1
|
Nagayama I, Takei Y, Takahashi S, Okada M, Maeshima A. The activin-follistatin system: Key regulator of kidney development, regeneration, inflammation, and fibrosis. Cytokine Growth Factor Rev 2024:S1359-6101(24)00092-3. [PMID: 39581798 DOI: 10.1016/j.cytogfr.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Activins, multifunctional cytokines of the transforming growth factor-beta superfamily, play critical roles in the regulation of growth and differentiation in multiple biological systems. Activin activity is finely regulated by the endogenous antagonist follistatin. Early studies reported that activins are involved in renal organogenesis, but subsequent research demonstrated that activins also play a significant role in kidney regeneration following injury. The results of more recent studies suggest activins play roles in both inflammatory kidney diseases and renal fibrosis, conditions that often culminate in end-stage renal disease. Given these findings, the inhibition of activin activity represents a promising therapeutic approach for treating a range of kidney disorders. This review discusses the latest discoveries concerning the role of the activin-follistatin system in renal development and pathophysiology and explores the potential therapeutic implications of targeting this system in the management of kidney diseases.
Collapse
Affiliation(s)
- Izumi Nagayama
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan
| | | | - Shunsuke Takahashi
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan
| | - Mari Okada
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan
| | - Akito Maeshima
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, Japan.
| |
Collapse
|
2
|
Ozaka S, Sonoda A, Kudo Y, Ito K, Kamiyama N, Sachi N, Chalalai T, Kagoshima Y, Soga Y, Ekronarongchai S, Ariki S, Mizukami K, Ishizawa S, Nishiyama M, Murakami K, Takeda K, Kobayashi T. Daikenchuto, a Japanese herbal medicine, ameliorates experimental colitis in a murine model by inducing secretory leukocyte protease inhibitor and modulating the gut microbiota. Front Immunol 2024; 15:1457562. [PMID: 39524440 PMCID: PMC11543465 DOI: 10.3389/fimmu.2024.1457562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) is a refractory inflammatory disorder of the intestine, which is probably triggered by dysfunction of the intestinal epithelial barrier. Secretory leukocyte protease inhibitor (SLPI) secreted by colon epithelial cells protects against intestinal inflammation by exerting anti-protease and anti-microbial activities. Daikenchuto (DKT) is one of the most commonly prescribed Japanese traditional herbal medicines for various digestive diseases. Although several animal studies have revealed that DKT exerts anti-inflammatory effects, its detailed molecular mechanism is unclear. This study aimed to clarify the anti-inflammatory mechanism of DKT using a murine colitis model, and to evaluate its potential as a therapeutic agent for IBD. Methods Experimental colitis was induced in wild-type (WT) mice and SLPI-deficient (KO) mice by dextran sulfate sodium (DSS) after oral administration of DKT. The resultant clinical symptoms, histological changes, and pro-inflammatory cytokine levels in the colon were assessed. Expression of SLPI in the colon was detected by Western blotting and immunohistochemistry. Composition of the gut microbiota was analyzed by 16S rRNA metagenome sequencing and intestinal metabolites were measured by gas chromatography-mass spectrometry analysis. Intestinal epithelial barrier function was assessed by oral administration of FITC-dextran and immunostaining of tight junction proteins (TJPs). Results Oral administration of DKT increased the number of butyrate-producing bacteria, such as Parabacteroides, Allobaculum, and Akkermansia, enhanced the levels of short-chain fatty acids, including butyrate, in the colon, induced SLPI expression, and ameliorated DSS-induced colitis in WT mice. We found that mouse colon carcinoma cell line treatment with either DKT or butyrate significantly enhanced the expression of SLPI. Moreover, supplementation of DKT protected the intestinal epithelial barrier with augmented expression of TJPs in WT mice, but not in KO mice. Finally, the composition of the gut microbiota was changed by DKT in WT mice, but not in KO mice, suggesting that DKT alters the colonic bacterial community in an SLPI-dependent manner. Conclusion These results indicate that DKT exerts anti-inflammatory effects on the intestinal epithelial barrier by SLPI induction, due, at least in part, to increased butyrate-producing bacteria and enhanced butyrate levels in the colon. These results provide insight into the mechanism of the therapeutic effects of DKT on IBD.
Collapse
Affiliation(s)
- Sotaro Ozaka
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Akira Sonoda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yoko Kudo
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kanako Ito
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Nozomi Sachi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Thanyakorn Chalalai
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yomei Kagoshima
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yasuhiro Soga
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | | | - Shimpei Ariki
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Shiori Ishizawa
- Tsumura Advanced Technology Research Laboratories, Research and Development Division, Tsumura & Co., Inashiki, Japan
| | - Mitsue Nishiyama
- Tsumura Advanced Technology Research Laboratories, Research and Development Division, Tsumura & Co., Inashiki, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
- Research Center for GLOBAL and LOCAL Infectious Diseases, Oita University, Yufu, Japan
| |
Collapse
|
3
|
Jiang BC, Ling YJ, Xu ML, Gu J, Wu XB, Sha WL, Tian T, Bai XH, Li N, Jiang CY, Chen O, Ma LJ, Zhang ZJ, Qin YB, Zhu M, Yuan HJ, Wu LJ, Ji RR, Gao YJ. Follistatin drives neuropathic pain in mice through IGF1R signaling in nociceptive neurons. Sci Transl Med 2024; 16:eadi1564. [PMID: 39413164 DOI: 10.1126/scitranslmed.adi1564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Neuropathic pain is a debilitating chronic condition that lacks effective treatment. The role of cytokine- and chemokine-mediated neuroinflammation in its pathogenesis has been well documented. Follistatin (FST) is a secreted protein known to antagonize the biological activity of cytokines in the transforming growth factor-β (TGF-β) superfamily. The involvement of FST in neuropathic pain and the underlying mechanism remain largely unknown. Here, we report that FST was up-regulated in A-fiber sensory neurons after spinal nerve ligation (SNL) in mice. Inhibition or deletion of FST alleviated neuropathic pain and reduced the nociceptive neuron hyperexcitability induced by SNL. Conversely, intrathecal or intraplantar injection of recombinant FST, or overexpression of FST in the dorsal root ganglion (DRG) neurons, induced pain hypersensitivity. Furthermore, exogenous FST increased neuronal excitability in nociceptive neurons. The biolayer interferometry (BLI) assay and coimmunoprecipitation (co-IP) demonstrated direct binding of FST to the insulin-like growth factor-1 receptor (IGF1R), and IGF1R inhibition reduced FST-induced activation of extracellular signal-regulated kinase (ERK) and protein kinase B (AKT), as well as neuronal hyperexcitability. Further co-IP analysis revealed that the N-terminal domain of FST exhibits the highest affinity for IGF1R, and blocking this interaction with a peptide derived from FST attenuated Nav1.7-mediated neuronal hyperexcitability and neuropathic pain after SNL. In addition, FST enhanced neuronal excitability in human DRG neurons through IGF1R. Collectively, our findings suggest that FST, released from A-fiber neurons, enhances Nav1.7-mediated hyperexcitability of nociceptive neurons by binding to IGF1R, making it a potential target for neuropathic pain treatment.
