1
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. Gut Microbes 2025; 17:2458189. [PMID: 39930324 PMCID: PMC11817531 DOI: 10.1080/19490976.2025.2458189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal growth barrier dysfunction, and aberrant inflammatory responses. Further, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, mostly commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no U.S. Food and Drug Administration (FDA) approved countermeasures that can treat radiation-induced GI injuries. To meet this critical need, Synedgen Inc. has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract, which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy partial body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, Vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory responses mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera while suppressing potentially pathogenic bacteria Enterococcus and Staphylococcus compared with Vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and reducing pro-inflammatory responses. Further development of this drug as an FDA-approved medical countermeasure is of critical importance.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
2
|
Liu MC, Shu YA, Wang YC, Tseng HY, Li MJ, Yu YT, Cheng HC, Tsai PJ, Yang YJ. Faecalibacterium prausnitzii Colonization Attenuates Gut Inflammation and Epithelial Damage in a DSS-Induced Colitis Mice Model. Mediators Inflamm 2025; 2025:7280675. [PMID: 40224484 PMCID: PMC11986197 DOI: 10.1155/mi/7280675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/04/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Reduction of Faecalibacterium prausnitzii abundance is related to inflammatory bowel diseases (IBDs), and supplement of it exists protective effects. Aim: This study aimed to establish a F. prausnitzii-colonized mouse model and investigate that the presence of F. prausnitzii in the gut can ameliorate the severity of dextran sulfate sodium (DSS)-induced colitis. Methods: A F. prausnitzii (ATCC 27768) strain was maintained on the PS-BHI agar plates and manipulated in a strictly anaerobic chamber. A F. prausnitzii-colonized C57BL/6 mice model was tested by a rectal enema with 1 × 109 bacteria/day for 3 days. The 5% DSS was added to drinking water for 3 days to induce colitis and diarrhea in experimental mice. The clinical, cytological, and histological severities were compared between groups. Results: The F. prausnitzii-colonized mice model was successfully established via rectal enema with the property of transfer to offspring. DSS treatment altered gut microbiota and significantly attenuated the abundance of F. prausnitzii in colonized mice. Mice with F. prausnitzii colonization had significantly improved weight loss, anal bleeding, stool consistency, cecum weight, colon length, and serum amyloid A (SAA) level than those without after DSS treatment. Furthermore, the F. prausnitzii-colonized mice significantly reduced the transcription levels of TNF-α, INF-γ and IL-18, and epithelial damage and PMN infiltration in the lamina propria and had better preservation of goblet cells than the control group. Conclusion: We have successfully established a mouse model colonized with F. prausnitzii via rectal enema administration and showed colonization of F. prausnitzii in the gut has a protective effect against DSS-induced colitis.
Collapse
Affiliation(s)
- Meng-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-An Shu
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chin Wang
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Ying Tseng
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Jia Li
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ting Yu
- Department of Pathology, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiu-Chi Cheng
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Jong Yang
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
3
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619652. [PMID: 39484519 PMCID: PMC11526895 DOI: 10.1101/2024.10.22.619652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal epithelial barrier dysfunction, and aberrant inflammatory responses. In addition, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, reduction in the abundance of beneficial commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no FDA-approved countermeasures that can treat radiation-induced GI injury. To meet this critical need, Synedgen Inc., has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy total body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory response mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera, while suppressing potentially pathogenic bacteria compared with vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and effectively reducing proinflammatory responses. Further development of this drug as an FDA-approved medical countermeasure will be of critical importance in the event of a radiation public health emergency.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
4
|
Marvin JC, Liu EJ, Chen HH, Shiovitz DA, Andarawis-Puri N. Proteins Derived From MRL/MpJ Tendon Provisional Extracellular Matrix and Secretome Promote Pro-Regenerative Tenocyte Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602500. [PMID: 39026846 PMCID: PMC11257490 DOI: 10.1101/2024.07.08.602500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Tendinopathies are prevalent musculoskeletal conditions that have no effective therapies to attenuate scar formation. In contrast to other adult mammals, the tendons of Murphy Roths Large (MRL/MpJ) mice possess a superior healing capacity following acute and overuse injuries. Here, we hypothesized that the application of biological cues derived from the local MRL/MpJ tendon environment would direct otherwise scar-mediated tenocytes towards a pro-regenerative MRL/MpJ-like phenotype. We identified soluble factors enriched in the secretome of MRL/MpJ tenocytes using bioreactor systems and quantitative proteomics. We then demonstrated that the combined administration of structural and soluble constituents isolated from decellularized MRL/MpJ tendon provisional ECM (dPECM) and the secretome stimulate scar-mediated rodent tenocytes towards enhanced mechanosensitivity, proliferation, intercellular communication, and ECM deposition associated with MRL/MpJ cell behavior. Our findings highlight key biological mechanisms that drive MRL/MpJ tenocyte activity and their interspecies utility to be harnessed for therapeutic strategies that promote pro-regenerative healing outcomes. Teaser Proteins enriched in a super-healer mouse strain elicit interspecies utility in promoting pro-regenerative tenocyte behavior.
Collapse
|
5
|
Wang CPJ, Ko GR, Lee YY, Park J, Park W, Park TE, Jin Y, Kim SN, Lee JS, Park CG. Polymeric DNase-I nanozymes targeting neutrophil extracellular traps for the treatment of bowel inflammation. NANO CONVERGENCE 2024; 11:6. [PMID: 38332364 PMCID: PMC10853102 DOI: 10.1186/s40580-024-00414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a family of chronic disorders along the gastrointestinal tract. Because of its idiopathic nature, IBD does not have a fundamental cure; current available therapies for IBD are limited to prolonged doses of immunomodulatory agents. While these treatments may reduce inflammation, limited therapeutic efficacy, inconsistency across patients, and adverse side effects from aggressive medications remain as major drawbacks. Recently, excessive production and accumulation of neutrophil extracellular traps (NETs) also known as NETosis have been identified to exacerbate inflammatory responses and induce further tissue damage in IBD. Such discovery invited many researchers to investigate NETs as a potential therapeutic target. DNase-I is a natural agent that can effectively destroy NETs and, therefore, potentially reduce NETs-induced inflammations even without the use of aggressive drugs. However, low stability and rapid clearance of DNase-I remain as major limitations for further therapeutic applications. In this research, polymeric nanozymes were fabricated to increase the delivery and therapeutic efficacy of DNase-I. DNase-I was immobilized on the surface of polymeric nanoparticles to maintain its enzymatic properties while extending its activity in the colon. Delivery of DNase-I using this platform allowed enhanced stability and prolonged activity of DNase-I with minimal toxicity. When administered to animal models of IBD, DNase-I nanozymes successfully alleviated various pathophysiological symptoms of IBD. More importantly, DNase-I nanozyme administration successfully attenuated neutrophil infiltration and NETosis in the colon compared to free DNase-I or mesalamine.
Collapse
Affiliation(s)
- Chi-Pin James Wang
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
| | - Ga Ryang Ko
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
| | - Yun Young Lee
- Department of Biomedical Engineering, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, 96813, USA
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Se-Na Kim
- Research and Development Center, MediArk Inc., Cheongju, Chungbuk, 28644, Republic of Korea.
- Department of Industrial Cosmetic Science, College of Bio-Health University System, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| | - Jung Seung Lee
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
6
|
Sarmikasoglou E, Chu L, Yue F, Faciola AP. Effects of ruminal lipopolysaccharide exposure on primary bovine ruminal epithelial cells. J Dairy Sci 2024; 107:1244-1262. [PMID: 37777002 DOI: 10.3168/jds.2023-23736] [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: 05/11/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023]
Abstract
The objective of this study was to investigate the immunopotential of ruminal lipopolysaccharides (LPS) on cultured primary bovine rumen epithelial cells (REC). Primary bovine REC were isolated from 6 yearling steers and grown in culture for 3 experiments. Experiment 1 aimed to determine the immunopotential of ruminal LPS, experiment 2 aimed to assess tolerance to chronic LPS exposure, and experiment 3 aimed to evaluate antagonistic interactions between ruminal and Escherichia coli LPS. In experiments 1 and 2, REC were exposed to nonpyrogenic water, 20 μg/mL E. coli LPS (EC20), 10 μg/mL ruminal LPS, 20 μg/mL ruminal LPS, and 40 μg/mL ruminal LPS, either continuously or intermittently. For the continuous exposure, REC underwent a 6 h exposure, whereas for the intermittent exposure, the procedure was: (1) a 12 h continuous exposure to treatments followed by LPS removal for 24 h and then another 12 h of exposure (RPT), and (2) a 12 h continuous exposure to treatments followed by LPS removal and a recovery period of 36 h (RCV). In experiment 3, REC were exposed to nonpyrogenic water, 1 μg/mL E. coli LPS, 1 μg/mL ruminal LPS to 1 μg/mL E. coli LPS, 10 μg/mL ruminal LPS to 1 μg/mL E. coli LPS, and 50 μg/mL ruminal LPS to 1 μg/mL E. coli LPS. Each experiment was done as a complete randomized block design with 6 REC donors. The REC-donor was used as blocking factor. Each treatment had 2 technical replicates, and treatment responses for all data were analyzed with the MIXED procedure of SAS. For all experiments, total RNA was extracted from REC and real-time quantitative PCR was performed to determine the relative expression of genes for toll-like receptors (TLR2 and TLR4), proinflammatory cytokines (TNF, IL1B, and IL6), chemokines (CXCL2 and CXCL8), growth factor-like cytokines (CSF2 and TGFB1), and a lipid mediator (PTGS2). In experiment 1, the targeted genes were upregulated by EC20, whereas all ruminal LPS treatments resulted in a lower transcript abundance. Regarding RPT, and RCV condition, in experiment 2, the expression of targeted genes was not affected or was at a lower abundance to EC20 when compared with ruminal LPS treatments. Lastly, in experiment 3, all targeted genes resulted in lower or similar transcript abundance on all ruminal LPS ratios. Overall, our results indicate that ruminal LPS have a limited capacity to activate the TLR4/NF-kB pathway and to induce the expression of inflammatory genes.
Collapse
Affiliation(s)
- E Sarmikasoglou
- Department of Animal Sciences, University of Florida, Gainesville, FL 32608
| | - L Chu
- Department of Animal Sciences, University of Florida, Gainesville, FL 32608
| | - F Yue
- Department of Animal Sciences, University of Florida, Gainesville, FL 32608
| | - A P Faciola
- Department of Animal Sciences, University of Florida, Gainesville, FL 32608.
| |
Collapse
|
7
|
Vezza T, Molina-Tijeras JA, Rodríguez-Nogales A, Garrido-Mesa J, Cádiz-Gurrea MDLL, Segura-Carretero A, González-Tejero MR, Rodríguez-Cabezas ME, Gálvez J, Algieri F. The Antioxidant Properties of Salvia verbenaca Extract Contribute to Its Intestinal Antiinflammatory Effects in Experimental Colitis in Rats. Antioxidants (Basel) 2023; 12:2071. [PMID: 38136191 PMCID: PMC10741154 DOI: 10.3390/antiox12122071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal inflammation with unpredictable symptom fluctuations. While there is no effective cure for IBD, various treatments aim to manage symptoms and improve the quality of life for affected individuals. In recent years, there has been growing interest in the potential benefits of certain natural plants and herbs in the management of IBD. In this regard, this study aimed to evaluate the immunomodulatory and anti-inflammatory effects of a well-characterized extract of Salvia verbenaca (S. verbenaca) in an experimental model of colitis in rats. Interestingly, the daily administration of S. verbenaca (10 and 25 mg/kg) effectively alleviated colitis symptoms, as evidenced by reduced weight/length ratio and colonic damage. Moreover, it reduced oxidative stress markers (MPO and GSH), decreased pro-inflammatory cytokine expression (Il-6, Il-12a, Il-1β, Il-23, Icam-1, Mcp-1, Cinc-1), and preserved the integrity of the intestinal barrier (Villin, Muc-2, Muc-3). These effects suggest S. verbenaca extract could represent a potential complementary candidate to treat gastrointestinal disorders. Its beneficial actions can be related to its antioxidant properties as well as the downregulation of the immune response, which can result in the improvement in the intestine epithelial barrier.
Collapse
Affiliation(s)
- Teresa Vezza
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), 18012 Granada, Spain
- Servicio de Digestivo, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| | - Jose Alberto Molina-Tijeras
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), 18012 Granada, Spain
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), 18012 Granada, Spain
| | - Jose Garrido-Mesa
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
| | - María de la Luz Cádiz-Gurrea
- Department of Analytical Chemistry, Faculty of Science, University of Granada, 18071 Granada, Spain; (M.d.l.L.C.-G.); (A.S.-C.)
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Science, University of Granada, 18071 Granada, Spain; (M.d.l.L.C.-G.); (A.S.-C.)
| | | | - María Elena Rodríguez-Cabezas
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), 18012 Granada, Spain
| | - Julio Gálvez
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), 18012 Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francesca Algieri
- Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (T.V.); (J.A.M.-T.); (J.G.-M.); (J.G.); (F.A.)
