [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] [Indexed: 04/17/2024]
Abstract
BACKGROUND Impaired wound healing is a health problem around the world, and the search for a novel product to repair wounded skin is a major topic in the field. GW9508 is a synthetic molecule described as a selective agonist of free fatty acid receptors (FFARs) 1 and 4, and there is evidence of its anti-inflammatory effects on several organs of the body. PURPOSE Here, we aimed to evaluate the effects of topical GW9508 on wound healing in mice. RESEARCH DESIGN First, we used bioinformatic methods to determine the expression of FFAR1 and FFAR4 mRNA in the skin from a human cell atlas assembled with single-cell transcriptomes. Next, we employed 6-week-old C57BL6J mice with 2 wounds inflicted in the back. The mice were randomly divided into 2 groups, a control group, which received topical vehicle, and a treatment group, which received GW9508, for 12 days. The wound was monitored by photographic documentation every 2 days, and samples were collected at day 6 and 12 post injury for RT-PCR, western blot and histology analyses. RESULTS FFAR1 and FFAR4 mRNA are expressed in skin cells in similar amounts to those in other tissues. Topical GW9508 accelerated wound healing and decreased gene expression of IL-10 and metalloproteinase 9 on days 6 and 12 post injury. It increased the quantity of Collagen I and improved the organization of collagen fibres. Conclusions: Our results show that GW9508 could be an attractive drug treatment for wounded skin. Future studies need to be performed to assess the impact of GW9508 in chronic wound models.
Collapse
Affiliation(s)
- Thais P Prado
- Nursing School, Laboratory of Cell Signaling Obesity and Comorbidities Center, OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
| | - Carlos P Jara
- Nursing School, Laboratory of Cell Signaling Obesity and Comorbidities Center, OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
| | - Vanessa C Dias Bóbbo
- Nursing School, Laboratory of Cell Signaling Obesity and Comorbidities Center, OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
| | - Rodrigo S Carraro
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Davi Sidarta-Oliveira
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Guilherme R A de Mendonça
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Eliana P Araújo
- Nursing School, Laboratory of Cell Signaling Obesity and Comorbidities Center, OCRC, University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences, Laboratory of Cell Signaling, Obesity and Comorbidities Center - OCRC, University of Campinas, Campinas, Brazil
| |
Collapse
|
7
|
McGill AR, Markoutsa E, Mayilsamy K, Green R, Sivakumar K, Mohapatra S, Mohapatra SS. Acetate-encapsulated Linolenic Acid Liposomes Reduce SARS-CoV-2 and RSV Infection. Viruses 2023; 15:1429. [PMID: 37515117 PMCID: PMC10385125 DOI: 10.3390/v15071429] [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: 04/26/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 07/30/2023] Open
Abstract
Emergent Coronaviridae viruses, such as SARS-CoV-1 in 2003, MERS-CoV in 2012, and SARS-CoV-2 (CoV-2) in 2019, have caused millions of deaths. These viruses have added to the existing respiratory infection burden along with respiratory syncytial virus (RSV) and influenza. There are limited therapies for respiratory viruses, with broad-spectrum treatment remaining an unmet need. Since gut fermentation of fiber produces short-chain fatty acids (SCFA) with antiviral potential, developing a fatty acid-based broad-spectrum antiviral was investigated. Molecular docking of fatty acids showed α-linolenic acid (ALA) is likely to interact with CoV-2-S, NL63-CoV-S, and RSV-F, and an ALA-containing liposome interacted with CoV-2 directly, degrading the particle. Furthermore, a combination of ALA and a SCFA-acetate synergistically inhibited CoV2-N expression and significantly reduced viral plaque formation and IL-6 and IL-1β transcript expression in Calu-3 cells, while increasing the expression of IFN-β. A similar effect was also observed in RSV-infected A549 cells. Moreover, mice infected with a murine-adapted SARS-CoV-2 (MA10) and treated with an ALA-liposome encapsulating acetate showed significant reductions in plaque-forming units present in lung tissue and in infection-associated lung inflammation and cytokines. Taken together, these results demonstrate that the ALA liposome-encapsulating acetate can be a promising broad antiviral therapy against respiratory infections.