Collapse
Affiliation(s)
- Bao-Chun Jiang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Yue-Juan Ling
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Meng-Lin Xu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Jun Gu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Xiao-Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Wei-Lin Sha
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Tian Tian
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Xue-Hui Bai
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Nan Li
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, China
| | - Chang-Yu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, China
| | - Ouyang Chen
- Center for Translational Pain Medicine, Departments of Anesthesiology, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ling-Jie Ma
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Zhi-Jun Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Yi-Bin Qin
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Meixuan Zhu
- Department of Psychology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hong-Jie Yuan
- Department of Pain Management, Nantong Hospital of Traditional Chinese Medicine, Jiangsu 226001, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Departments of Anesthesiology, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| |
Collapse
|
4
|
Shen HC, Lee WJ, Sun CY, Yu WK, Chen WC, Hsiao FY, Yang KY, Chen LK. Follistatin-respiratory connection predicting all-cause mortality among community-dwelling middle-to-old age individuals: Results from the I-Lan Longitudinal Study. J Nutr Health Aging 2024; 28:100285. [PMID: 38861881 DOI: 10.1016/j.jnha.2024.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVES The link between aging and pulmonary function decline is well-established, but the underlying mechanisms have yet to be fully revealed. Serum follistatin, a myokine implicated in muscle degeneration, may play a role in age-related pulmonary changes. This study aims to investigate the relationship between serum follistatin levels and pulmonary function decline in community-dwelling older adults, and evaluate their combined association with all-cause mortality. RESEARCH DESIGN AND METHODS This longitudinal cohort study utilized data from 751 participants aged ≥50 years in the I-Lan Longitudinal Aging Study between 2018-2019. Serum follistatin levels, spirometry results, demographic and clinical data were retrieved. Participants were stratified based on their follistatin levels. Survival curves and group comparisons based on follistatin levels and decline in peak expiratory flow (PEF) using Kaplan-Meier analysis and log-rank tests. Multivariate Cox proportional hazards models were further used to identify independent predictors of all-cause mortality during the 52-month follow-up. RESULTS Elevated follistatin levels significantly correlated with worse pulmonary function, particularly decreased PEF (p = 0.030). Kaplan-Meier analysis revealed the combination of elevated follistatin levels and decreased PEF was associated with increased risk of all-cause mortality (Log-rank p = 0.023). Cox proportional hazards models further identified that concurrent presence of higher follistatin levels and decreased PEF predicted higher risk of all-cause mortality (adjusted HR 3.58, 95% CI: 1.22-10.53, p = 0.020). CONCLUSION Higher serum follistatin levels correlate with decreased pulmonary function, specifically PEF decline, in community-dwelling older adults. Furthermore, the coexistence of elevated follistatin levels and decreased PEF was associated with risk of all-cause mortality. Follistatin may serve as a biomarker for pulmonary aging and related adverse outcomes.
Collapse
Affiliation(s)
- Hsiao-Chin Shen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Evidence-based Medicine, Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Ju Lee
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Family Medicine, Taipei Veterans General Hospital Yuanshan Branch, Yilan, Taiwan
| | - Chuan-Yen Sun
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Kuang Yu
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chih Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fei-Yuan Hsiao
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan; School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuang-Yao Yang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Liang-Kung Chen
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan; Taipei Municipal Gan-Dau Hospital (Managed by Taipei Veterans General Hospital), Taipei, Taiwan
| |
Collapse
|
5
|
Elsheikh AA, Shalaby AM, Alabiad MA, Abd-Almotaleb NA, Alorini M, Alnasser SM, Elhasadi I, El-Nagdy SA. Trigonelline Chloride Ameliorated Triphenyltin-Induced Testicular Autophagy, Inflammation, and Apoptosis: Role of Recovery. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2024; 30:133-150. [PMID: 38156731 DOI: 10.1093/micmic/ozad137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
Triphenyltin chloride (TPT-Cl) is an organometallic organotin. This study aimed to investigate the role of trigonelline (TG) along with the impact of TPT withdrawal on the testicular toxicity induced by TPT-Cl. Thirty-six adult male albino rats were divided into control, TG (40 mg/kg/day), TPT-Cl (0.5 mg/kg/day), TG + TPT-Cl, and recovery groups. Animals were daily gavaged for 12 weeks. Both TG and TPT-Cl withdrawal improved TPT-Cl-induced testicular toxicity features involving testis and relative testis weight reduction, luteinizing hormone, follicular stimulating hormone, and sex hormone-binding globulin elevation, reduction of inhibin B, free testosterone levels, and sperm count reduction with increased abnormal sperm forms. Moreover, both TG and TPT-Cl withdrawal reduced inflammatory activin A, follistatin, tumor necrosis factor α, interleukin-1β, and proapoptotic Bax and elevated antiapoptotic Bcl2 in testicular tissues mediated by TPT-Cl. TG and TPT-Cl withdrawal restored the excessive autophagy triggered by TPT-Cl via elevation of mTOR, AKT, PI3K, and P62/SQSTM1 and reduction of AMPK, ULK1, Beclin1, and LC3 mRNA gene expressions and regained the deteriorated testicular structure. In conclusion, TG and TPT-Cl withdrawal had an ameliorative role in partially reversing TPT-Cl-induced testicular toxicity. However, the findings indicated that the use of TG as an adjunctive factor is more favorable than TPT-Cl withdrawal, suggesting the capability of the testis for partial self-improvement.