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), 18012 Granada, Spain
| |
Collapse
|
8
|
Peña-Cearra A, Castelo J, Lavín JL, Gonzalez-Lopez M, Pascual-Itoiz MA, Fuertes M, Gutiérrez de Juan V, Bárcena L, Martín-Ruiz I, Pellón A, Seoane I, Barriales D, Palacios A, Fullaondo A, Rodríguez-Lago I, Martinez-Chantar ML, Aransay AM, Rodriguez H, Anguita J, Abecia L. Mitochondrial dysfunction-associated microbiota establishes a transmissible refractory response to anti-TNF therapy during ulcerative colitis. Gut Microbes 2023; 15:2266626. [PMID: 37842919 PMCID: PMC10586225 DOI: 10.1080/19490976.2023.2266626] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023] Open
Abstract
Anti-TNF therapy can induce and maintain a remission status during intestinal bowel disease. However, up to 30% of patients do not respond to this therapy by mechanisms that are unknown. Here, we show that the absence of MCJ, a natural inhibitor of the respiratory chain Complex I, induces gut microbiota changes that are critical determinants of the lack of response in a murine model of DSS-induced inflammation. First, we found that MCJ expression is restricted to macrophages in human colonic tissue. Therefore, we demonstrate by transcriptomic analysis of colon macrophages from DSS-induced mice that MCJ-deficiency is linked to the expression of genes belonging to the FcγR signaling pathway and contains an anti-TNF refractory gene signature identified in ulcerative colitis patients. The gut microbial composition changes observed upon DSS treatment in the MCJ-deficient mice revealed the increased presence of specific colitogenic members, including Ruminococcus gnavus and Oscillospira, which could be associated with the non-response to TNF inhibitors. Further, we show that the presence of a microbiota associated resistance to treatment is dominant and transmissible to responsive individuals. Collectively, our findings underscore the critical role played by macrophage mitochondrial function in the gut ecological niche that can substantially affect not only the severity of inflammation but also the ability to successfully respond to current therapies.
Collapse
Affiliation(s)
- Ainize Peña-Cearra
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Janire Castelo
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Jose Luis Lavín
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Applied Mathematics Department, NEIKER-Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | | | - Miguel Fuertes
- Animal Health Department, NEIKER-Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Laura Bárcena
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Itziar Martín-Ruiz
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Aize Pellón
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Iratxe Seoane
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Diego Barriales
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Ainhoa Palacios
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Asier Fullaondo
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | | | - María L. Martinez-Chantar
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- CIBERehd, ISCIII, Madrid, Spain
| | - Ana Mª Aransay
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- CIBERehd, ISCIII, Madrid, Spain
| | - Hector Rodriguez
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Juan Anguita
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Leticia Abecia
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| |
Collapse
|
9
|
Peters DE, Norris LD, Tenora L, Šnajdr I, Ponti AK, Zhu X, Sakamoto S, Veeravalli V, Pradhan M, Alt J, Thomas AG, Majer P, Rais R, McDonald C, Slusher BS. A gut-restricted glutamate carboxypeptidase II inhibitor reduces monocytic inflammation and improves preclinical colitis. Sci Transl Med 2023; 15:eabn7491. [PMID: 37556558 PMCID: PMC10661206 DOI: 10.1126/scitranslmed.abn7491] [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: 12/17/2021] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
There is an urgent need to develop therapeutics for inflammatory bowel disease (IBD) because up to 40% of patients with moderate-to-severe IBD are not adequately controlled with existing drugs. Glutamate carboxypeptidase II (GCPII) has emerged as a promising therapeutic target. This enzyme is minimally expressed in normal ileum and colon, but it is markedly up-regulated in biopsies from patients with IBD and preclinical colitis models. Here, we generated a class of GCPII inhibitors designed to be gut-restricted for oral administration, and we interrogated efficacy and mechanism using in vitro and in vivo models. The lead inhibitor, (S)-IBD3540, was potent (half maximal inhibitory concentration = 4 nanomolar), selective, gut-restricted (AUCcolon/plasma > 50 in mice with colitis), and efficacious in acute and chronic rodent colitis models. In dextran sulfate sodium-induced colitis, oral (S)-IBD3540 inhibited >75% of colon GCPII activity, dose-dependently improved gross and histologic disease, and markedly attenuated monocytic inflammation. In spontaneous colitis in interleukin-10 (IL-10) knockout mice, once-daily oral (S)-IBD3540 initiated after disease onset improved disease, normalized colon histology, and attenuated inflammation as evidenced by reduced fecal lipocalin 2 and colon pro-inflammatory cytokines/chemokines, including tumor necrosis factor-α and IL-17. Using primary human colon epithelial air-liquid interface monolayers to interrogate the mechanism, we further found that (S)-IBD3540 protected against submersion-induced oxidative stress injury by decreasing barrier permeability, normalizing tight junction protein expression, and reducing procaspase-3 activation. Together, this work demonstrated that local inhibition of dysregulated gastrointestinal GCPII using the gut-restricted, orally active, small-molecule (S)-IBD3540 is a promising approach for IBD treatment.
Collapse
Affiliation(s)
- Diane E. Peters
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren D. Norris
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lukáš Tenora
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, 160 00 Prague, Czechia
| | - Ivan Šnajdr
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, 160 00 Prague, Czechia
| | - András K. Ponti
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shinji Sakamoto
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vijayabhaskar Veeravalli
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Manisha Pradhan
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, 160 00 Prague, Czechia
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christine McDonald
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
10
|
Neutrophils recruited to immunization sites initiating vaccine-induced antibody responses by locally expressing BAFF. iScience 2022; 25:104453. [PMID: 35874922 PMCID: PMC9301880 DOI: 10.1016/j.isci.2022.104453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/23/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
Abstract
Neutrophils played a key role in the innate immune responses. Less is known about whether and how the neutrophils recruited in the immunization sites affecting the vaccine-induced antibody responses. In the process of evaluating the efficacy of an oil-in-water emulsion-formulated vaccine in mice, we found that neutrophils were rapidly and massively recruited to immunization sites but were barely detected in the draining lymph nodes. Interestingly, B cell-activating factor (BAFF) was abundantly expressed in the recruiting neutrophils at a very early stage. The initial neutrophil-derived BAFF firstly brought about the B cell responses in the local part, then subsequently in lymphoid organs. Activated B cells produced more BAFF through TLR9-IRF5 signaling pathway, thereby amplifying the vaccine-induced antibody responses. Suppressing BAFF in the neutrophils could weaken the B cell activation and reduce the antibody production. The data indicate that vaccines endow neutrophils with the potential to orchestrate antibody responses at immunization sites. Neutrophils at immunization sites influencing subsequent immune responses Neutrophil-driven BAFF at immunization sites assisting B cell responses to vaccines Activated B cells produce more BAFF through TLR9-IRF5 signaling pathway BAFF-producing neutrophils orchestrate antibody responses at immunization sites
Collapse
|
11
|
Ruiz Castro PA, Yepiskoposyan H, Gubian S, Calvino-Martin F, Kogel U, Renggli K, Peitsch MC, Hoeng J, Talikka M. Systems biology approach highlights mechanistic differences between Crohn's disease and ulcerative colitis. Sci Rep 2021; 11:11519. [PMID: 34075172 PMCID: PMC8169754 DOI: 10.1038/s41598-021-91124-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
The molecular mechanisms of IBD have been the subject of intensive exploration. We, therefore, assembled the available information into a suite of causal biological network models, which offer comprehensive visualization of the processes underlying IBD. Scientific text was curated by using Biological Expression Language (BEL) and compiled with OpenBEL 3.0.0. Network properties were analysed by Cytoscape. Network perturbation amplitudes were computed to score the network models with transcriptomic data from public data repositories. The IBD network model suite consists of three independent models that represent signalling pathways that contribute to IBD. In the “intestinal permeability” model, programmed cell death factors were downregulated in CD and upregulated in UC. In the “inflammation” model, PPARG, IL6, and IFN-associated pathways were prominent regulatory factors in both diseases. In the “wound healing” model, factors promoting wound healing were upregulated in CD and downregulated in UC. Scoring of publicly available transcriptomic datasets onto these network models demonstrated that the IBD models capture the perturbation in each dataset accurately. The IBD network model suite can provide better mechanistic insights of the transcriptional changes in IBD and constitutes a valuable tool in personalized medicine to further understand individual drug responses in IBD.
Collapse
Affiliation(s)
- Pedro A Ruiz Castro
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland.
| | - Hasmik Yepiskoposyan
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland.
| | - Sylvain Gubian
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Florian Calvino-Martin
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Ulrike Kogel
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Kasper Renggli
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland.
| |
Collapse
|
12
|
Flis E, Barber G, Nulty C, Keogh B, McGuirk P, Anand A, O’Sullivan J, Quante M, Creagh EM. Identification of TLR2 Signalling Mechanisms Which Contribute to Barrett's and Oesophageal Adenocarcinoma Disease Progression. Cancers (Basel) 2021; 13:cancers13092065. [PMID: 33922955 PMCID: PMC8123271 DOI: 10.3390/cancers13092065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Oesophageal adenocarcinoma (EAC) is a common type of oesophageal cancer with a rapidly rising incidence. Risk factors such as reflux, smoking, obesity and Barrett’s oesophagus cause chronic irritation and inflammation in the oesophagus. A receptor that causes inflammation, called Toll-like receptor 2 (TLR2), is expressed at higher levels in oesophageal cells from patients with Barrett’s and EAC, compared to disease-free patients. This study aimed to identify mechanisms involved in TLR2-mediated inflammation in oesophageal cells; and to assess whether TLR2 represents a therapeutic target to limit EAC development. Findings reveal that TLR2 activation in Barrett’s organoids and oesophageal cancer cells amplifies inflammation and promotes cancer development by causing the secretion of several inflammatory factors, most notably the nuclear protein, HMGB1. We demonstrate that TLR2 neutralisation efficiently blocks the inflammatory effects of TLR2 in these systems, revealing the therapeutic potential of TLR2 targeting to limit oesophageal disease and cancer progression. Abstract Chronic inflammation plays an important role in the pathogenesis of oesophageal adenocarcinoma (EAC) and its only known precursor, Barrett’s oesophagus (BE). Recent studies have shown that oesophageal TLR2 levels increase from normal epithelium towards EAC. TLR2 signalling is therefore likely to be important during EAC development and progression, which requires an inflammatory microenvironment. Here, we show that, in response to TLR2 stimulation, BE organoids and early-stage EAC cells secrete pro-inflammatory cytokines and chemokines which recruit macrophages to the tumour site. Factors secreted from TLR2-stimulated EAC cells are shown to subsequently activate TLR2 on naïve macrophages, priming them for inflammasome activation and inducing their differentiation to an M2/TAM-like phenotype. We identify the endogenous TLR2 ligand, HMGB1, as the factor secreted from EAC cells responsible for the observed TLR2-mediated effects on macrophages. Our results indicate that HMGB1 signalling between EAC cells and macrophages creates an inflammatory tumour microenvironment to facilitate EAC progression. In addition to identifying HMGB1 as a potential target for early-stage EAC treatment, our data suggest that blocking TLR2 signalling represents a mechanism to limit HMGB1 release, inflammatory cell infiltration and inflammation during EAC progression.
Collapse
Affiliation(s)
- Ewelina Flis
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland; (E.F.); (G.B.); (C.N.); (B.K.); (P.M.)
| | - Gillian Barber
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland; (E.F.); (G.B.); (C.N.); (B.K.); (P.M.)
| | - Ciara Nulty
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland; (E.F.); (G.B.); (C.N.); (B.K.); (P.M.)
| | - Brian Keogh
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland; (E.F.); (G.B.); (C.N.); (B.K.); (P.M.)
| | - Peter McGuirk
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland; (E.F.); (G.B.); (C.N.); (B.K.); (P.M.)
| | - Akanksha Anand
- Department of Internal Medicine, Technical University of Munich, D-80333 Munich, Germany; (A.A.); (M.Q.)
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James’s Hospital, D08 W9RT Dublin, Ireland;
| | - Michael Quante
- Department of Internal Medicine, Technical University of Munich, D-80333 Munich, Germany; (A.A.); (M.Q.)
| | - Emma M. Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, D02 R590 Dublin, Ireland; (E.F.); (G.B.); (C.N.); (B.K.); (P.M.)
- Correspondence: ; Tel.: +353-1-8962539
| |
Collapse
|
13
|
Lambertini C, Zannoni A, Romagnoli N, Bombardi C, Morini M, Dondi F, Bernardini C, Forni M, Rinnovati R, Spadari A. Expression of Proteinase-Activated Receptor 2 During Colon Volvulus in the Horse. Front Vet Sci 2020; 7:589367. [PMID: 33330716 PMCID: PMC7728609 DOI: 10.3389/fvets.2020.589367] [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: 07/30/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022] Open
Abstract
Large colon volvulus in horses is associated with a poor prognosis, especially when ischemic-reperfusion injury of the affected intestinal tract develops. Proteinase-activated receptor 2 (PAR2) plays an important role in the pathogenesis of inflammation in the gastrointestinal tract. The aim of this study was to evaluate the distribution and expression of PAR2 in colonic pelvic flexure of horses spontaneously affected by large colon volvulus (CVH group). Eight horses admitted for severe abdominal colon volvolus and which underwent surgery were included. Colon samples were collected after enterotomy. Data previously obtained from healthy horses were used as a control group. Histologic evaluation was carried out to grade the severity of the colon lesions. Immunofluorescence, western blot and quantitative polymerase chain reaction (RT-qPCR) were carried out on colon samples to evaluate PAR2 expression. In addition, the transcriptional profile of cytokines and chemokines was evaluated using RT2 Profiler™ PCR Array Horse Cytokines & Chemokines. Three out of the eight patients were euthanised due to clinical deterioration. Immunostaining for PAR2 was observed in the enterocytes, intestinal glands and neurons of the submucosal and myenteric plexi. In the CVH horses, the expression of PAR2 mesenger RNA (mRNA) did not differ significantly from that of the healthy animals; western blots of the mucosa of the colon tracts showed a clear band of the expected molecular weight for PAR2 (~44 kDa) and a band smaller than the expected molecular weight for PAR2 (25kDa), suggesting its activation. The gene expressions for C-X-C motif ligand 1 (CXCL1); interleukin 8 (IL8), macrophage inflammatory protein 2 beta (MIP-2BETA) were upregulated in the colic horses as compared with the colons of the healthy horses. Therefore, in the present study, the expression and activation of PAR2 in the colons of horses in the presence of an inflammatory reaction like that occurring in those with spontaneous colon volvulus was confirmed.