Collapse
Affiliation(s)
- Andrew R McGill
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Eleni Markoutsa
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Taneja College of Pharmacy Graduate Programs, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
| | - Karthick Mayilsamy
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Ryan Green
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kavya Sivakumar
- Taneja College of Pharmacy Graduate Programs, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Taneja College of Pharmacy Graduate Programs, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
| |
Collapse
|
8
|
Moonwiriyakit A, Pathomthongtaweechai N, Steinhagen PR, Chantawichitwong P, Satianrapapong W, Pongkorpsakol P. Tight junctions: from molecules to gastrointestinal diseases. Tissue Barriers 2022; 11:2077620. [PMID: 35621376 PMCID: PMC10161963 DOI: 10.1080/21688370.2022.2077620] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Intestinal epithelium functions as a tissue barrier to prevent interaction between the internal compartment and the external milieu. Intestinal barrier function also determines epithelial polarity for the absorption of nutrients and the secretion of waste products. These vital functions require strong integrity of tight junction proteins. In fact, intestinal tight junctions that seal the paracellular space can restrict mucosal-to-serosal transport of hostile luminal contents. Tight junctions can form both an absolute barrier and a paracellular ion channel. Although defective tight junctions potentially lead to compromised intestinal barrier and the development and progression of gastrointestinal (GI) diseases, no FDA-approved therapies that recover the epithelial tight junction barrier are currently available in clinical practice. Here, we discuss the impacts and regulatory mechanisms of tight junction disruption in the gut and related diseases. We also provide an overview of potential therapeutic targets to restore the epithelial tight junction barrier in the GI tract.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Nutthapoom Pathomthongtaweechai
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Peter R Steinhagen
- Department of Hepatology and Gastroenterology, Charité Medical School, Berlin, Germany
| | | | | | - Pawin Pongkorpsakol
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
9
|
Königs V, Pierre S, Schicht M, Welss J, Hahnefeld L, Rimola V, Lütjen-Drecoll E, Geisslinger G, Scholich K. GPR40 Activation Abolishes Diabetes-Induced Painful Neuropathy by Suppressing VEGF-A Expression. Diabetes 2022; 71:774-787. [PMID: 35061031 DOI: 10.2337/db21-0711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022]
Abstract
G-protein-coupled receptor 40 (GPR40) is a promising target to support glucose-induced insulin release in patients with type 2 diabetes. We studied the role of GPR40 in the regulation of blood-nerve barrier integrity and its involvement in diabetes-induced neuropathies. Because GPR40 modulates insulin release, we used the streptozotocin model for type 1 diabetes, in which GPR40 functions can be investigated independently of its effects on insulin release. Diabetic wild-type mice exhibited increased vascular endothelial permeability and showed epineural microlesions in sciatic nerves, which were also observed in naïve GPR40-/- mice. Fittingly, expression of vascular endothelial growth factor-A (VEGF-A), an inducer of vascular permeability, was increased in diabetic wild-type and naïve GPR40-/- mice. GPR40 antagonists increased VEGF-A expression in murine and human endothelial cells as well as permeability of transendothelial barriers. In contrast, GPR40 agonists suppressed VEGF-A release and mRNA expression. The VEGF receptor inhibitor axitinib prevented diabetes-induced hypersensitivities and reduced endothelial and epineural permeability. Importantly, the GPR40 agonist GW9508 reverted established diabetes-induced hypersensitivity, an effect that was blocked by VEGF-A administration. Thus, GPR40 activation suppresses VEGF-A expression, thereby reducing diabetes-induced blood-nerve barrier permeability and reverting diabetes-induced hypersensitivities.