Collapse
Affiliation(s)
- Arwa A Elsheikh
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amany Mohamed Shalaby
- Histology and Cell Biology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Mohamed Ali Alabiad
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Noha Ali Abd-Almotaleb
- Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mohammed Alorini
- Department of Basic Medical Sciences, Unaizah College of Medicine and Medical Sciences, Qassim University, Unaizah 51911, Saudi Arabia
| | - Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51911, Saudi Arabia
| | - Ibtesam Elhasadi
- Department of Pathology, Faculty of Medicine, University of Benghazi, Benghazi, Libya
| | - Samah A El-Nagdy
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
6
|
Tobolski D, Zwierzchowski G, Lukasik K, Skarżyński DJ, Pascottini OB, Opsomer G, Barański W. Progesterone-independent endometrial mRNA expression in dairy cows with clinical or subclinical endometritis. Theriogenology 2024; 216:146-154. [PMID: 38183931 DOI: 10.1016/j.theriogenology.2023.12.031] [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: 10/13/2023] [Accepted: 12/27/2023] [Indexed: 01/08/2024]
Abstract
Up to 50 % of dairy cows fail to resolve uterine involution and develop chronic clinical (CE) or subclinical endometritis (SE) 21 days after calving. Clinical endometritis is associated with purulent discharge, while SE is not associated with overt clinical signs. Along with numerous knowledge gaps related to its pathogenesis, SE does not allow for a straightforward and effective therapy. Therefore, it is crucial to unravel differences in the expression of genes among healthy, CE, and SE cows. This might contribute to the discovery of new drug candidates and, in consequence, a potentially effective treatment. In the present study, cows between 21 and 28 days postpartum (PP) were examined using vaginoscopy for the presence of vaginal discharge and endometrial cytology for the determination of the endometrial polymorphonuclear cell (PMN) percentage. Next, an endometrial biopsy sample was taken to investigate the expression of 13 selected candidate genes by qPCR. Uterine health status was assigned to healthy (absence of abnormal vaginal discharge and ≤5 % PMN, n = 13), SE (absence of abnormal vaginal discharge and >5 % PMN, n = 30), and CE (mucopurulent or purulent vaginal discharge and >5 % PMN, n = 9). At the same time, a blood sample was collected to assess serum progesterone concentration and to categorize cows as low (≤1 ng/mL) or high (>1 ng/mL) in progesterone. High expression of IL1B, IL6, IL17A, CXCL8, PTGES, PTGS1, PTGS2, and INHBA genes and low expression of FST was noted in the endometrium of CE compared to healthy cows. Increased endometrial INHBA expression was observed in both SE and CE compared to healthy cows. Interestingly, greater expression of PTGES and PRXL2B genes and lower expression of PTGS2 were characteristic of SE versus CE or healthy. Among cows with no overt clinical symptoms of uterine disease (healthy and SE), the endometrial expression of IL1 B, CXCL8, and PTGES was greater in cows with high versus low serum progesterone. Several genes were differentially expressed among healthy, SE, and CE cows indicating different pathways for the development of different uterine diseases. In conclusion, we found progesterone-independent SE markers, which suggests that low endometrial PTGS2 expression may be indicative of an inadequate immune response and thus contribute to the pathogenesis of SE.
Collapse
Affiliation(s)
- Dawid Tobolski
- Department of Internal Diseases with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Oczapowskiego 14, Olsztyn, Poland.
| | - Grzegorz Zwierzchowski
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, 1a Oczapowskiego Str., Olsztyn, 10-719, Poland
| | - Karolina Lukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10 -748, Olsztyn, Poland
| | - Dariusz Jan Skarżyński
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10 -748, Olsztyn, Poland
| | - Osvaldo Bogado Pascottini
- Department of Internal Medicine, Reproduction and Population Medicine at the Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Geert Opsomer
- Department of Internal Medicine, Reproduction and Population Medicine at the Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wojciech Barański
- Department of Animal Reproduction with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Oczapowskiego 14, Olsztyn, Poland
| |
Collapse
|
7
|
Zeplin PH. Activin A and Follistatin Serum Concentrations in Breast Augmentation Patients. Plast Surg (Oakv) 2023; 31:377-382. [PMID: 37915344 PMCID: PMC10617457 DOI: 10.1177/22925503211051120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/17/2021] [Accepted: 08/28/2021] [Indexed: 11/03/2023] Open
Abstract
Background: Capsular contracture is caused by an excessive fibrotic reaction similar as observed in other progressive fibrotic disorders. For their pathogenesis, several studies confirmed the importance of activins and follistatin. The aim of this study was to determine and analyze serum levels of Activin A and follistatin in patients with capsular contracture after aesthetic breast augmentation. Methods: The study included 361 female patients who underwent primary aesthetic breast augmentation, came for control examination after breast augmentation or for revision operation because of capsular contracture. Blood samples were taken and using a specific ELISA to determine the serum concentration levels of Activin A and Follistatin. Results: Ninety-six patients (n = 96), who developed a capsular contracture Baker ≥°III and underwent revision surgery were collected (capsular fibrosis group). One-hundred and fourteen patients (n = 114) were asymptomatic for capsular fibrosis Baker ≥°III after primary breast augmentation and 33 (n = 33) of them had developed no capsular fibrosis after more than 10 years (long-term group). For control group, blood samples were taken from 167 patients (n = 167) before primary aesthetic breast augmentation. Serum Activin A levels were significantly higher in the long-term Group compared with those in the capsular fibrosis- and the control groups. Follistatin levels were significantly lower in the capsular fibrosis group compared to the control- and the long-term groups. A small amount of control group patients (n = 16) developed a capsular fibrosis within 2 years after primary breast augmentation with significant lower follistatin levels. Retrospectively, they showed significantly lower serum follistatin levels than the control group even before the onset of capsular contracture. Conclusions: Capsular fibrosis has no effect on Activin A serum levels. In contrast, follistatin serum levels are lower in patients with capsular fibrosis. These results show that besides many other factors, a dysregulation of the Activin-follistatin axis may have importance on the pathogenesis of capsular contracture.