Collapse
Affiliation(s)
- Carlotta Lambertini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Noemi Romagnoli
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Cristiano Bombardi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Maria Morini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Francesco Dondi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Riccardo Rinnovati
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Spadari
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
14
|
Baumann P, Wiegert S, Greco F, Ersch J, Cannizzaro V. Strain-specific differences in lung tissue viscoelasticity of mechanically ventilated infant Sprague-Dawley and Wistar rats. Am J Physiol Lung Cell Mol Physiol 2020; 320:L220-L231. [PMID: 33207919 DOI: 10.1152/ajplung.00100.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Rats are often used in ventilator-induced lung injury (VILI) models. However, strain-specific susceptibility for VILI has not been elucidated yet. The aim of this study was to demonstrate strain-specific differences in VILI in infant Sprague-Dawley and Wistar rats. VILI was compared in 2-wk-old pups after 8 h of protective or injurious ventilation. Pups were ventilated with tidal volumes (VT) of ∼7 mL/kg and positive end-expiratory pressures (PEEP) of 6 cmH2O (VT7 PEEP6) or with VT of ∼21 mL/kg and PEEP 2 cmH2O (VT21 PEEP2). Interleukin-6, macrophage inflammatory protein-2 (MIP-2), inflammatory cells, and albumin in bronchoalveolar lavage fluid (BALF); histology; and low-frequency forced oscillation technique (LFOT) and pressure-volume (PV) maneuvers were assessed. Alveolar macrophages, neutrophils, and MIP-2 derived from BALF revealed more pronounced VILI after VT21 PEEP2 in both strains. LFOT and PV analyses demonstrated rat strain-specific differences both at baseline and particularly in response to VT21 PEEP2 ventilation. Sprague-Dawley rats showed higher airway and tissue resistance and elastance values with no difference in hysteresivity between ventilation strategies. Wister rats challenged by VT21 PEEP2 experienced significantly more energy dissipation when compared with VT7 PEEP6 ventilation. In conclusion, both rat strains are useful for VILI models. The degree of VILI severity depends on ventilation strategy and selected strain. However, fundamental and time-dependent differences in respiratory system mechanics exist and reflect different lung tissue viscoelasticity. Hence, strain-specific characteristics of the respiratory system need to be considered when planning and interpreting VILI studies with infant rats.
Collapse
Affiliation(s)
- Philipp Baumann
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Susanne Wiegert
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Francesco Greco
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Joerg Ersch
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Vincenzo Cannizzaro
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Department of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Kent-Dennis C, Aschenbach JR, Griebel PJ, Penner GB. Effects of lipopolysaccharide exposure in primary bovine ruminal epithelial cells. J Dairy Sci 2020; 103:9587-9603. [PMID: 32747102 DOI: 10.3168/jds.2020-18652] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022]
Abstract
The objective of this study was to investigate whether cultured ruminal epithelial cells (REC) responded to lipopolysaccharide (LPS) stimulation and determine whether LPS induced a proinflammatory response. Primary bovine REC were isolated and grown in culture for 2 studies. In study 1, REC were isolated from Holstein bull calves (n = 8) and grown in culture for 10 to 12 d. Cells were then exposed to 0, 10,000, 50,000, or 200,000 endotoxin (E)U/mL of LPS (Escherichia coli O55:B5) for either 6 or 24 h. The effect of LPS exposure on cell viability was analyzed by flow cytometry using a propidium iodide stain. In study 2, cells were isolated from Holstein bull calves (n = 5) and yearling beef heifers (n = 4). Cells were exposed to either 1,000 or 50,000 EU/mL of LPS using the following conditions: (1) medium alone time-matched controls, (2) 12-h LPS exposure, (3) 24 h of LPS exposure, (4) 36 h of LPS exposure, (5) 12 h of LPS exposure followed by LPS removal for 24 h before restimulating with LPS for an additional 12 h (RPT), and (6) 12 h of LPS exposure followed by LPS removal for 36 (RVY). For both experiments, total RNA was extracted from REC and real-time quantitative PCR was performed to determine relative expression of genes for toll-like receptors (TLR2 and TLR4), proinflammatory cytokines (TNF and IL1B), chemokines (CXCL2 and CXCL8), a lipid mediator (PTGS2), and growth factor-like cytokines (CSF2 and IL7). In study 1, LPS exposure did not negatively affect cell viability. Treatment of cells with LPS resulted in increased transcript abundance for all genes analyzed. The TLR2, IL7, and TLR4 had a greater magnitude of change at 6 h compared with 24 h. Quadratic expression patterns were detected for TNF, IL1B, CXCL2, CXCL8, and CSF2. These results suggested that REC increase expression of proinflammatory genes following exposure to LPS. In study 2, all genes analyzed were upregulated in a quadratic manner following exposure to LPS for different time intervals. The TLR4, TNF, CXCL2, CXCL8, CSF2, and IL7 gene expression was significantly greater after a single 12 h of LPS exposure than after RPT exposure, suggesting repeated exposure of REC to LPS may induce a tolerogenic effect. When LPS was removed from the medium (RVY), transcript abundance for all genes analyzed decreased and expression of TLR2, TLR4, and IL7 returned to baseline levels, suggesting REC recovered following exposure to LPS. Overall, the data suggest cultured REC respond to LPS stimulation by increasing transcription of proinflammatory genes and this transcriptional response was influenced by the dose, duration, and frequency of LPS exposure.
Collapse
Affiliation(s)
- C Kent-Dennis
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada, S7N 5A8
| | - J R Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, D-14163, Berlin, Germany
| | - P J Griebel
- Vaccine and Infectious Disease Organization/Intervac, University of Saskatchewan, Saskatoon, SK, Canada, S7N 5E3; School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada, S7N 2Z4
| | - G B Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada, S7N 5A8.
| |
Collapse
|
16
|
Nerolidol Mitigates Colonic Inflammation: An Experimental Study Using both In Vivo and In Vitro Models. Nutrients 2020; 12:nu12072032. [PMID: 32650602 PMCID: PMC7400891 DOI: 10.3390/nu12072032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Nerolidol (NED) is a naturally occurring sesquiterpene alcohol present in various plants with potent anti-inflammatory effects. In the current study, we investigated NED as a putative anti-inflammatory compound in an experimental model of colonic inflammation. C57BL/6J male black mice (C57BL/6J) were administered 3% dextran sodium sulfate (DSS) in drinking water for 7 days to induce colitis. Six groups received either vehicle alone or DSS alone or DSS with oral NED (50, 100, and 150 mg/kg body weight/day by oral gavage) or DSS with sulfasalazine. Disease activity index (DAI), colonic histology, and biochemical parameters were measured. TNF-α-treated HT-29 cells were used as in vitro model of colonic inflammation to study NED (25 µM and 50 µM). NED significantly decreased the DAI and reduced the inflammation-associated changes in colon length as well as macroscopic and microscopic architecture of the colon. Changes in tissue Myeloperoxidase (MPO) concentrations, neutrophil and macrophage mRNA expression (CXCL2 and CCL2), and proinflammatory cytokine content (IL-1β, IL-6, and TNF-α) both at the protein and mRNA level were significantly reduced by NED. The increase in content of the proinflammatory enzymes, COX-2 and iNOS induced by DSS were also significantly inhibited by NED along with tissue nitrate levels. NED promoted Nrf2 nuclear translocation dose dependently. NED significantly increased antioxidant enzymes activity (Superoxide dismutase (SOD) and Catalase (CAT)), Hemeoxygenase-1 (HO-1), and SOD3 mRNA levels. NED treatment in TNF-α-challenged HT-29 cells significantly decreased proinflammatory chemokines (CXCL1, IL-8, CCL2) and COX-2 mRNA levels. NED supplementation attenuates colon inflammation through its potent antioxidant and anti-inflammatory activity both in in vivo and in vitro models of colonic inflammation.
Collapse
|
17
|
Semaniakou A, Brothers S, Gould G, Zahiremani M, Paton J, Chappe F, Li A, Anini Y, Croll RP, Chappe V. Disrupted local innervation results in less VIP expression in CF mice tissues. J Cyst Fibros 2020; 20:154-164. [PMID: 32600901 DOI: 10.1016/j.jcf.2020.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 11/16/2022]
Abstract
Vasoactive Intestinal Peptide (VIP) is the major physiological agonist of the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) chloride channel activity. VIP functions as a neuromodulator and neurotransmitter secreted by neurons innervating all exocrine glands. VIP is also a potent vasodilator and bronchodilator that regulates exocrine gland secretions, contributing to local innate defense by stimulating the movement of water and chloride transport across intestinal and tracheobronchial epithelia. Previous human studies have shown that the rich intrinsic neuronal networks for VIP secretion around exocrine glands could be lost in tissues from patients with cystic fibrosis. Our research has since confirmed, in vitro and in vivo, the need for chronic VIP exposure to maintain functional CFTR chloride channels at the cell surface of airways and intestinal epithelium, as well as normal exocrine tissues morphology [1]. The goal of the present study was to examine changes in VIP in the lung, duodenum and sweat glands of 8- and 17-weeks old F508del/F508del mice and to investigate VIPergic innervation in the small intestine of CF mice, before important signs of the disease development. Our data show that a low amount of VIP is found in CF tissues prior to tissue damage. Moreover, we found a specific reduction in VIPergic and cholinergic innervation of the small intestine. The general innervation of the primary and secondary myenteric plexus was lost in CF tissues, with the presence of enlarged ganglionic cells in the tertiary layer. We propose that low amount of VIP in CF tissues is due to a reduction in VIPergic and cholinergic innervation and represents an early defect that constitutes an aggravating factor for CF disease progression.
Collapse
Affiliation(s)
- Anna Semaniakou
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Sarah Brothers
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Grayson Gould
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Mehrsa Zahiremani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jamie Paton
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Frederic Chappe
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Audrey Li
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Younes Anini
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada; Department of Obstetrics and Gynecology, IWK Health Center, Halifax, NS, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Valerie Chappe
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
18
|
Kumar R, Derbigny WA. TLR3 Deficiency Leads to a Dysregulation in the Global Gene-Expression Profile in Murine Oviduct Epithelial Cells Infected with Chlamydia muridarum. ACTA ACUST UNITED AC 2020; 1:1-13. [PMID: 31891165 PMCID: PMC6937138 DOI: 10.18689/ijmr-1000101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chlamydia trachomatis replicates primarily in the epithelial cells lining the genital tract and induces the innate immune response by triggering cellular pathogen recognition receptors (PRRs). Our previous studies showed that Toll-like receptor 3 (TLR3) is expressed in murine oviduct epithelial (OE) cells, is the primary PRR triggered by C. muridarum (Cm) early during infection to induce IFN-β synthesis, and that TLR3 signaling regulates the chlamydial induced synthesis of a plethora of other innate inflammatory modulators including IL-6, CXCL10, CXCL16 and CCL5. We also showed that the expression of these cytokines induced by Chlamydia was severely diminished during TLR3 deficiency; however, the replication of Chlamydiain TLR3 deficient OE cells was more robust than in WT cells. These data suggested that TLR3 had a biological impact on the inflammatory response to Chlamydia infection; however, the global effects of TLR3 signaling in the cellular response to Chlamydia infection in murine OE cells has not yet been investigated. To determine the impact of TLR3 signaling on Chlamydia infection in OE cell at the transcriptome level, we infected wild-type (OE-WT) and TLR3-deficient (OE-TLR3KO) cells with Cm, and performed transcriptome analyses using microarray. Genome-wide expression and ingenuity pathway analysis (IPA) identified enhanced expression of host genes encoding for components found in multiple cellular processes encompassing: (1) pro-inflammatory, (2) cell adhesion, (3) chemoattraction, (4) cellular matrix and small molecule transport, (5) apoptosis, and (6) antigen-processing and presentation. These results support a role for TLR3 in modulating the host cellular responses to Cm infection that extend beyond inflammation and fibrosis, and shows that TLR3 could serve a potential therapeutic target for drug and/or vaccine development.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana-46202, USA
| | - Wilbert A Derbigny
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana-46202, USA
| |
Collapse
|
19
|
Long SR, Lanter BB, Pazos MA, Mou H, Barrios J, Su CW, Wang ZQ, Walker WA, Hurley BP, Shi HN. Intestinal helminth infection enhances bacteria-induced recruitment of neutrophils to the airspace. Sci Rep 2019; 9:15703. [PMID: 31673002 PMCID: PMC6823376 DOI: 10.1038/s41598-019-51991-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 10/08/2019] [Indexed: 12/15/2022] Open
Abstract
Intestinal helminth infections elicit Th2-type immunity, which influences host immune responses to additional threats, such as allergens, metabolic disease, and other pathogens. Th2 immunity involves a shift of the CD4+ T-cell population from type-0 to type-2 (Th2) with increased abundance of interleukin (IL)-4 and IL-13. This study sought to investigate if existing gut-restricted intestinal helminth infections impact bacterial-induced acute airway neutrophil recruitment. C57BL/6 mice were divided into four groups: uninfected; helminth-Heligmosomoides polygyrus infected; Pseudomonas aeruginosa infected; and coinfected. Mice infected with H. polygyrus were incubated for 2 weeks, followed by P. aeruginosa intranasal inoculation. Bronchial alveolar lavage, blood, and lung samples were analyzed. Interestingly, infection with gut-restricted helminths resulted in immunological and structural changes in the lung. These changes include increased lung CD4+ T cells, increased Th2 cytokine expression, and airway goblet cell hyperplasia. Furthermore, coinfected mice exhibited significantly more airspace neutrophil infiltration at 6 hours following P. aeruginosa infection and exhibited an improved rate of survival compared with bacterial infected alone. These results suggest that chronic helminth infection of the intestines can influence and enhance acute airway neutrophil responses to P. aeruginosa infection.