Collapse
Affiliation(s)
- Vanessa Königs
- Fraunhofer Institute for Translational Medicine and Pharmacology, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Frankfurt am Main, Germany
| | - Sandra Pierre
- Institute of Clinical Pharmacology, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jessica Welss
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Frankfurt am Main, Germany
- Institute of Clinical Pharmacology, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Vittoria Rimola
- Institute of Clinical Pharmacology, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Elke Lütjen-Drecoll
- Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Frankfurt am Main, Germany
- Institute of Clinical Pharmacology, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Klaus Scholich
- Fraunhofer Institute for Translational Medicine and Pharmacology, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Frankfurt am Main, Germany
- Institute of Clinical Pharmacology, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Liu R, Cheng F, Zeng K, Li W, Lan J. GPR120 Agonist GW9508 Ameliorated Cellular Senescence Induced by ox-LDL. ACS OMEGA 2020; 5:32195-32202. [PMID: 33376857 PMCID: PMC7758881 DOI: 10.1021/acsomega.0c03581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/15/2020] [Indexed: 05/03/2023]
Abstract
Introduction Oxidized low-density lipoprotein (ox-LDL)-induced endothelial senescence is involved in the pathogenesis of atherosclerosis and many cardiovascular diseases. G-protein-coupled receptor 120 (GPR120), a type of orphan G-protein-coupled receptors (GPRs), plays a vital role in mediating anti-inflammatory and insulin-sensitizing effects. The biological function of GPR120 in vascular endothelial cells is largely unknown. Methods The human aortic endothelial cells (HAECs) were treated with ox-LDL (100 μg/mL) in the presence or absence of GW9508 (50 μM) or AH9614 (1 μM) for 24 h. The LDH assay was used to determine cell death. The dihydroethidium (DHE) staining assay was used to measure intracellular levels of reactive oxidative species (ROS), and a senescence β-galactosidase assay kit was used to determine endothelial senescence. Gene and protein expressions were measured using real-time polymerase chain reaction (PCR) and western blot analysis, respectively. Results Ox-LDL treatment decreased the expression of GPR120 by more than half in HAECs. Typically, 100 μg/mL of ox-LDL- induced 35.2% LDH release, which was reduced to 16.9% by 50 μM GW9508, the agonist of GPR120. Importantly, GW9508 relieved cytotoxicity and suppressed the ox-LDL-induced increase in the activity of senescence-associated β-galactosidase (SA-β-Gal) (from 3.3-fold to 1.6-fold of the control group) and the generation of cellular reactive oxidative species (ROS) (from 3.8-fold to 1.6-fold of the control group). Furthermore, we found that GW9508 ameliorated ox-LDL-induced endothelial cell cycle arrest at the G0/G1 phase and the expression of key senescence proteins, including p53 and plasminogen activator inhibitor-1(PAI-1). Mechanistically, we showed that GW9508 promoted ox-LDL-induced transcriptional factor NF-E2-related factor 2 (NRF2) (increase by 47.3%) translocation into the nucleus. The effect of GW9508 is dependent on its receptor GPR120, the blockage of which by its specific antagonist, AH7614, abolished the antisenescence effect of GW9508. Conclusion Collectively, this study revealed the protective effect of GPR120 activation in vascular endothelial cells, implying that GPR120 is a promising therapeutic target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Ruijie Liu
- Department
of Cardiology, The First Affiliated Hospital
of Ji’nan University, Guangzhou, Guangdong 510630, China
- Department
of Cardiology, Dongguan Songshanhu Central
Hospital, Dongguan, Guangdong 523326, China
| | - Fei Cheng
- Department
of Cardiology, Dongguan Songshanhu Central
Hospital, Dongguan, Guangdong 523326, China
| | - Kanghua Zeng
- Department
of Cardiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi 341000, China
| | - Wenfeng Li
- Department
of Cardiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi 341000, China
| | - Jun Lan
- Department
of Cardiology, Dongguan Songshanhu Central
Hospital, Dongguan, Guangdong 523326, China
- . Tel/Fax: +86-769-81368666
| |
Collapse
|
11
|