Collapse
Affiliation(s)
- Philip H. Zeplin
- Schlosspark Klinik Ludwigsburg, Privatklinik für Plastische und Ästhetische Chirurgie, Ludwigsburg, Germany
| |
Collapse
|
8
|
Xie Z, Zhou G, Zhang M, Han J, Wang Y, Li X, Wu Q, Li M, Zhang S. Recent developments on BMPs and their antagonists in inflammatory bowel diseases. Cell Death Discov 2023; 9:210. [PMID: 37391444 DOI: 10.1038/s41420-023-01520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis, and Crohn's disease, are intestinal disorders characterized by chronic relapsing inflammation. A large proportion of patients with IBD will progress to develop colitis-associated colorectal cancer due to the chronic intestinal inflammation. Biologic agents that target tumour necrosis factor-α, integrin α4β7, and interleukin (IL)12/23p40 have been more successful than conventional therapies in treating IBD. However, drug intolerance and loss of response are serious drawbacks of current biologics, necessitating the development of novel drugs that target specific pathways in IBD pathogenesis. One promising group of candidate molecules are bone morphogenetic proteins (BMPs), members of the TGF-β family involved in regulating morphogenesis, homeostasis, stemness, and inflammatory responses in the gastrointestinal tract. Also worth examining are BMP antagonists, major regulators of these proteins. Evidence has shown that BMPs (especially BMP4/6/7) and BMP antagonists (especially Gremlin1 and follistatin-like protein 1) play essential roles in IBD pathogenesis. In this review, we provide an updated overview on the involvement of BMPs and BMP antagonists in IBD pathogenesis and in regulating the fate of intestinal stem cells. We also described the expression patterns of BMPs and BMP antagonists along the intestinal crypt-villus axis. Lastly, we synthesized available research on negative regulators of BMP signalling. This review summarizes recent developments on BMPs and BMP antagonists in IBD pathogenesis, which provides novel insights into future therapeutic strategies.
Collapse
Affiliation(s)
- Zhuo Xie
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Gaoshi Zhou
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Mudan Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jing Han
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Ying Wang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiaoling Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Qirui Wu
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Manying Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China.
| |
Collapse
|
9
|
Hatamzade Esfahani N, Day AS. The Role of TGF-β, Activin and Follistatin in Inflammatory Bowel Disease. GASTROINTESTINAL DISORDERS 2023; 5:167-186. [DOI: 10.3390/gidisord5020015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory condition predominantly affecting the gastrointestinal (GI) tract. An increasing prevalence of IBD has been observed globally. The pathogenesis of IBD includes a complex interplay between the intestinal microbiome, diet, genetic factors and immune responses. The consequent imbalance of inflammatory mediators ultimately leads to intestinal mucosal damage and defective repair. Growth factors, given their specific roles in maintaining the homeostasis and integrity of the intestinal epithelium, are of particular interest in the setting of IBD. Furthermore, direct targeting of growth factor signalling pathways involved in the regeneration of the damaged epithelium and the regulation of inflammation could be considered as therapeutic options for individuals with IBD. Several members of the transforming growth factor (TGF)-β superfamily, particularly TGF-β, activin and follistatin, are key candidates as they exhibit various roles in inflammatory processes and contribute to maintenance and homeostasis in the GI tract. This article aimed firstly to review the events involved in the pathogenesis of IBD with particular emphasis on TGF-β, activin and follistatin and secondly to outline the potential role of therapeutic manipulation of these pathways.
Collapse
Affiliation(s)
| | - Andrew S. Day
- Paediatric Department, University of Otago Christchurch, Christchurch 8140, New Zealand
| |
Collapse
|
10
|
Ameliorating Effects of Vitamin K2 on Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Int J Mol Sci 2023; 24:ijms24032986. [PMID: 36769323 PMCID: PMC9917520 DOI: 10.3390/ijms24032986] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic recurrent inflammatory illness of the gastrointestinal system. The purpose of this study was to explore the alleviating effect of vitamin K2 (VK2) on UC, as well as its mechanism. C57BL/6J mice were given 3% DSS for seven days to establish UC, and they then received VK2 (15, 30, or 60 mg/kg·bw) and 5-aminosalicylic acid (100 mg/kg·bw) for two weeks. We recorded the clinical signs, body weights, colon lengths, and histological changes during the experiment. We detected the inflammatory factor expressions using enzyme-linked immunosorbent assay (ELISA) kits, and we detected the tight junction proteins using Western blotting. We analyzed the intestinal microbiota alterations and short-chain fatty acids (SCFAs) using 16S rRNA sequencing and targeted metabolomics. According to the results, VK2 restored the colon lengths, improved the colonic histopathology, reduced the levels of proinflammatory cytokines (such as IL-1β, TNF-α, and IL-6), and boosted the level of the immunosuppressive cytokine IL-10 in the colon tissues of the colitis mice. Moreover, VK2 promoted the expression of mucin and tight junction proteins (such as occludin and zonula occludens-1) in order to preserve the intestinal mucosal barrier function and prevent UC in mice. Additionally, after the VK2 intervention, the SCFAs and SCFA-producing genera, such as Eubacterium_ruminantium_group and Faecalibaculum, were elevated in the colon. In conclusion, VK2 alleviated the DSS-induced colitis in the mice, perhaps by boosting the dominant intestinal microflora, such as Faecalibaculum, by reducing intestinal microflora dysbiosis, and by modulating the expression of SCFAs, inflammatory factors, and intestinal barrier proteins.
Collapse
|
11
|
Yu TY, Feng YM, Kong WS, Li SN, Sun XJ, Zhou G, Xie RF, Zhou X. Gallic acid ameliorates dextran sulfate sodium-induced ulcerative colitis in mice via inhibiting NLRP3 inflammasome. Front Pharmacol 2023; 14:1095721. [PMID: 36762118 PMCID: PMC9905138 DOI: 10.3389/fphar.2023.1095721] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Background: Ulcerative colitis (UC) is a chronic recurrent inflammatory bowel disease (IBD). The conventional drugs for UC may induce severe side effects. Herbal medicine is considered as a complementary and alternative choice for UC. Purpose: This study aims to estimate the effect of natural polyphenol gallic acid (GA) on the NLRP3 inflammasome with dextran sulfate sodium (DSS)-induced colitis in mice. Study design: The body weights and symptoms of BALB/c mice were recorded. Histological evaluation, ELISA, q-PCR, immunohistochemistry, and western blotting were carried out to observe the morphology, cytokine contents, mRNA expressions, and protein expressions, respectively. Lipopolysaccharide (LPS)-induced RAW264.7 macrophage was used to probe GA's effect on relative protein expression. Results: GA attenuated weight loss (p < 0.05), relieved symptoms, and ameliorated colonic morphological injury (p < 0.05) in mice with colitis induced by DSS. GA also lowered the contents of TNF-α, IL-1β, IL-18, IL-33, and IFN-γ in the serum and colon of mice, which were elevated by DSS, downregulated protein, and mRNA expressions of the NLRP3 pathway in the colon tissue. Furthermore, GA downregulated the expressions of NLRP3 (p < 0.05), iNOS (p < 0.01), COX2 (p < 0.01), and P-p65 (p < 0.05), and suppressed NO release (p < 0.001) in LPS-induced RAW264.7 cells. Conclusion: GA ameliorated DSS-induced UC in mice via inhibiting the NLRP3 inflammasome. These findings furnish evidence for the anti-inflammatory effect of herbal medicines containing GA on UC.