Collapse
Affiliation(s)
- Shao Rong Long
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan, China
| | - Bernard B Lanter
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Michael A Pazos
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Juliana Barrios
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Chien-Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan, China
| | - W Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Bryan P Hurley
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | - Hai Ning Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
20
|
Chang YL, Lo HY, Cheng SP, Chang KT, Lin XF, Lee SP, Ma J, Kuo ML, Hsieh MF, Chan CK. Therapeutic Efficacy of Subcutaneous and Intraperitoneal Injections of a Single Dose of Human Umbilical Mesenchymal Stem Cells in Acute and Chronic Colitis in a Mouse Model. J Med Biol Eng 2019. [DOI: 10.1007/s40846-019-00494-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
21
|
Gorth DJ, Shapiro IM, Risbud MV. A New Understanding of the Role of IL-1 in Age-Related Intervertebral Disc Degeneration in a Murine Model. J Bone Miner Res 2019; 34:1531-1542. [PMID: 30875127 PMCID: PMC6697204 DOI: 10.1002/jbmr.3714] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/05/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022]
Abstract
Increased cytokine expression, in particular interleukin-1β (IL-1β), is considered a hallmark of intervertebral disc degeneration. However, the causative relationship between IL-1 and age-dependent degeneration has not been established. To investigate the role of IL-1 in driving age-related disc degeneration, we studied the spine phenotype of global IL-1α/β double knockout (IL-1KO) mice at 12 and 20 months. Multiplex ELISA analysis of blood revealed significant reductions in the concentrations of IFN-γ, IL-5, IL-15, TNF-α, IP-10, and a trend of reduced concentrations of IL-10, macrophage inflammatory protein 1α (MIP-1α), keratinocyte chemoattractant/human growth-regulated oncogene (KC/GRO), and IL-6. However, the circulating level of MIP-2, a neutrophil chemoattractant, was increased in the IL-1KO. The alterations in systemic cytokine levels coincided with altered bone morphology-IL-1KO mice exhibited significantly thicker caudal cortical bone at 12 and 20 months. Despite these systemic inflammatory and bony changes, IL-1 deletion only minimally affected disc health. Both wild-type (WT) and IL-1KO mice showed age-dependent disc degeneration. Unexpectedly, rather than protecting the animals from degeneration, the aging phenotype was more pronounced in IL-1KO animals: knockout mice evidenced significantly more degenerative changes in the annulus fibrosis (AF) together with alterations in collagen type and maturity. At 20 months, there were no changes in nucleus pulposus (NP) extracellular matrix composition or cellular marker expression; however, the IL-1KO NP cells occupied a smaller proportion of the NP compartment that those of WT controls. Taken together, these results show that IL-1 deletion altered the systemic inflammatory environment and vertebral bone morphology. However, instead of protecting discs from age-related disc degeneration, global IL-1 deletion amplified the degenerative phenotype. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Deborah J Gorth
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
22
|
Ko CL, Lin JA, Chen KY, Hsu AC, Wu SY, Tai YT, Lin KH, Chung WC, Li MH. Netrin-1 Dampens Hypobaric Hypoxia-Induced Lung Injury in Mice. High Alt Med Biol 2019; 20:293-302. [PMID: 31329475 DOI: 10.1089/ham.2018.0116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: This study aimed to explore the effects of netrin-1 on hypobaric hypoxia-induced lung injury in mice. Methods: We exposed 6-8-week-old C57BL/6 mice to hypobaric stress at 340 mmHg for 30 minutes followed by 260 mmHg for different periods (6, 12, 18, and 24 hours) to observe the severity of lung injury (O2 concentration, 21%; 54.6 mmHg). The wet/dry weight ratio and protein leakage from the mouse lung were used to determine the suitable exposure time. Netrin-1 was injected into the tail vein of mice before 18-hour decompression. Inflammatory cytokines, lung injury scores, and activity of nuclear factor κB were evaluated. The expression of apoptosis-related proteins was also examined. Results: Protein concentration in the bronchoalveolar lavage fluid was significantly higher in the 18-hour group (p < 0.05). Pulmonary pathology revealed neutrophil infiltration, alveolar septum thickening, and tissue edema. Injury score and macrophage inflammatory protein 2 levels were also increased. Intrinsic apoptosis pathway was activated. Hypoxia decreased the expression of Bcl2 protein, the number of active caspase-3-stained cells, and UNC5HB receptors. Pretreatment with netrin-1 reduced protein leakage, inhibited neutrophil migration, lowered the injury score, attenuated apoptosis, and increased UNC5HB receptor expression. Conclusion: Netrin-1 dampens hypobaric hypoxia-induced lung injury by inhibiting neutrophil migration and attenuating apoptosis.
Collapse
Affiliation(s)
- Ching-Lung Ko
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jui-An Lin
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kung-Yen Chen
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - An-Chih Hsu
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ting Tai
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ko-Huan Lin
- Division of Psychiatry, Hualien Armed Forces General Hospital, Hualien, Taiwan
| | - Wei-Chen Chung
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Min-Hui Li
- Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| |
Collapse
|
23
|
Nunes NS, Chandran P, Sundby M, Visioli F, da Costa Gonçalves F, Burks SR, Paz AH, Frank JA. Therapeutic ultrasound attenuates DSS-induced colitis through the cholinergic anti-inflammatory pathway. EBioMedicine 2019; 45:495-510. [PMID: 31253515 PMCID: PMC6642284 DOI: 10.1016/j.ebiom.2019.06.033] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/05/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ulcerative Colitis (UC) is an Inflammatory Bowel Disease (IBD) characterized by uncontrolled immune response, diarrhoea, weight loss and bloody stools, where sustained remission is not currently achievable. Dextran Sulphate Sodium (DSS)-induced colitis is an animal model that closely mimics human UC. Ultrasound (US) has been shown to prevent experimental acute kidney injury through vagus nerve (VN) stimulation and activation of the cholinergic anti-inflammatory pathway (CAIP). Since IBD patients may present dysfunctional VN activity, our aim was to determine the effects of therapeutic ultrasound (TUS) in DSS-induced colitis. METHODS Acute colitis was induced by 2% DSS in drinking water for 7 days and TUS was administered to the abdominal area for 7 min/day from days 4-10. Clinical symptoms were analysed, and biological samples were collected for proteomics, macroscopic and microscopic analysis, flow cytometry and immunohistochemistry. FINDINGS TUS attenuated colitis by reducing clinical scores, colon shortening and histological damage, inducing proteomic tolerogenic response in the gut during the injury phase and early recovery of experimental colitis. TUS did not improve clinical and pathological outcomes in splenectomised mice, while α7nAChR (α7 nicotinic acetylcholine receptor - indicator of CAIP involvement) knockout animals presented with disease worsening. Increased levels of colonic F4/80+α7nAChR+ macrophages in wild type mice suggest CAIP activation. INTERPRETATION These results indicate TUS improved DSS-induced colitis through stimulation of the splenic nerve along with possible contribution by VN with CAIP activation. FUND: Intramural Research Programs of the Clinical Centre, the National Institute of Biomedical Imaging and Bioengineering at the NIH and CAPES/Brazil.
Collapse
Affiliation(s)
- Natalia Schneider Nunes
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States; Gastroenterology and Hepatology Sciences Graduate Program, UFRGS, Porto Alegre, RS, Brazil.
| | - Parwathy Chandran
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States
| | - Fernanda Visioli
- Faculty of Dentistry, Oral Pathology, UFRGS, Porto Alegre, RS, Brazil
| | | | - Scott Robert Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States
| | - Ana Helena Paz
- Gastroenterology and Hepatology Sciences Graduate Program, UFRGS, Porto Alegre, RS, Brazil
| | - Joseph Alan Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States; National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, MD, United States
| |
Collapse
|
24
|
Boshagh MA, Foroutan P, Moloudi MR, Fakhari S, Malakouti P, Nikkhoo B, Jalili A. ELR positive CXCL chemokines are highly expressed in an animal model of ulcerative colitis. J Inflamm Res 2019; 12:167-174. [PMID: 31417300 PMCID: PMC6599894 DOI: 10.2147/jir.s203714] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Background: The presence of neutrophil-rich inflammation in colon tissues of patients with ulcerative colitis (UC) is one of the most important histological characteristics of this disease. However, the expression of CXCL chemokines governing the infiltration of neutrophils in UC has not been well elucidated. Materials and methods: In this experimental study, the UC model was induced in Wistar rats by administration of 2 mL 4% acetic acid into the large colon through the rectum. Animals were anesthetized after 48 hrs; their colon tissue samples were isolated for macroscopic and histopathological examinations. The expression of CXCL family was assessed by reverse transcription polymerase chain reaction (qRT-PCR) technique. Results: Heavy infiltration of neutrophils, coagulation necrosis, and ulcers were observed in H&E staining, which pathologically proved the UC model. qRT-PCR results showed that ELR+ CXC chemokines such as CXCL6 and CXCL3 had the highest expression in the UC group, which was 49 and 28 times higher than that of the control group, respectively. In addition, other chemokines of this group including CXCL1, CXCL2, and CXCL7 had a significant increase compared to the control group (P≤0.05). However, ELR− CXC chemokines such as CXCL4, CXCL13, and CXCL16 showed a smaller upregulation, while CXCL14 chemokine showed a significant decrease compared to the control group (P≤0.05). However, the expression of CXCL9-12 and CXCL17 did not change. Conclusion: The results showed that the ELR+ CXC chemokines, especially CXCL6 and CXCL3, many involved in the pathogenesis of UC; therefore, CXCL6 and CXCL3 chemokines can be used as therapeutic targets for UC, although more studies using human samples are required.
Collapse
Affiliation(s)
- Mohammad Amin Boshagh
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Immunology & Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Poorya Foroutan
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Immunology & Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Raman Moloudi
- Liver and Digestive Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Shohreh Fakhari
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Parisa Malakouti
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkhoo
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ali Jalili
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Immunology & Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
25
|
Robinson SC, Chaudhary R, Jiménez-Saiz R, Rayner LGA, Bayer L, Jordana M, Daniel JM. Kaiso-induced intestinal inflammation is preceded by diminished E-cadherin expression and intestinal integrity. PLoS One 2019; 14:e0217220. [PMID: 31199830 PMCID: PMC6568390 DOI: 10.1371/journal.pone.0217220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/07/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic intestinal inflammation contributes to pathologies such as inflammatory bowel disease (IBD) and colon cancer. While the precise etiology remains controversial, IBD is believed to manifest as a result of various factors. We previously reported that intestinal-specific overexpression of the transcription factor Kaiso results in an intestinal inflammatory response; however, the cause of this inflammation is unknown. To elucidate the underlying mechanism(s) of the Kaiso-mediated intestinal inflammatory phenotype, we evaluated two independent transgenic mouse lines that express varying levels of Kaiso (KaisoTg). Histological analyses of KaisoTg mice revealed intestinal damage including thickening of the mucosa, intestinal “lesions” and crypt abscesses, which are reminiscent of IBD pathology. Additionally, higher Kaiso levels induced intestinal neutrophilia as early as 12 weeks, which worsened as the mice aged. Notably, the Kaiso-induced intestinal inflammation correlated with a leaky intestinal barrier and mis-regulation of E-cadherin expression and localization. Interestingly, Kaiso overexpression resulted in reduced proliferation but enhanced migration of intestinal epithelial cells prior to the onset of inflammation. Collectively, these data suggest that Kaiso plays a role in regulating intestinal epithelial cell integrity and function, dysregulation of which contributes to a chronic inflammatory phenotype as mice age.