Drzazga A, Cichońska E, Koziołkiewicz M, Gendaszewska-Darmach E. Formation of βTC3 and MIN6 Pseudoislets Changes the Expression Pattern of Gpr40, Gpr55, and Gpr119 Receptors and Improves Lysophosphatidylcholines-Potentiated Glucose-Stimulated Insulin Secretion. Cells 2020; 9:E2062. [PMID: 32917053 PMCID: PMC7565006 DOI: 10.3390/cells9092062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
The impaired spatial arrangement and connections between cells creating islets of Langerhans as well as altered expression of G protein-coupled receptors (GPCRs) often lead to dysfunction of insulin-secreting pancreatic β cells and can significantly contribute to the development of diabetes. Differences in glucose-stimulated insulin secretion (GSIS) are noticeable not only in diabetic individuals but also in model pancreatic β cells, e.g., βTC3 and MIN6 β cell lines with impaired and normal insulin secretion, respectively. Now, we compare the ability of GPCR agonists (lysophosphatidylcholines bearing fatty acid chains of different lengths) to potentiate GSIS in βTC3 and MIN6 β cell models, cultured as adherent monolayers and in a form of pseudoislets (PIs) with pancreatic MS1 endothelial cells. Our aim was also to investigate differences in expression of the GPCRs responsive to LPCs in these experimental systems. Aggregation of β cells into islet-like structures greatly enhanced the expression of Gpr40, Gpr55, and Gpr119 receptors. In contrast, the co-culture of βTC3 cells with endothelial cells converted the GPCR expression pattern closer to the pattern observed in MIN6 cells. Additionally, the efficiencies of various LPC species in βTC3-MS1 PIs also shifted toward the MIN6 cell model.
Collapse
Affiliation(s)
- Anna Drzazga
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (E.C.); (M.K.)
| | | | | | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (E.C.); (M.K.)
| |
Collapse
|
12
|
Moonwiriyakit A, Koval M, Muanprasat C. Pharmacological stimulation of G-protein coupled receptor 40 alleviates cytokine-induced epithelial barrier disruption in airway epithelial Calu-3 cells. Int Immunopharmacol 2019; 73:353-361. [PMID: 31129422 DOI: 10.1016/j.intimp.2019.05.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
Abstract
Impairment of airway tight junctions induced by elevated levels of proinflammatory cytokines is implicated in the pathogenesis of inflammatory airway diseases. Pharmacological stimulation of G-protein coupled receptor (GPR) 40, a receptor of polyunsaturated fatty acids, have recently been shown to promote tight junction assembly in airway epithelial cells under non-inflammatory conditions. However, roles of GPR40 in regulating airway epithelial integrity in response to inflammatory insults are unknown. This study was aimed to investigate the effect of GPR40 stimulation on proinflammatory cytokine (TNFα and IL-1β)-induced tight junction disruption in human airway epithelial Calu-3 cells using GW9508, a GPR40 agonist. We found that stimulation of GPR40 by GW9508 attenuated the cytokine-induced airway epithelial barrier leakage as analyzed by measurements of transepithelial electrical resistance and transepithelial flux of fluorescently labeled dextran (molecular weight of 4 kDa). Furthermore, GW9508 prevented the cytokine-induced dislocalization of zonula occludens (ZO)-1, occludin and claudin-1. The barrier-protective effect of GW9508 was abolished by a GPR40 antagonist, but not a GPR120 antagonist. Immunofluorescence staining of NF-ĸB indicated that GW9508 had no effect on cytokine-induced NF-ĸB activation. Intriguingly, GW9508 inhibited cytokine-induced airway epithelial barrier disruption through suppression of extracellular signal-regulated kinase (ERK) phosphorylation in a phospholipase C (PLC) and calcium/calmodulin-dependent protein kinase kinase beta (CaMKKβ)-dependent manner. Collectively, this study uncovered the novel role of GPR40 in preventing cytokine-induced tight junction disruption in airway epithelial cells through mechanisms involving PLC-CaMKKβ-mediated suppression of ERK signaling. Pharmacological stimulation of GPR40 may be beneficial in the treatment of airway diseases.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Chatchai Muanprasat
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand; Excellent Center for Drug Discovery (ECDD), Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand; Research Center of Transport Proteins for Medical Innovation, Faculty of Science, Mahidol University, Rama VI Road, Rajathevi, Bangkok 10400, Thailand.
| |
Collapse
|