Collapse
Affiliation(s)
- Tian-Yuan Yu
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yi-Ming Feng
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei-Song Kong
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shan-Ni Li
- Shanghai Nanyang Model Private High School, Shanghai, China
| | - Xue-Jiao Sun
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Fengdu County People’s Hospital of Chongqing, Chongqing, China
| | - Gui Zhou
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui-Fang Xie
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Zhou
- Department of Pharmacy, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Xin Zhou,
| |
Collapse
|
12
|
Ariki S, Ozaka S, Sachi N, Chalalai T, Soga Y, Fukuda C, Kagoshima Y, Ekronarongchai S, Mizukami K, Kamiyama N, Murakami K, Kobayashi T. GM-CSF-producing CCR2 + CCR6 + Th17 cells are pathogenic in dextran sodium sulfate-induced colitis model in mice. Genes Cells 2023; 28:267-276. [PMID: 36641236 DOI: 10.1111/gtc.13008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Although excessive immune responses by Th17 cells, a helper T cell subset, are implicated in the pathogenesis of inflammatory bowel disease (IBD), the mechanism by which its localization in an inflamed colon is regulated remains unclear. Chemokines and their receptors are involved in the pathogenesis of IBD, however, the relative significance of each receptor on Th17 cells remains unknown. We generated C-C motif chemokine receptor 2 (CCR2) knockout (KO) and CCR6 KO mice in the syngeneic background using the CRISPR/Cas9 system and found that the phenotypes of experimental colitis worsened in both mutant mice. Surprisingly, the phenotype of colitis in CCR2/CCR6-double knockout (CCR2/6 DKO) mice was opposite to that of the single-deficient mice, with significantly milder experimental colitis (p < .05). The same was true for the symptoms in CCR6 KO mice, but not in wild type mice treated with a CCR2 inhibitor, propagermanium. Colonic CCR2+ CCR6+ Th17 cells produced a potentially pathogenic cytokine GM-CSF whose levels in the gut were significantly reduced in CCR2/6 DKO mice (p < .05). These results suggest that GM-CSF-producing CCR2+ CCR6+ Th17 cells are pathogenic and are attracted to the inflamed colon by either CCR2 or CCR6 gradient, which subsequently exacerbates experimental colitis in mice.
Collapse
Affiliation(s)
- Shimpei Ariki
- Department of Infectious Disease Control, Oita University, Oita, Japan.,Department of Gastroenterology, Oita University, Oita, Japan
| | - Sotaro Ozaka
- Department of Infectious Disease Control, Oita University, Oita, Japan.,Department of Gastroenterology, Oita University, Oita, Japan
| | - Nozomi Sachi
- Department of Infectious Disease Control, Oita University, Oita, Japan
| | | | - Yasuhiro Soga
- Department of Infectious Disease Control, Oita University, Oita, Japan
| | - Chiaki Fukuda
- Department of Infectious Disease Control, Oita University, Oita, Japan
| | - Yomei Kagoshima
- Department of Infectious Disease Control, Oita University, Oita, Japan.,Department of Gastroenterology, Oita University, Oita, Japan
| | | | - Kazuhiro Mizukami
- Department of Gastroenterology, Oita University, Oita, Japan.,Hospital Clinical Training Institute for Interns, Faculty of Medicine, Oita University, Oita, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Oita University, Oita, Japan
| | | | - Takashi Kobayashi
- Department of Infectious Disease Control, Oita University, Oita, Japan.,Research Center for GLOBAL and LOCAL Infectious Diseases, Oita, Japan
| |
Collapse
|
13
|
Ozaka S, Sonoda A, Ariki S, Minata M, Kamiyama N, Hidano S, Sachi N, Ito K, Kudo Y, Dewayani A, Chalalai T, Ozaki T, Soga Y, Fukuda C, Mizukami K, Ishizawa S, Nishiyama M, Fujitsuka N, Mogami S, Kubota K, Murakami K, Kobayashi T. Saireito, a Japanese herbal medicine, alleviates leaky gut associated with antibiotic-induced dysbiosis in mice. PLoS One 2022; 17:e0269698. [PMID: 35704618 PMCID: PMC9200308 DOI: 10.1371/journal.pone.0269698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Antibiotics disrupt normal gut microbiota and cause dysbiosis, leading to a reduction in intestinal epithelial barrier function. Disruption of the intestinal epithelial barrier, which is known as “leaky gut”, results in increased intestinal permeability and contributes to the development or exacerbation of gastrointestinal diseases such as inflammatory bowel disease and irritable bowel syndrome. We have previously reported on a murine model of intestinal epithelial barrier dysfunction associated with dysbiosis induced by the administration of ampicillin and vancomycin. Saireito, a traditional Japanese herbal medicine, is often used to treat autoimmune disorders including ulcerative colitis; the possible mechanism of action and its efficacy, however, remains unclear. In this study, we examined the efficacy of Saireito in our animal model for leaky gut associated with dysbiosis. C57BL/6 mice were fed a Saireito diet for the entirety of the protocol (day1-28). To induce colitis, ampicillin and vancomycin were administered in drinking water for the last seven consecutive days (day22-28). As previously demonstrated, treatment with antibiotics caused fecal occult bleeding, cecum enlargement with black discoloration, colon inflammation with epithelial cell apoptosis, and upregulation of pro-inflammatory cytokines. Oral administration of Saireito significantly improved antibiotics-induced fecal occult bleeding and cecum enlargement by suppressing inflammation in the colon. Furthermore, Saireito treatment ensured the integrity of the intestinal epithelial barrier by suppressing apoptosis and inducing cell adhesion proteins including ZO-1, occludin, and E-cadherin in intestinal epithelial cells, which in turn decreased intestinal epithelial permeability. Moreover, the reduced microbial diversity seen in the gut of mice treated with antibiotics was remarkably improved with the administration of Saireito. In addition, Saireito altered the composition of gut microbiota in these mice. These results suggest that Saireito alleviates leaky gut caused by antibiotic-induced dysbiosis. Our findings provide a potentially new therapeutic strategy for antibiotic-related gastrointestinal disorders.