Collapse
Affiliation(s)
| | - Roopali Chaudhary
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Rodrigo Jiménez-Saiz
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, Ontario, Canada
| | | | - Luke Bayer
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Manel Jordana
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, Ontario, Canada
| | - Juliet M. Daniel
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
26
|
Chang YL, Lo HY, Cheng SP, Chang KT, Lin XF, Lee SP, Hsieh MF, Chan CK. Therapeutic effects of a single injection of human umbilical mesenchymal stem cells on acute and chronic colitis in mice. Sci Rep 2019; 9:5832. [PMID: 30967579 PMCID: PMC6456500 DOI: 10.1038/s41598-019-41910-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 03/20/2019] [Indexed: 12/16/2022] Open
Abstract
Multiple injections of bone marrow mesenchymal stem cells (BMMSCs) have been used for treatment of chronic colitis in mice. We aimed to report the therapeutic effects of a single injection of human umbilical cord mesenchymal stem cells (hUCMSCs) on acute and chronic colitis. Male C57BL/6JNarl mice were divided into control, phosphate-buffered saline (PBS), and hUCMSCs treated groups, respectively. Acute and chronic colitis were induced in the mice (except controls) using 3% dextran sulfate sodium (DSS). The mice in the hUCMSCs group underwent a single injection of hUCMSCs. The disease activity index (DAI), colon length, histology, colon inflammation score, in vivo stem cells images, and blood cytokine levels were recorded. The DAI was significantly higher in the hUCMSCs group than in the control group and lower than in the PBS group on all days. The colon length was significantly longer and the colon inflammation score was significantly lower in the hUCMSCs group than in the PBS group on days 8 and 25. IL17A, Gro-α, MIP-1α, MIP-2, and eotaxin were significantly lower in the hUCMSCs group than in the PBS group on days 8 and 25. Single-injection hUCMSCs improved DSS–induced acute colitis and decreased progression of acute colitis to chronic colitis.
Collapse
Affiliation(s)
- Yu-Lung Chang
- Department of Urology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan.,Department of Biomedical Engineering, College of Engineering, Chung Yuan Christian University, Chung Li, Taiwan.,Department of Urology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Translational Medicine Center, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Huei-Yu Lo
- Department of Rehabilitation, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan.,Translational Medicine Center, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan.,Department of Chemistry, Chung Yuan Christian University, Chung Li, Taiwan
| | - Shun-Ping Cheng
- Department of Rehabilitation, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan.,Translational Medicine Center, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Kuo-Ting Chang
- Translational Medicine Center, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Xiu-Fang Lin
- Department of Biomedical Engineering, College of Engineering, Chung Yuan Christian University, Chung Li, Taiwan
| | - Sheng-Ping Lee
- Department of Urology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Ming-Fa Hsieh
- Department of Biomedical Engineering, College of Engineering, Chung Yuan Christian University, Chung Li, Taiwan.
| | - Chin-Kan Chan
- Department of Pediatrics, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan. .,Department of Biotechnology, School of Health Technology, Ming Chuan University, Taoyuan, Taiwan. .,Translational Medicine Center, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan.
| |
Collapse
|
27
|
Hillman M, Weström B, Aalaei K, Erlanson-Albertsson C, Wolinski J, Lozinska L, Sjöholm I, Rayner M, Landin-Olsson M. Skim milk powder with high content of Maillard reaction products affect weight gain, organ development and intestinal inflammation in early life in rats. Food Chem Toxicol 2018; 125:78-84. [PMID: 30553875 DOI: 10.1016/j.fct.2018.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/04/2018] [Accepted: 12/12/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND The intestinal tract is important for development of immune tolerance and disturbances are suggested to trigger autoimmune disorders. The aim of this study was to explore the effect of Maillard products in skim milk powder obtained after long storage, compared to fresh skim milk powder. METHODS Young rats were weaned onto a diet based on skim milk powder with high concentration of Maillard products (HM-SM, n = 18) or low (C-SM, n = 18) for one week or four weeks. Weekly body weight and feed consumption were noted. At the end, organ weights, intestinal histology, permeability and inflammatory cytokines were evaluated. RESULTS Rats fed with HM-SM had after one week, 15% less weight gain than controls, despite equal feed intake. After one week thymus and spleen were smaller, intestinal mucosa thickness was increased and acute inflammatory cytokines (IL-17, IL-1β, MCP-1) were elevated. After four weeks, cytokines associated with chronic intestinal inflammation (fractalkine, IP-10, leptin, LIX, MIP-2, RANTES and VEGF) were increased in rats fed with HM-SM compared to C-SM. CONCLUSION High content of Maillard products in stored milk powder caused an intestinal inflammation. Whether this is relevant for tolerance development and future autoimmune diseases remains to be explored.
Collapse
Affiliation(s)
- M Hillman
- Lund University, Faculty of Medicine, Department of Clinical Sciences, Lund, Sweden
| | - B Weström
- Lund University, Faculty of Science, Department of Biology, Lund, Sweden, Sweden
| | - K Aalaei
- Lund University, Faculty of Engineering, Department of Food Technology Engineering and Nutrition, Sweden
| | - C Erlanson-Albertsson
- Lund University, Faculty of Medicine, Department of Experimental Sciences, Lund, Sweden
| | - J Wolinski
- Polish Academy of Sciences, Kielanowski Institute of Animal Nutrition and Physiology, Department of Endocrinology, Jablonna, Poland
| | - L Lozinska
- Lund University, Faculty of Science, Department of Biology, Lund, Sweden, Sweden
| | - I Sjöholm
- Lund University, Faculty of Engineering, Department of Food Technology Engineering and Nutrition, Sweden
| | - M Rayner
- Lund University, Faculty of Engineering, Department of Food Technology Engineering and Nutrition, Sweden
| | - M Landin-Olsson
- Lund University, Faculty of Medicine, Department of Clinical Sciences, Lund, Sweden; Skane University Hospital, Department of Endocrinology, Lund, Sweden.
| |
Collapse
|
28
|
Chen F, Yang W, Huang X, Cao AT, Bilotta AJ, Xiao Y, Sun M, Chen L, Ma C, Liu X, Liu CG, Yao S, Dann SM, Liu Z, Cong Y. Neutrophils Promote Amphiregulin Production in Intestinal Epithelial Cells through TGF-β and Contribute to Intestinal Homeostasis. THE JOURNAL OF IMMUNOLOGY 2018; 201:2492-2501. [PMID: 30171165 DOI: 10.4049/jimmunol.1800003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/09/2018] [Indexed: 12/19/2022]
Abstract
Neutrophils are the first responders to sites of inflammation when the intestinal epithelial barrier is breached and the gut microbiota invade. Despite current efforts in understanding the role of neutrophils in intestinal homeostasis, the complex interactions between neutrophils and intestinal epithelial cells (IECs) is still not well characterized. In this study, we demonstrated that neutrophils enhanced production of amphiregulin (AREG), a member of the EGFR ligand family, by IECs, which promoted IEC barrier function and tissue repair. Depletion of neutrophils resulted in more severe colitis in mice because of decreased AREG production by IECs upon dextran sodium sulfate (DSS) insult. Administration of AREG restored epithelial barrier function and ameliorated colitis. Furthermore, neutrophil-derived TGF-β promoted AREG production by IECs. Mechanistically, TGF-β activated MEK1/2 signaling, and inhibition of MEK1/2 abrogated TGF-β-induced AREG production by IECs. Collectively, these findings reveal that neutrophils play an important role in the maintenance of IEC barrier function and homeostasis.
Collapse
Affiliation(s)
- Feidi Chen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555.,Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xiangsheng Huang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Anthony T Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Anthony J Bilotta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555.,Department of Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Yi Xiao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Mingming Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555.,Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Liang Chen
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555.,Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Chunyan Ma
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Xiuping Liu
- Department of Experimental Therapeutics, MD Anderson Cancer Center, University of Texas, Houston, TX 77230
| | - Chang-Gong Liu
- Department of Experimental Therapeutics, MD Anderson Cancer Center, University of Texas, Houston, TX 77230
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Sara M Dann
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555.,Department of Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Yingzi Cong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555; .,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
29
|
Dubé PE, Liu CY, Girish N, Washington MK, Polk DB. Pharmacological activation of epidermal growth factor receptor signaling inhibits colitis-associated cancer in mice. Sci Rep 2018; 8:9119. [PMID: 29904166 PMCID: PMC6002410 DOI: 10.1038/s41598-018-27353-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/30/2018] [Indexed: 12/15/2022] Open
Abstract
Current treatments for inflammatory bowel disease (IBD) target the overactive immune response of the intestinal mucosa. However, epidermal growth factor (EGF), an activating ligand of the EGF receptor (EGFR), has been shown to induce disease remission through direct targeting of intestinal mucosal healing. Despite promising preclinical and clinical results, this EGFR-activating therapy has not progressed, in part due to the potential for carcinogenesis associated with long-term use and the increased risk of colitis-associated cancer (CAC) in IBD. Here we tested whether pharmacological modulation of EGFR altered outcomes of CAC in the murine azoxymethane/dextran sulfate sodium model. We found that administering EGF during the period of maximum colitis severity ("early"), coincident with the initiation and early promotion of tumors, improved outcomes of colitis and reduced tumor size. In contrast, daily EGF administration beginning ~2 months after tumor initiation ("late") increased tumor size. Administration of the EGFR kinase inhibitor gefitinib increased the tumor size when the drug was given early and decreased the tumor size when the drug was administered late. EGF administration not only reduced colonic cytokine and chemokine expression during injury, but also baseline chemokine expression in homeostasis. These results suggest that EGFR activation during acute bouts of colitis may reduce the long-term burden of CAC.
Collapse
Affiliation(s)
- Philip E Dubé
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Taconic Biosciences, Hudson, NY, USA
| | - Cambrian Y Liu
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Nandini Girish
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - D Brent Polk
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Puzan M, Hosic S, Ghio C, Koppes A. Enteric Nervous System Regulation of Intestinal Stem Cell Differentiation and Epithelial Monolayer Function. Sci Rep 2018; 8:6313. [PMID: 29679034 PMCID: PMC5910425 DOI: 10.1038/s41598-018-24768-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/10/2018] [Indexed: 12/15/2022] Open
Abstract
The Enteric Nervous System (ENS) is a complex network of neurons and glia, which regulates sensorimotor function throughout the gastroinestinal tract (GI). Here we investigated the role of the ENS and intestinal myofibroblasts in the maintenance of a primary intestinal epithelial barrier through regulation of monolayer permeability, cytokine production, and differentiation of intestinal stem cells. Utilizing a novel, in vitro, transwell-based coculture system, murine small intestinal stem cells were isolated and cultured with ENS neurons and glia or subepithelial myofibroblasts. Results show that the ENS contributes to regulation of intestinal stem cell fate, promoting differentiation into chemosensory enteroendocrine cells, with 0.9% of cells expressing chromogranin A when cultured with ENS versus 0.6% in cocultures with myofibroblasts and 0.3% in epithelial cultures alone. Additionally, enteric neurons and myofibroblasts differentially release cytokines Macrophage Inflammatory Protein 2 (MIP-2), Transforming Growth Factor beta 1 (TGF-β1), and Interleukin 10 (IL-10) when cultured with intestinal epithelial cells, with a 1.5 fold increase of IL-10 and a 3 fold increase in MIP-2 in ENS cocultures compared to coculture with myofibroblasts. These results indicate the importance of enteric populations in the regulation of intestinal barrier function.
Collapse
Affiliation(s)
- Marissa Puzan
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Sanjin Hosic
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Caroline Ghio
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Abigail Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Biology, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Gatej SM, Marino V, Bright R, Fitzsimmons TR, Gully N, Zilm P, Gibson RJ, Edwards S, Bartold PM. Probiotic Lactobacillus rhamnosus GG
prevents alveolar bone loss in a mouse model of experimental periodontitis. J Clin Periodontol 2017; 45:204-212. [DOI: 10.1111/jcpe.12838] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Simona M. Gatej
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Victor Marino
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Richard Bright
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Tracy R. Fitzsimmons
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Neville Gully
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Peter Zilm
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Rachel J. Gibson
- Faculty of Health and Medical Sciences; Adelaide Medical School; The University of Adelaide; Adelaide SA Australia
- Division of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Suzanne Edwards
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| | - Peter M. Bartold
- Faculty of Health and Medical Sciences; Adelaide Dental School; The University of Adelaide; Adelaide SA Australia
| |
Collapse
|
32
|
Han X, Li B, Ye X, Mulatibieke T, Wu J, Dai J, Wu D, Ni J, Zhang R, Xue J, Wan R, Wang X, Hu G. Dopamine D 2 receptor signalling controls inflammation in acute pancreatitis via a PP2A-dependent Akt/NF-κB signalling pathway. Br J Pharmacol 2017; 174:4751-4770. [PMID: 28963856 PMCID: PMC5727253 DOI: 10.1111/bph.14057] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Dopamine has multiple anti-inflammatory effects, but its role and molecular mechanism in acute pancreatitis (AP) are unclear. We investigated the role of dopamine signalling in the inflammatory response in AP. EXPERIMENTAL APPROACH Changes in pancreatic dopaminergic system and effects of dopamine, antagonists and agonists of D1 and D2 dopamine receptors were analysed in wild-type and pancreas-specific Drd2-/- mice with AP (induced by caerulein and LPS or L-arginine) and pancreatic acinar cells with or without cholecystokinin (CCK) stimulation. The severity of pancreatitis was assessed by measuring serum amylase and lipase and histological assessments. The NF-κB signalling pathway was evaluated, and macrophage and neutrophil migration assessed by Transwell assay. KEY RESULTS Pancreatic dopamine synthetase and metabolic enzyme levels were increased, whereas D1 and D2 receptors were decreased in AP. Dopamine reduced inflammation in CCK-stimulated pancreatic acinar cells by inhibiting the NF-κB pathway. Moreover, the protective effects of dopamine were blocked by a D2 antagonist, but not a D1 antagonist. A D2 agonist reduced pancreatic damage and levels of p-IκBα, p-NF-κBp65, TNFα, IL-1β and IL-6 in AP. Pancreas-specific Drd2-/- aggravated AP. Also, the D2 agonist activated PP2A and inhibited the phosphorylation of Akt, IKK, IκBα and NF-κB and production of inflammatory cytokines and chemokines. Furthermore, it inhibited the migration of macrophages and neutrophils by reducing the expression of CCL2 and CXCL2. A PP2A inhibitor attenuated these protective effects of the D2 agonist. CONCLUSIONS AND IMPLICATIONS D2 receptors control pancreatic inflammation in AP by inhibiting NF-κB activation via a PP2A-dependent Akt signalling pathway.
Collapse
Affiliation(s)
- Xiao Han
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bin Li
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Ye
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tunike Mulatibieke
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianghong Wu
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Juanjuan Dai
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Deqing Wu
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ruling Zhang
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jing Xue
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Centre, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Rong Wan
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guoyong Hu
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
33
|
McLean MH, Andrews C, Hanson ML, Baseler WA, Anver MR, Senkevitch E, Staniszewska AK, Smith C, Davies LC, Hixon J, Li W, Shen W, Steidler L, Durum SK. Interleukin-27 Is a Potential Rescue Therapy for Acute Severe Colitis Through Interleukin-10-Dependent, T-Cell-Independent Attenuation of Colonic Mucosal Innate Immune Responses. Inflamm Bowel Dis 2017; 23:1983-1995. [PMID: 29019857 PMCID: PMC5796760 DOI: 10.1097/mib.0000000000001274] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND If treatment with intravenous steroids fail, inflammatory bowel disease patients with acute severe colitis face systemic anti-tumor necrosis factor biologic rescue therapy or colectomy. Interleukin (IL)-27 is a cytokine with an immunosuppressive role in adaptive immune responses. However, the IL-27 receptor complex is also expressed on innate immune cells, and there is evidence that IL-27 can impact the function of innate cell subsets, although this particular functionality in vivo is not understood. Our aim was to define the efficacy of IL-27 in acute severe colitis and characterize novel IL-27-driven mechanisms of immunosuppression in the colonic mucosa. METHODS We assessed oral delivery of Lactococcus lactis expressing an IL-27 hyperkine on the innate immune response in vivo in a genetically intact, noninfective, acute murine colitis model induced by intrarectal instillation of 2,4,6-trinitrobenzenesulfonic acid in SJL/J mice. RESULTS IL-27 attenuates acute severe colitis through the reduction of colonic mucosal neutrophil infiltrate associated with a decreased CXC chemokine gradient. This suppression was T cell independent and IL-10 dependent, initially featuring enhanced mucosal IL-10. IL-27 was associated with a reduction in colonic proinflammatory cytokines and induced a multifocal, strong, positive nuclear expression of phosphorylated STAT-1 in mucosal epithelial cells. CONCLUSION We have defined novel mechanisms of IL-27 immunosuppression toward colonic innate immune responses in vivo. Mucosal delivery of IL-27 has translational potential as a novel therapeutic for inflammatory bowel disease, and it is a future mucosal directed rescue therapy in acute severe inflammatory bowel disease.
Collapse
Affiliation(s)
- Mairi H McLean
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Scotland, AB25 2ZD, UK
| | - Caroline Andrews
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Miranda L Hanson
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Walter A Baseler
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Miriam R Anver
- Pathology/Histotechnology Laboratory, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21702, USA
| | - Emilee Senkevitch
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Aleksandra K Staniszewska
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Scotland, AB25 2ZD, UK
| | - Christopher Smith
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Scotland, AB25 2ZD, UK
| | - Luke C Davies
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Julie Hixon
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Wenqeng Li
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | - Wei Shen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| | | | - Scott K Durum
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, 21702, USA
| |
Collapse
|
34
|
Wunschel EJ, Schirmer B, Seifert R, Neumann D. Lack of Histamine H 4-Receptor Expression Aggravates TNBS-Induced Acute Colitis Symptoms in Mice. Front Pharmacol 2017; 8:642. [PMID: 28955241 PMCID: PMC5601386 DOI: 10.3389/fphar.2017.00642] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/30/2017] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are a growing health problem worldwide, severely affecting patients’ life qualities and life expectancies. Therapeutic options, which are rare and focus on symptoms associated with the disease, suffer from increasing numbers of patients refractory to the established strategies. Thus, in order to generate new therapeutic regimens, the detailed understanding of the pathogenic mechanisms causing IBD is necessary. Histamine is an inflammatory mediator associated with IBD. Four histamine receptors are currently known of which the histamine H4-receptor (H4R) has been shown to possess a pro-inflammatory function in several experimental models of inflammatory diseases, including dextran sodium sulfate (DSS)-induced colitis in mice. No single model reflects the complexity of human IBD, but each model provides valuable information on specific aspects of IBD pathogenesis. While DSS-induced colitis mostly relies on innate immune mechanisms, trinitrobenzene sulfonic acid (TNBS)-induced colitis rather reflects T-cell mechanisms. Consequently, an observation made in a single model has to be verified in at least one other model. Therefore, in the present study we investigated the effect of genetic blockade of H4R-signaling in mice subjected to the model of TNBS-induced acute colitis. We analyzed severity and progression of clinical signs of colitis, as well as histopathologic alterations in the colon and local cytokine production. Genetic ablation of H4R expression worsened clinical signs of acute colitis and histological appearance of colon inflammation after TNBS application. Moreover, TNBS instillation enhanced local synthesis of inflammatory mediators associated with a neutrophilic response, i.e., CXCL1, CXCL2, and interleukin-6. Lastly, also myeloperoxidase concentration, indicative for the presence of neutrophils, was elevated in cola of TNBS-treated mice due to the absence of H4R expression. Our results indicate an anti-inflammatory role of histamine via H4R in TNBS-induced acute neutrophilic colitis in mice, thus questioning the strategy of pharmacological H4R blocked as new therapeutic option for patients suffering from IBD.
Collapse
Affiliation(s)
- Eva J Wunschel
- Institute of Pharmacology, Hannover Medical SchoolHanover, Germany
| | - Bastian Schirmer
- Institute of Pharmacology, Hannover Medical SchoolHanover, Germany
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical SchoolHanover, Germany
| | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical SchoolHanover, Germany
| |
Collapse
|
35
|
Manresa MC, Taylor CT. Hypoxia Inducible Factor (HIF) Hydroxylases as Regulators of Intestinal Epithelial Barrier Function. Cell Mol Gastroenterol Hepatol 2017; 3:303-315. [PMID: 28462372 PMCID: PMC5404106 DOI: 10.1016/j.jcmgh.2017.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
Human health is dependent on the ability of the body to extract nutrients, fluids, and oxygen from the external environment while at the same time maintaining a state of internal sterility. Therefore, the cell layers that cover the surface areas of the body such as the lung, skin, and gastrointestinal mucosa provide vital semipermeable barriers that allow the transport of essential nutrients, fluid, and waste products, while at the same time keeping the internal compartments free of microbial organisms. These epithelial surfaces are highly specialized and differ in their anatomic structure depending on their location to provide appropriate and effective site-specific barrier function. Given this important role, it is not surprising that significant disease often is associated with alterations in epithelial barrier function. Examples of such diseases include inflammatory bowel disease, chronic obstructive pulmonary disease, and atopic dermatitis. These chronic inflammatory disorders often are characterized by diminished tissue oxygen levels (hypoxia). Hypoxia triggers an adaptive transcriptional response governed by hypoxia-inducible factors (HIFs), which are repressed by a family of oxygen-sensing HIF hydroxylases. Here, we review recent evidence suggesting that pharmacologic hydroxylase inhibition may be of therapeutic benefit in inflammatory bowel disease through the promotion of intestinal epithelial barrier function through both HIF-dependent and HIF-independent mechanisms.
Collapse
Key Words
- CD, Crohn’s disease
- DMOG, dimethyloxalylglycine
- DSS, dextran sodium sulfate
- Epithelial Barrier
- FIH, factor inhibiting hypoxia-inducible factor
- HIF, hypoxia-inducible factor
- Hypoxia
- Hypoxia-Inducible Factor (HIF) Hydroxylases
- IBD, inflammatory bowel disease
- IL, interleukin
- Inflammatory Bowel Disease
- NF-κB, nuclear factor-κB
- PHD, hypoxia-inducible factor–prolyl hydroxylases
- TFF, trefoil factor
- TJ, tight junction
- TLR, Toll-like receptor
- TNF-α, tumor necrosis factor α
- UC, ulcerative colitis
- ZO, zonula occludens
Collapse
Affiliation(s)
- Mario C. Manresa
- Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin, Ireland
- Charles Institute of Dermatology, Belfield, Dublin, Ireland
| | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin, Ireland
- Charles Institute of Dermatology, Belfield, Dublin, Ireland
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
36
|
Inoue R, Yajima T, Tsukahara T. Expression of TLR2 and TLR4 in murine small intestine during postnatal development. Biosci Biotechnol Biochem 2017; 81:350-358. [DOI: 10.1080/09168451.2016.1254534] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abstract
The important role played by the gut microbiota in host immunity is mediated, in part, through toll-like receptors (TLRs). We evaluated the postnatal changes in expression of TLR2 and TLR4 in the murine small intestine and assessed how expression is influenced by gut microbiota. The expression of TLR2 and TLR4 in the murine small intestine was highly dynamic during development. The changes were especially profound during the suckling period, with the maximal mRNA levels detected in the mid-suckling period. Immunohistochemical and flow-cytometric analyses indicated that the changes in TLR2 and TLR4 expression involve primarily epithelial cells. The germ-free mice showed minor changes in TLR2/TLR4 mRNA and TLR2 protein during the suckling period. This study demonstrated that the postnatal expression of TLR2 and TLR4 in small intestinal epithelial cells is dynamic and depends on the presence of commensal intestinal microbiota.
Collapse
Affiliation(s)
- Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan
- Creative Research Initiative “Sousei” (CRIS), Hokkaido University, Sapporo, Japan
| | - Takaji Yajima
- Creative Research Initiative “Sousei” (CRIS), Hokkaido University, Sapporo, Japan
| | - Takamitsu Tsukahara
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan
- Kyoto Institute of Nutrition and Pathology, Kyoto, Japan
| |
Collapse
|
37
|
Algieri F, Rodriguez-Nogales A, Vezza T, Garrido-Mesa J, Garrido-Mesa N, Utrilla MP, González-Tejero MR, Casares-Porcel M, Molero-Mesa J, Del Mar Contreras M, Segura-Carretero A, Pérez-Palacio J, Diaz C, Vergara N, Vicente F, Rodriguez-Cabezas ME, Galvez J. Anti-inflammatory activity of hydroalcoholic extracts of Lavandula dentata L. and Lavandula stoechas L. JOURNAL OF ETHNOPHARMACOLOGY 2016; 190:142-158. [PMID: 27269390 DOI: 10.1016/j.jep.2016.05.063] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/09/2016] [Accepted: 05/31/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plants from genus Lavandula have been used as anti-inflammatory drugs in Mediterranean traditional medicine. Nowadays, there is a growing interest for complementary medicine, including herbal remedies, to treat inflammatory bowel disease (IBD). AIM OF THE STUDY To test the anti-inflammatory properties of Lavandula dentata and Lavandula stoechas extracts in two inflammatory experimental models: TNBS model of rat colitis and the carrageenan-induced paw edema in mice, in order to mimic the intestinal conditions and the extra-intestinal manifestations of human IBD, respectively. MATERIAL AND METHODS The extracts were characterized through the qualitative HPLC analysis. Then, they were assayed in vitro and in vivo. In vitro studies were performed in BMDMs and CMT-93 epithelial cells with different concentrations of the extracts (ranging from 0.1 to 100µg/ml). The extracts were tested in vivo in the TNBS model of rat colitis (10 and 25mg/kg) and in the carrageenan-induced paw edema in mice (10, 25 and 100mg/kg). RESULTS L. dentata and L. stoechas extracts displayed immunomodulatory properties in vitro down-regulating different mediators of inflammation like cytokines and nitric oxide. They also showed anti-inflammatory effects in the TNBS model of colitis as evidenced by reduced myeloperoxidase activity and increased total glutathione content, indicating a decrease of neutrophil infiltration and an improvement of the oxidative state. Besides, both extracts modulated the expression of pro-inflammatory cytokines and chemokines, and ameliorated the altered epithelial barrier function. They also displayed anti-inflammatory effects in the carrageenan-induced paw edema in mice, since a significant reduction of the paw thickness was observed. This was associated with a down-regulation of the expression of different inducible enzymes like MMP-9, iNOS and COX-2 and pro-inflammatory cytokines, all involved in the maintenance of the inflammatory condition. CONCLUSION L. dentata and L. stoechas extracts showed intestinal anti-inflammatory effect, confirming their potential use as herbal remedies in gastrointestinal disorders. In addition, their anti-inflammatory effect was also observed in other locations, thus suggesting a possible use for the treatment of the extra-intestinal symptoms of IBD.