Collapse
Affiliation(s)
- Sotaro Ozaka
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Akira Sonoda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Shimpei Ariki
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Mizuki Minata
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Shinya Hidano
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Nozomi Sachi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Kanako Ito
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Yoko Kudo
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Astri Dewayani
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Thanyakorn Chalalai
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Takashi Ozaki
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Yasuhiro Soga
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Chiaki Fukuda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Kazuhiro Mizukami
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Shiori Ishizawa
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Mitsue Nishiyama
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Naoki Fujitsuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Sachiko Mogami
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Kunitsugu Kubota
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Oita, Japan
- * E-mail:
| |
Collapse
|
14
|
Otake-Kasamoto Y, Kayama H, Kishikawa T, Shinzaki S, Tashiro T, Amano T, Tani M, Yoshihara T, Li B, Tani H, Liu L, Hayashi A, Okuzaki D, Motooka D, Nakamura S, Okada Y, Iijima H, Takeda K, Takehara T. Lysophosphatidylserines derived from microbiota in Crohn’s disease elicit pathological Th1 response. J Exp Med 2022; 219:213240. [PMID: 35608941 PMCID: PMC9134096 DOI: 10.1084/jem.20211291] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 03/17/2022] [Accepted: 05/02/2022] [Indexed: 12/31/2022] Open
Abstract
Microbiota alteration and IFN-γ–producing CD4+ T cell overactivation are implicated in Crohn’s disease (CD) pathogenesis. However, it remains unclear how dysbiosis enhances Th1 responses, leading to intestinal inflammation. Here, we identified key metabolites derived from dysbiotic microbiota that induce enhanced Th1 responses and exaggerate colitis in mouse models. Patients with CD showed elevated lysophosphatidylserine (LysoPS) concentration in their feces, accompanied by a higher relative abundance of microbiota possessing a gene encoding the phospholipid-hydrolyzing enzyme phospholipase A. LysoPS induced metabolic reprogramming, thereby eliciting aberrant effector responses in both human and mouse IFN-γ–producing CD4+ T cells. Administration of LysoPS into two mouse colitis models promoted large intestinal inflammation. LysoPS-induced aggravation of colitis was impaired in mice lacking P2ry10 and P2ry10b, and their CD4+ T cells were hyporesponsive to LysoPS. Thus, our findings elaborate on the mechanism by which metabolites elevated in patients with CD harboring dysbiotic microbiota promote Th1-mediated intestinal pathology.
Collapse
Affiliation(s)
- Yuriko Otake-Kasamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
| | - Toshihiro Kishikawa
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Otorhinolaryngology—Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Taku Tashiro
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takahiro Amano
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mizuki Tani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takeo Yoshihara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Bo Li
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Haruka Tani
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Li Liu
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akio Hayashi
- Discovery Technology Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Daisuke Okuzaki
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Daisuke Motooka
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Shota Nakamura
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Yukinori Okada
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
15
|
Laudisi F, Stolfi C, Bevivino G, Maresca C, Franzè E, Troncone E, Lolli E, Marafini I, Pietrucci D, Teofani A, Di Grazia A, Di Fusco D, Colantoni A, Ortenzi A, Desideri A, Monteleone I, Monteleone G. GATA6 Deficiency Leads to Epithelial Barrier Dysfunction and Enhances Susceptibility to Gut Inflammation. J Crohns Colitis 2022; 16:301-311. [PMID: 34374415 DOI: 10.1093/ecco-jcc/jjab145] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIMS Intestinal barrier dysfunction is a hallmark of inflammatory bowel diseases [IBD], but the mechanisms that lead to such a defect are not fully understood. This study was aimed at characterising the factors involved in the defective barrier function in IBD. METHODS Transcriptome analysis was performed on colon samples taken from healthy controls [CTR] and IBD patients. Expression of GATA-binding factor 6 [GATA6], a transcription factor involved in intestinal epithelial cell differentiation, was evaluated in colon samples taken from CTR and IBD patients by real-time polymerase chain reaction [PCR] and immunohistochemistry. Intestinal sections of wild-type and Gata6del mice, which exhibit a conditional Gata6 deletion in intestinal epithelial cells and which are either left untreated or receive subcutaneous indomethacin or rectal trinitrobenzene sulphonic acid, were stained with haematoxylin and eosin. In parallel, some Gata6del mice received antibiotics to deplete intestinal flora. Mucosal inflammatory cell infiltration and cytokine production were evaluated by flow cytometry and real-time PCR, respectively, and tight junction proteins were examined by immunofluorescence. Intestinal barrier integrity was assessed by fluorescein isothiocyanate [FITC]-dextran assay. RESULTS Multiple genes involved in cell commitment/proliferation and wound healing were differentially expressed in IBD compared with CTR. Among these, GATA6 was significantly decreased in the IBD epithelium compared with CTR. In mice, conditional deletion of GATA6 in the intestinal epithelium induced primarily epithelial damage, diminished zonula occludens-1 expression, and enhanced intestinal permeability, ultimately resulting in bacteria-driven local immune response and enhanced susceptibility to gut inflammation. CONCLUSIONS Reduced expression of GATA6 promotes intestinal barrier dysfunction, thus amplifying intestinal inflammatory pathology.
Collapse
Affiliation(s)
- Federica Laudisi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Division of Clinical Biochemistry and Clinical Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gerolamo Bevivino
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Maresca
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Franzè
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Edoardo Troncone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Elisabetta Lolli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Irene Marafini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Daniele Pietrucci
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.,Department for Innovation in Biological, Agro-Food and Forest Systems, DIBAF, University of Tuscia, Viterbo, Italy
| | - Adelaide Teofani
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Antonio Di Grazia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Davide Di Fusco
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alfredo Colantoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angela Ortenzi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Ivan Monteleone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | |
Collapse
|
16
|
Vlasov IN, Alieva AK, Novosadova EV, Arsenyeva EL, Rosinskaya AV, Partevian SA, Grivennikov IA, Shadrina MI. Transcriptome Analysis of Induced Pluripotent Stem Cells and Neuronal Progenitor Cells, Derived from Discordant Monozygotic Twins with Parkinson's Disease. Cells 2021; 10:3478. [PMID: 34943986 PMCID: PMC8700621 DOI: 10.3390/cells10123478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's Disease (PD) is a widespread severe neurodegenerative disease that is characterized by pronounced deficiency of the dopaminergic system and disruption of the function of other neuromodulator systems. Although heritable genetic factors contribute significantly to PD pathogenesis, only a small percentage of sporadic cases of PD can be explained using known genetic risk factors. Due to that, it could be inferred that changes in gene expression could be important for explaining a significant percentage of PD cases. One of the ways to investigate such changes, while minimizing the effect of genetic factors on experiment, are the study of PD discordant monozygotic twins. In the course of the analysis of transcriptome data obtained from IPSC and NPCs, 20 and 1906 differentially expressed genes were identified respectively. We have observed an overexpression of TNF in NPC cultures, derived from twin with PD. Through investigation of gene interactions and gene involvement in biological processes, we have arrived to a hypothesis that TNF could play a crucial role in PD-related changes occurring in NPC derived from twins with PD, and identified INHBA, WNT7A and DKK1 as possible downstream effectors of TNF.