Collapse
Affiliation(s)
- Francesca Algieri
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Alba Rodriguez-Nogales
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Teresa Vezza
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Jose Garrido-Mesa
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Natividad Garrido-Mesa
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - M Pilar Utrilla
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | | | | | - Maria Del Mar Contreras
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain; Research and Development Centre for Functional Food (CIDAF), Health-Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Avenida Fuentenueva s/n, 18071 Granada, Spain; Research and Development Centre for Functional Food (CIDAF), Health-Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
| | - José Pérez-Palacio
- Fundación Centro de Excelencia en Investigación de Medicamentos Innovadores de Andalucía (MEDINA), Granada, Spain
| | - Caridad Diaz
- Fundación Centro de Excelencia en Investigación de Medicamentos Innovadores de Andalucía (MEDINA), Granada, Spain
| | - Noemí Vergara
- Fundación Centro de Excelencia en Investigación de Medicamentos Innovadores de Andalucía (MEDINA), Granada, Spain
| | - Francisca Vicente
- Fundación Centro de Excelencia en Investigación de Medicamentos Innovadores de Andalucía (MEDINA), Granada, Spain
| | - M Elena Rodriguez-Cabezas
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Julio Galvez
- CIBER-EHD, Department of Pharmacology, IBS.GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain.
| |
Collapse
|
38
|
Gabrielli E, Sabbatini S, Roselletti E, Kasper L, Perito S, Hube B, Cassone A, Vecchiarelli A, Pericolini E. In vivo induction of neutrophil chemotaxis by secretory aspartyl proteinases of Candida albicans. Virulence 2016; 7:819-25. [PMID: 27127904 PMCID: PMC5029300 DOI: 10.1080/21505594.2016.1184385] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Secretory aspartyl proteinases (Saps) of Candida albicans are key virulence traits which cause inflammasome-dependent, aseptic inflammation in a mouse model of vaginitis. In this paper, neutrophil migration in response to Sap2, Sap6 and chemo-attractive products released from Sap-treated vaginal epithelium was measured in vitro, ex vivo and in vivo. Our results show that Sap2 and Sap6 induce neutrophil migration and production of potent chemoattractive chemokines such as IL-8 and MIP-2 by vaginal epithelial cells. Our data suggest that at least part of MIP-2 production depends upon IL-1β activity. The vaginal fluid of Candida-infected mice contained a heat-labile inhibitor of neutrophil candidacidal activity that was absent from the vaginal fluid of Sap-treated mice. Overall, our data provide additional information on the capacity of C. albicans Saps to cause aseptic vaginal inflammation and highlight the potential role of some chemokines released from vaginal epithelial cells in this phenomenon.
Collapse
Affiliation(s)
- Elena Gabrielli
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Samuele Sabbatini
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Elena Roselletti
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Lydia Kasper
- b Department of Microbial Pathogenicity Mechanisms , Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute , Jena , Germany
| | - Stefano Perito
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Bernhard Hube
- b Department of Microbial Pathogenicity Mechanisms , Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute , Jena , Germany.,c Friedrich Schiller University , Jena , Germany.,d Center for Sepsis Control and Care (CSCC) , Jena , Germany
| | - Antonio Cassone
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Anna Vecchiarelli
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| | - Eva Pericolini
- a Department of Experimental Medicine , University of Perugia , Perugia , Italy
| |
Collapse
|
39
|
Long-Term Relationships: the Complicated Interplay between the Host and the Developmental Stages of Toxoplasma gondii during Acute and Chronic Infections. Microbiol Mol Biol Rev 2016; 79:387-401. [PMID: 26335719 DOI: 10.1128/mmbr.00027-15] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Toxoplasma gondii represents one of the most common parasitic infections in the world. The asexual cycle can occur within any warm-blooded animal, but the sexual cycle is restricted to the feline intestinal epithelium. T. gondii is acquired through consumption of tissue cysts in undercooked meat as well as food and water contaminated with oocysts. Once ingested, it differentiates into a rapidly replicating asexual form and disseminates throughout the body during acute infection. After stimulation of the host immune response, T. gondii differentiates into a slow-growing, asexual cyst form that is the hallmark of chronic infection. One-third of the human population is chronically infected with T. gondii cysts, which can reactivate and are especially dangerous to individuals with reduced immune surveillance. Serious complications can also occur in healthy individuals if infected with certain T. gondii strains or if infection is acquired congenitally. No drugs are available to clear the cyst form during the chronic stages of infection. This therapeutic gap is due in part to an incomplete understanding of both host and pathogen responses during the progression of T. gondii infection. While many individual aspects of T. gondii infection are well understood, viewing the interconnections between host and parasite during acute and chronic infection may lead to better approaches for future treatment. The aim of this review is to provide an overview of what is known and unknown about the complex relationship between the host and parasite during the progression of T. gondii infection, with the ultimate goal of bridging these events.
Collapse
|
40
|
Anticoccidial activities of Chitosan on Eimeria papillata-infected mice. Parasitol Res 2016; 115:2845-52. [PMID: 27041340 DOI: 10.1007/s00436-016-5035-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/27/2016] [Indexed: 12/28/2022]
Abstract
Eimeria spp. multiply within the intestinal tract causing severe inflammatory responses. Chitosan (CS), meanwhile, has been shown to exhibit anti-inflammatory activities in different experimental models. Here, we investigated the effect of CS on the outcome of inflammation caused by Eimeria papillata in the mouse intestine. Investigations were undertaken into the oocyst output in feces and developmental stages and goblet cells in intestinal tissue. Assays for lipid peroxidation, nitric oxide (NO), and myeloperoxidase (MPO) were also performed. T cells in intestinal tissue were counted using immunohistochemistry while total IgA in serum or intestinal wash was assayed using ELISA. In addition, mRNA expression of tumor necrosis factor alpha (TNF-α), transforming growth factor β (TGF-β), interleukin (IL)-10, and IL-4 were detected using real-time PCR. The data indicated a reduction in both oocyst output and in the number of parasite developmental stages following CS treatment, while the goblet cell hypoplasia in infected mice was also inhibited. CS decreased lipid peroxidation, NO, and MPO but did not alter the T cell count or IgA levels in comparison to the infected group. The expression of TNF-α and TGF-β decreased but IL-10 and IL-4 increased after CS treatment in comparison to the non-treated infected group. In conclusion, CS showed anti-inflammatory and protective effects against E. papillata infection.
Collapse
|
41
|
Brand C, da Costa TP, Bernardes ES, Machado CML, Andrade LR, Chammas R, de Oliveira FL, El-Cheikh MC. Differential development of oil granulomas induced by pristane injection in galectin-3 deficient mice. BMC Immunol 2015; 16:68. [PMID: 26572128 PMCID: PMC4647586 DOI: 10.1186/s12865-015-0133-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022] Open
Abstract
Background Galectin-3 is known to be a lectin that plays an important role in inflammatory processes, acting as pro-inflammatory mediator in activation and migration of neutrophils and macrophages, as well as in the phagocytic function of these cells. The injection of mineral oils into the peritoneal cavity of mice, such as 2, 6, 10, 14-tetramethylpentadecane (pristane), induce a chronic granulomatous inflammatory reaction which is rich in macrophages, B cells and peritoneal plasma cells known as oil granuloma. In addition, this inflammatory microenvironment provided by oil granulomas is also an important site of plasmacytoma induction, which are dependent on cytokine production and cellular mobilization. Here, we have analyzed the role of galectin-3 in inflammatory cells mobilization and organization after pristane injection characterizing granulomatous reaction through the formation of oil granulomas. Results In galectin-3 deficient mice (gal-3−/−), the mobilization of inflammatory cells, between peritoneal cavity and bone marrow, was responsible for the formation of disorganized oil granulomas, which presented scattered cells, large necrotic areas and low amounts of extracellular matrix. The production of inflammatory cytokines partially explained the distribution of cells through peritoneal cavity, since high levels of IL-6 in gal-3−/− mice led to drastically reduction of B1 cells. The previous pro-inflammatory status of these animals also explains the excess of cell death and disruption of oil granulomas architecture. Conclusions Our data indicate, for the first time, that the disruption in the inflammatory cells migration in the absence of galectin-3 is a crucial event in the formation and organization of oil granulomas. Electronic supplementary material The online version of this article (doi:10.1186/s12865-015-0133-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camila Brand
- Laboratório de Proliferação e Diferenciação Celular, Programa de Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| | - Thayse Pinheiro da Costa
- Laboratório de Proliferação e Diferenciação Celular, Programa de Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| | - Emerson Soares Bernardes
- Centro de Radiofarmácia, Instituto de Pesquisas Energéticas Nucleares (IPEN), São Paulo, Brazil.
| | - Camila Maria Longo Machado
- Laboratório de Oncologia Experimental-LIM24, Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil. .,Laboratório de Investigação Médica Radioisótopos-LIM43, Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil.
| | - Leonardo Rodrigues Andrade
- Laboratório de Biomineralização, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| | - Roger Chammas
- Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brasil.
| | - Felipe Leite de Oliveira
- Laboratório de Proliferação e Diferenciação Celular, Programa de Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| | - Márcia Cury El-Cheikh
- Laboratório de Proliferação e Diferenciação Celular, Programa de Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
42
|
Ssemakalu CC, Ubomba-Jaswa E, Motaung KS, Pillay M. The Effect of Solar Irradiated Vibrio cholerae on the Secretion of Pro-Inflammatory Cytokines and Chemokines by the JAWS II Dendritic Cell Line In Vitro. PLoS One 2015; 10:e0130190. [PMID: 26066787 PMCID: PMC4465907 DOI: 10.1371/journal.pone.0130190] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 05/16/2015] [Indexed: 11/18/2022] Open
Abstract
The use of solar irradiation to sterilize water prior to its consumption has resulted in the reduction of water related illnesses in waterborne disease endemic communities worldwide. Currently, research on solar water disinfection (SODIS) has been directed towards understanding the underlying mechanisms through which solar irradiation inactivates the culturability of microorganisms in water, enhancement of the disinfection process, and the health impact of SODIS water consumption. However, the immunological consequences of SODIS water consumption have not been explored. In this study, we investigated the effect that solar irradiated V. cholerae may have had on the secretion of cytokines and chemokines by the JAWS II dendritic cell line in vitro. The JAWS II dendritic cell line was stimulated with the different strains of V. cholerae that had been: (i) prepared in PBS, (ii) inactivated through a combination of heat and chemical, (iii) solar irradiated, and (iv) non-solar irradiated, in bottled water. As controls, LPS (1 μg/ml) and CTB (1 μg/ml) were used as stimulants. After 48 hours of stimulation the tissue culture media from each treatment was qualitatively and quantitatively analysed for the presence of IL-1α, IL-1β, IL-6, IL-7, IL-10, IL-12p40, IL-12p70, IL-15, MIP-1α, MIP-1β, MIP-2, RANTES, TNF-α, IL-23 and IL-27. Results showed that solar irradiated cultures of V. cholerae induced dendritic cells to secrete significant (p<0.05) levels of pro-inflammatory cytokines in comparison to the unstimulated dendritic cells. Furthermore, the amount of pro-inflammatory cytokines secreted by the dendritic cells in response to solar irradiated cultures of V. cholerae was not as high as observed in treatments involving non-solar irradiated cultures of V. cholerae or LPS. Our results suggest that solar irradiated microorganisms are capable of inducing the secretion of pro-inflammatory cytokines and chemokines. This novel finding is key towards understanding the possible immunological consequences of consuming SODIS treated water.
Collapse
Affiliation(s)
- Cornelius Cano Ssemakalu
- Department of Biotechnology, Faculty of Applied and Computer Sciences, Vaal University of Technology, Vanderbijlpark, 1900, Gauteng, South Africa
| | - Eunice Ubomba-Jaswa
- Council for Scientific and Industrial Research, Natural Resources and the Environment, P.O. Box 395, Pretoria, 0001, Gauteng, South Africa
| | - Keolebogile Shirley Motaung
- Department of Biomedical Sciences, Tshwane University of Technology, 175 Nelson Mandela Drive, Arcadia Campus, Pretoria, 0001, Gauteng, South Africa
| | - Michael Pillay
- Department of Biotechnology, Faculty of Applied and Computer Sciences, Vaal University of Technology, Vanderbijlpark, 1900, Gauteng, South Africa
- * E-mail:
| |
Collapse
|
43
|
Domingos-Pereira S, Hojeij R, Reggi E, Derré L, Chevalier MF, Romero P, Jichlinski P, Nardelli-Haefliger D. Local Salmonella immunostimulation recruits vaccine-specific CD8 T cells and increases regression of bladder tumor. Oncoimmunology 2015; 4:e1016697. [PMID: 26140240 DOI: 10.1080/2162402x.2015.1016697] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 02/02/2015] [Accepted: 02/03/2015] [Indexed: 12/13/2022] Open
Abstract
The efficacy of antitumoral responses can be increased using combinatorial vaccine strategies. We recently showed that vaccination could be optimized by local administration of diverse molecular or bacterial agents to target and augment antitumoral CD8 T cells in the genital mucosa (GM) and increase regression of cervical cancer in an animal model. Non muscle-invasive bladder cancer is another disease that is easily amenable to local therapies. In contrast to data obtained in the GM, in this study we show that intravesical (IVES) instillation of synthetic toll-like receptor (TLR) agonists only modestly induced recruitment of CD8 T cells to the bladder. However, IVES administration of Ty21a, a live bacterial vaccine against typhoid fever, was much more effective and increased the number of total and vaccine-specific CD8 T cells in the bladder approximately 10 fold. Comparison of chemokines induced in the bladder by either CpG (a TLR-9 agonist) or Ty21a highlighted the preferential increase in complement component 5a, CXCL5, CXCL2, CCL8, and CCL5 by Ty21a, suggesting their involvement in the attraction of T cells to the bladder. IVES treatment with Ty21a after vaccination also significantly increased tumor regression compared to vaccination alone, resulting in 90% survival in an orthotopic murine model of bladder cancer expressing a prototype tumor antigen. Our data demonstrate that combining vaccination with local immunostimulation may be an effective treatment strategy for different types of cancer and also highlight the great potential of the Ty21a vaccine, which is routinely used worldwide, in such combinatorial therapies.
Collapse
Key Words
- BCG, Bacillus Calmette Guerin
- BMDC, bone marrow-derived dendritic cell
- C5a, complement component 5a
- ESL, E-selectin ligands
- GM, genital mucosa
- IVAG, intravaginal
- IVES, intravesical
- NMIBC, non-muscle invasive bladder cancer
- PBS, phosphate buffered saline
- PE, phycoerythrin
- PIC, poly (I:C)
- SEM, standard error of the mean
- Salmonella Ty21a
- TLR, toll-like receptor
- TUR, transurethral resection
- bacterial immunostimulant
- bladder cancer
- combinatorial therapy
- s.c., subcutaneously
- therapeutic vaccination
Collapse
Affiliation(s)
| | - Rim Hojeij
- Dept. Urology; Lausanne University Hospital (CHUV); Lausanne, Switzerland
| | - Erica Reggi
- Dept. Urology; Lausanne University Hospital (CHUV); Lausanne, Switzerland
| | - Laurent Derré
- Dept. Urology; Lausanne University Hospital (CHUV); Lausanne, Switzerland
| | | | - Pedro Romero
- Ludwig Center for Cancer Research of University of Lausanne ; Lausanne, Switzerland
| | - Patrice Jichlinski
- Dept. Urology; Lausanne University Hospital (CHUV); Lausanne, Switzerland
| | | |
Collapse
|
44
|
Araujo AP, Giorgio S. Immunohistochemical evidence of stress and inflammatory markers in mouse models of cutaneous leishmaniosis. Arch Dermatol Res 2015; 307:671-82. [PMID: 25896942 DOI: 10.1007/s00403-015-1564-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/06/2015] [Accepted: 04/11/2015] [Indexed: 01/20/2023]
Abstract
Leishmanioses are chronic parasitic diseases and host responses are associated with pro- or anti-inflammatory cytokines involved, respectively, in the control or exacerbation of infection. The relevance of other inflammatory mediators and stress markers has not been widely studied and there is a need to search for biomarkers to leishmaniasis. In this work, the stress and inflammatory molecules p38 mitogen-activated protein kinase, cyclooxygenase-2, migration inhibitory factor, macrophage inflammatory protein 2, heat shock protein 70 kDa, vascular endothelial factor (VEGF), hypoxia-inducible factors (HIF-1α and HIF-2α), heme oxygenase and galectin-3 expression were assessed immunohistochemically in self-controlled lesions in C57BL/6 mice and severe lesions in Balb/c mice infected with Leishmania amazonensis. The results indicated that the majority of molecules were expressed in the cutaneous lesions of both C57BL/6 and Balb/c mice during various phases of infection, suggesting no obvious correlation between the stress and inflammatory molecule expression and the control/exacerbation of leishmanial lesions. However, the cytokine VEGF was only detected in C57BL/6 footpad lesions and small lesions in Balb/c mice treated with antimonial pentavalent. These findings suggest that VEGF expression could be a predictive factor for murine leishmanial control, a hypothesis that should be tested in human leishmaniosis.
Collapse
Affiliation(s)
- Alexandra Paiva Araujo
- Department of Animal Biology, Biology Institute, Universidade Estadual de Campinas, Caixa Postal 6109, Campinas, São Paulo, 13083-970, Brazil
| | - Selma Giorgio
- Department of Animal Biology, Biology Institute, Universidade Estadual de Campinas, Caixa Postal 6109, Campinas, São Paulo, 13083-970, Brazil.
| |
Collapse
|
45
|
Ohtsuka Y. Food intolerance and mucosal inflammation. Pediatr Int 2015; 57:22-9. [PMID: 25442377 DOI: 10.1111/ped.12546] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/28/2014] [Accepted: 11/17/2014] [Indexed: 01/13/2023]
Abstract
Most infants are immunologically active and are able to develop a tolerance to oligoclonal antigens by producing IgA, along with activation of regulatory T cells, in early infancy. Cytokines and their signaling molecules are important mediators in the intestine, regulating both oral tolerance and mucosal inflammation. This system works efficiently in most individuals, but for an as yet undefined reason, some people react to food and other proteins as though they were pathogens, with induction of chronic inflammation in the mucosa. The adverse reaction caused by ingested foods is defined as food intolerance. The clinical features of food intolerance include vomiting, diarrhea, bloody stool, eczema, failure to thrive, and a protean range of other symptoms. Intolerance can be divided into two categories depending on whether or not they are immunologically mediated. Food intolerance and mucosal inflammation are deeply related because tolerance cannot be established when there is an inflammation in the intestinal mucosa. Mast cells, eosinophils, mucosal lymphocytes, and epithelial cells are deeply involved and related to each other in the development of mucosal inflammation. Meanwhile, rectal bleeding in infancy is related to lymphoid hyperplasia with eosinophil infiltration into the colonic mucosa facilitated by C-C motif ligand 11 (CCL11, known as eotaxin-1) and C-X-C motif chemokine ligand 13 (CXCL13). Rectal bleeding in infancy may not be simply caused by allergic reactions against specific antigens, but may be due to migrated lymphocytes developing immunological tolerance; including IgA synthesizing, in the intestinal mucosa.
Collapse
Affiliation(s)
- Yoshikazu Ohtsuka
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
Leptin receptor mutation results in defective neutrophil recruitment to the colon during Entamoeba histolytica infection. mBio 2014; 5:mBio.02046-14. [PMID: 25516614 PMCID: PMC4271549 DOI: 10.1128/mbio.02046-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Amebiasis is an enteric infection caused by Entamoeba histolytica, with symptoms ranging in severity from asymptomatic colonization to dysentery. Humans with the Q223R leptin receptor mutation have increased susceptibility to amebiasis, but the mechanism has been unclear. Using a mouse model expressing the mutation, we tested the impact of the Q223R mutation on the innate immune response to E. histolytica infection. The 223R mutation resulted in delayed clearance of amebae from the cecum, as had been previously observed. We found that neutrophil influx to the site of the infection was reduced 12 h after infection in 223R mice. Depletion of neutrophils with anti-Ly6G monoclonal antibody increased susceptibility of wild-type mice to infection, supporting the importance of neutrophils in innate defense. Leptin expression was increased in the cecum by E. histolytica infection, suggesting that leptin could serve as a homing signal for neutrophils to the gut. Interestingly, neutrophils from mice with the 223R mutation had diminished chemotaxis toward leptin. This impaired chemotaxis likely explained the reduced gut infiltration of neutrophils. The newly recognized effect of the leptin receptor Q223R mutation on neutrophil chemotaxis and the impact of this mutation on multiple infectious diseases suggest a broader impact of this mutation on susceptibility to disease. The Q223R leptin receptor mutation results in increased susceptibility of children and adults to E. histolytica, one of the leading causes of diarrhea morbidity and mortality in children of the developing world. Here we show that the mutation results in reduced neutrophil infiltration to the site of infection. This decreased infiltration is likely due to the mutation’s impact on neutrophil chemotaxis toward leptin, an inflammatory agent upregulated in the cecum after infection. The significance of this work thus extends beyond understanding E. histolytica susceptibility by also providing insight into the potential impact of leptin on neutrophil function in other states of altered leptin signaling, which include both malnutrition and obesity.
Collapse
|
47
|
Hornef MW, Fulde M. Ontogeny of intestinal epithelial innate immune responses. Front Immunol 2014; 5:474. [PMID: 25346729 PMCID: PMC4191320 DOI: 10.3389/fimmu.2014.00474] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/16/2014] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence indicates that processes during postnatal development might significantly influence the establishment of mucosal host-microbial homeostasis. Developmental and adaptive immunological processes but also environmental and microbial exposure early after birth might thus affect disease susceptibility and health during adult life. The present review aims at summarizing the current understanding of the intestinal epithelial innate immune system and its developmental and adaptive changes after birth.
Collapse
Affiliation(s)
- Mathias W Hornef
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School , Hannover , Germany ; Institute of Medical Microbiology, RWTH University , Aachen , Germany
| | - Marcus Fulde
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School , Hannover , Germany
| |
Collapse
|
48
|
Kristek M, Collins LE, DeCourcey J, McEvoy FA, Loscher CE. Soluble factors from colonic epithelial cells contribute to gut homeostasis by modulating macrophage phenotype. Innate Immun 2014; 21:358-69. [PMID: 25298104 DOI: 10.1177/1753425914538294] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 05/06/2014] [Indexed: 11/17/2022] Open
Abstract
Intestinal macrophages originate from inflammatory blood monocytes which migrate to the intestine, where they differentiate into anti-inflammatory macrophages through a number of transitional stages. These macrophages typically remain hypo-responsive to commensal bacteria and food Ags in the intestine, yet also retain the ability to react to invading pathogens. In this study we examined the role of epithelial cells in inducing this intestinal macrophage phenotype. Using an in vitro system we showed that, in two-dimensional culture, epithelial cell-derived factors from a murine cell line, CMT-93, are sufficient to induce phenotypic changes in macrophages. Exposure of monocyte-derived macrophages, J774A.1, to soluble factors derived from epithelial cells, induced an altered phenotype similar to that of intestinal macrophages with decreased production of IL-12p40, IL-6 and IL-23 and expression of MHC ІІ and CD80 following TLR ligation. Furthermore, these conditioned macrophages showed enhanced phagocytic activity in parallel with low respiratory burst and NO production, similar to the response seen in intestinal macrophages. Our findings suggest a role for colonic epithelial cells in modulation of macrophage phenotype for maintenance of gut homeostasis. Further understanding of the cell interactions that maintain homeostasis in the gut could reveal novel therapeutic strategies to restore the balance in disease.
Collapse
Affiliation(s)
- Maja Kristek
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Ireland
| | - Laura E Collins
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Ireland
| | - Joseph DeCourcey
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Ireland
| | - Fiona A McEvoy
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Ireland
| | - Christine E Loscher
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Ireland
| |
Collapse
|
49
|
Azuma K, Osaki T, Tsuka T, Imagawa T, Okamoto Y, Minami S. Effects of fish scale collagen peptide on an experimental ulcerative colitis mouse model. PHARMANUTRITION 2014. [DOI: 10.1016/j.phanu.2014.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Azuma K, Osaki T, Kurozumi S, Kiyose M, Tsuka T, Murahata Y, Imagawa T, Itoh N, Minami S, Sato K, Okamoto Y. Anti-inflammatory effects of orally administered glucosamine oligomer in an experimental model of inflammatory bowel disease. Carbohydr Polym 2014; 115:448-56. [PMID: 25439918 DOI: 10.1016/j.carbpol.2014.09.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/08/2014] [Accepted: 09/11/2014] [Indexed: 01/12/2023]
Abstract
Anti-inflammatory effects of oral administration of the glucosamine oligomers (chito-oligosaccharides: COS) were evaluated in an experimental model of inflammatory bowel disease (IBD). Oral administration of COS improved shortening of colon length and tissue injury (as assessed by histology) in mice. Oral administration of COS inhibited inflammation in the colonic mucosa by suppression of myeloperoxidase activation in inflammatory cells, as well as activation of nuclear factor-kappa B, cyclooxygenase-2, and inducible nitric oxide synthase. Oral administration of COS also reduced serum levels of pro-inflammatory cytokines (tumor necrosis factor-α and interleukin-6). Moreover, it prolonged survival time in mice. These data suggest that COS have anti-inflammatory effects in an experimental model of IBD, and could be new functional foods for IBD patients.
Collapse
Affiliation(s)
- Kazuo Azuma
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan.
| | - Tomohiro Osaki
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan
| | - Seiji Kurozumi
- Koyo Chemical Co. Ltd., 3-11-15 Iidabashi, Chiyodaku, Tokyo 102-0072, Japan
| | - Masatoshi Kiyose
- Koyo Chemical Co. Ltd., 3-11-15 Iidabashi, Chiyodaku, Tokyo 102-0072, Japan
| | - Takeshi Tsuka
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan
| | - Yusuke Murahata
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan
| | - Tomohiro Imagawa
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan
| | - Norihiko Itoh
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan
| | - Saburo Minami
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan
| | - Kimihiko Sato
- Koyo Chemical Co. Ltd., 3-11-15 Iidabashi, Chiyodaku, Tokyo 102-0072, Japan
| | - Yoshiharu Okamoto
- Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Tottori University, 4-101 Koyama-minami, Tottori 680-8553, Japan.
| |
Collapse
|