Collapse
Affiliation(s)
- Ivan N. Vlasov
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| | - Anelya Kh. Alieva
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| | - Ekaterina V. Novosadova
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| | - Elena L. Arsenyeva
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| | - Anna V. Rosinskaya
- State Public Health Institution Primorsk Regional Clinical Hospital No. 1, 57 Aleutskaya St., 690091 Vladivostok, Russia;
| | - Suzanna A. Partevian
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| | - Igor A. Grivennikov
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| | - Maria I. Shadrina
- Institute of Molecular Genetics of National Research Centre, Kurchatov Institute, 2 Kurchatova Sq., 123182 Moscow, Russia; (A.K.A.); (E.V.N.); (E.L.A.); (S.A.P.); (I.A.G.); (M.I.S.)
| |
Collapse
|
17
|
Ozaka S, Sonoda A, Ariki S, Kamiyama N, Hidano S, Sachi N, Ito K, Kudo Y, Minata M, Saechue B, Dewayani A, Chalalai T, Soga Y, Takahashi Y, Fukuda C, Mizukami K, Okumura R, Kayama H, Murakami K, Takeda K, Kobayashi T. Protease inhibitory activity of secretory leukocyte protease inhibitor ameliorates murine experimental colitis by protecting the intestinal epithelial barrier. Genes Cells 2021; 26:807-822. [PMID: 34379860 DOI: 10.1111/gtc.12888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/04/2021] [Accepted: 08/07/2021] [Indexed: 12/13/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder in the intestine, and the dysfunction of intestinal epithelial barrier (IEB) may trigger the onset of IBD. Secretory leukocyte protease inhibitor (SLPI) is a serine protease inhibitor that has been implicated in the tissue-protective effect in the skin and lung. We found that SLPI was induced in lipopolysaccharides-treated colon carcinoma cell line and in the colon of dextran sulfate sodium (DSS)-treated mice. SLPI-deficient mice were administered DSS to induce colitis and sustained severe inflammation compared with wild-type mice. The colonic mucosa of SLPI-deficient mice showed more severe inflammation with neutrophil infiltration and higher levels of proinflammatory cytokines compared with control mice. Moreover, neutrophil elastase (NE) activity in SLPI-deficient mice was increased and IEB function was severely impaired in the colon, accompanied with the increased number of apoptotic cells. Importantly, we demonstrated that DSS-induced colitis was ameliorated by administration of protease inhibitor SSR69071 and recombinant SLPI. These results suggest that the protease inhibitory activity of SLPI protects from colitis by preventing IEB dysfunction caused by excessive NE activity, which provides insight into the novel function of SLPI in the regulation of gut homeostasis and therapeutic approaches for IBD.
Collapse
Affiliation(s)
- Sotaro Ozaka
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan.,Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Akira Sonoda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan.,Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Shimpei Ariki
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan.,Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Shinya Hidano
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Nozomi Sachi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kanako Ito
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yoko Kudo
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Mizuki Minata
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Benjawan Saechue
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Astri Dewayani
- Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Thanyakorn Chalalai
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yasuhiro Soga
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yuya Takahashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Chiaki Fukuda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kazuhiro Mizukami
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan.,Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Ryu Okumura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
18
|
Franzè E, Monteleone I, Laudisi F, Rizzo A, Dinallo V, Di Fusco D, Colantoni A, Ortenzi A, Giuffrida P, Di Carlo S, Sica GS, Di Sabatino A, Monteleone G. Cadherin-11 Is a Regulator of Intestinal Fibrosis. J Crohns Colitis 2020; 14:406-417. [PMID: 31504344 DOI: 10.1093/ecco-jcc/jjz147] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Although the mechanisms underlying the formation of intestinal fibrostrictures in Crohn's disease [CD] are not fully understood, activation of fibroblasts and excessive collagen deposition are supposed to contribute to the development of such complications. Here, we investigated the role of cadherin-11 [CDH-11], a fibroblast-derived protein that induces collagen production in various organs, in intestinal fibrosis. METHODS CDH-11 expression was evaluated in inflammatory [I] and fibrostricturing [FS] CD mucosal samples, ulcerative colitis [UC] mucosal samples, and ileal and colonic control samples, by real-time polymerase chain reaction, western blotting, and immunohistochemistry. CDH-11 expression was evaluated in normal and in CD intestinal fibroblasts stimulated with inflammatory/fibrogenic cytokines. FS CD fibroblasts were cultured either with a specific CDH-11 antisense oligonucleotide [AS], or activating CDH-11 fusion protein and activation of RhoA/ROCK, and TGF-β pathways and collagen production were evaluated by western blotting. Finally, we assessed the susceptibility of CDH-11-knockout [KO] mice to colitis-induced intestinal fibrosis. RESULTS CDH-11 RNA and protein expression were increased in both CD and UC as compared with controls. In CD, the greater expression of CDH-11 was seen in FS samples. Stimulation of fibroblasts with TNF-α, interleukin [IL]-6, IFN-γ, IL-13, and IL-1β enhanced CDH-11 expression. Knockdown of CDH-11 in FS CD fibroblasts impaired RhoA/ROCK/TGF-β signalling and reduced collagen synthesis, whereas activation of CDH-11 increased collagen secretion. CDH-11 KO mice were largely protected from intestinal fibrosis. CONCLUSIONS Data show that CDH-11 expression is up-regulated in inflammatory bowel disease [IBD] and suggest a role for this protein in the control of intestinal fibrosis.
Collapse
Affiliation(s)
- Eleonora Franzè
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Ivan Monteleone
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Angelamaria Rizzo
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Vincenzo Dinallo
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Davide Di Fusco
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Alfredo Colantoni
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Angela Ortenzi
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| | - Paolo Giuffrida
- Department of Internal Medicine, San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Sara Di Carlo
- Department of Surgery, University 'TOR VERGATA' of Rome, Rome, Italy
| | - Giuseppe S Sica
- Department of Surgery, University 'TOR VERGATA' of Rome, Rome, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine, San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome 'TOR VERGATA', Rome, Italy
| |
Collapse
|
19
|
Takei Y, Takahashi S, Nakasatomi M, Sakairi T, Ikeuchi H, Kaneko Y, Hiromura K, Nojima Y, Maeshima A. Urinary Activin A is a novel biomarker reflecting renal inflammation and tubular damage in ANCA-associated vasculitis. PLoS One 2019; 14:e0223703. [PMID: 31613925 PMCID: PMC6793943 DOI: 10.1371/journal.pone.0223703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 09/26/2019] [Indexed: 11/18/2022] Open
Abstract
Activin A, a member of the transforming growth factor-beta superfamily, is a critical modulator of inflammation and plays a key role in controlling the cytokine cascade that drives the inflammatory response. However, the role of activin A in inflammatory kidney diseases remains unknown. To address this issue, we examined here whether activin A can be detected in the kidney and/or urine from patients with antineutrophil cytoplasmic antibody (ANCA) -associated vasculitis (AAV). Fifty-one patients who had been diagnosed with AAV and were treated in our department between November 2011 to March 2018 were included in this study. Forty-one patients had renal complications (renal AAV). Serum and urinary activin A levels were measured by enzyme-linked immunosorbent assay. Correlation of urinary activin A concentration with clinical parameters was analyzed. Urinary activin A was undetectable in healthy volunteers. In contrast, urinary activin A concentration was significantly increased in patients with renal AAV but not in those with non-renal AAV. Urinary activin A concentration decreased rapidly after immunosuppressive treatment. There was a significant correlation of urinary activin A level with urinary protein, L-FABP, and NAG. Histologic evaluation revealed that urinary activin A levels were significantly higher in patients with cellular crescentic glomeruli than in those lacking this damage. In situ hybridization demonstrated that the mRNA encoding the activin A βA subunit was undetectable in normal kidneys but accumulated in the proximal tubules and crescentic glomeruli of the kidneys of patients with renal AAV. Immunostaining showed that activin A protein also was present in the proximal tubules, crescentic glomeruli, and macrophages infiltrating into the interstitium in the kidneys of patients with renal AAV. These data suggested that urinary activin A concentration reflects renal inflammation and tubular damage in AAV and may be a useful biomarker for monitoring renal AAV.
Collapse
Affiliation(s)
- Yoshinori Takei
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shunsuke Takahashi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masao Nakasatomi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Toru Sakairi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoriaki Kaneko
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshihisa Nojima
- Department of Nephrology and Rheumatology, Japanese Red Cross Hospital, Maebashi, Japan
| | - Akito Maeshima
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Nephrology and Rheumatology, Japanese Red Cross Hospital, Maebashi, Japan
- * E-mail:
| |
Collapse
|
20
|
Roudebush C, Catala-Valentin A, Andl T, Le Bras GF, Andl CD. Activin A-mediated epithelial de-differentiation contributes to injury repair in an in vitro gastrointestinal reflux model. Cytokine 2019; 123:154782. [PMID: 31369967 DOI: 10.1016/j.cyto.2019.154782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023]
Abstract
Reflux esophagitis is a result of esophageal exposure to acid and bile during episodes of gastroesophageal reflux. Aside from chemical injury to the esophageal epithelium, it has been shown that acid and bile induce cytokine-mediated injury by stimulating the release of pro-inflammatory cytokines. During the repair and healing process following reflux injury, the squamous esophageal cells are replaced with a columnar epithelium causing Barrett's metaplasia, which predisposes patients to esophageal adenocarcinoma. We identified a novel player in gastroesophageal reflux injury, the TGFβ family member Activin A (ActA), which is a known regulator of inflammation and tissue repair. In this study, we show that in response to bile salt and acidified media (pH 4) exposure, emulating the milieu to which the distal esophagus is exposed during gastroesophageal reflux, long-term treated, tolerant esophageal keratinocytes exhibit increased ActA secretion and a pro-inflammatory cytokine signature. Furthermore, we noted increased motility and expression of the stem cell markers SOX9, LGR5 and DCLK1 supporting the notion that repair mechanisms were activated in the bile salt/acid-tolerant keratinocytes. Additionally, these experiments demonstrated that de-differentiation as characterized by the induction of YAP1, FOXO3 and KRT17 was altered by ActA/TGFβ signaling. Collectively, our results suggest a pivotal role for ActA in the inflammatory GERD environment by modulating esophageal tissue repair and de-differentiation.
Collapse
Affiliation(s)
- Cedric Roudebush
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Alma Catala-Valentin
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Gregoire F Le Bras
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Claudia D Andl
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States.
| |
Collapse
|
21
|
Diller M, Frommer K, Dankbar B, Tarner I, Hülser ML, Tsiklauri L, Hasseli R, Sauerbier M, Pap T, Rehart S, Müller-Ladner U, Neumann E. The activin-follistatin anti-inflammatory cycle is deregulated in synovial fibroblasts. Arthritis Res Ther 2019; 21:144. [PMID: 31182152 PMCID: PMC6558802 DOI: 10.1186/s13075-019-1926-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/26/2019] [Indexed: 12/25/2022] Open
Abstract
Background Activin A and follistatin exhibit immunomodulatory functions, thus affecting autoinflammatory processes as found in rheumatoid arthritis (RA). The impact of both proteins on the behavior of synovial fibroblasts (SF) in RA as well as in osteoarthritis (OA) is unknown. Methods Immunohistochemical analyses of synovial tissue for expression of activin A and follistatin were performed. The influence of RASF overexpressing activin A on cartilage invasion in a SCID mouse model was examined. RASF and OASF were stimulated with either IL-1β or TNFα in combination with or solely with activin A, activin AB, or follistatin. Protein secretion was measured by ELISA and mRNA expression by RT-PCR. Smad signaling was confirmed by western blot. Results In human RA synovial tissue, the number of activin A-positive cells as well as its extracellular presence was higher than in the OA synovium. Single cells within the tissue expressed follistatin in RA and OA synovial tissue. In the SCID mouse model, activin A overexpression reduced RASF invasion. In human RASF, activin A was induced by IL-1β and TNFα. Activin A slightly increased IL-6 release by unstimulated RASF, but decreased protein and mRNA levels of follistatin. Conclusion The observed decrease of cartilage invasion by RASF overexpressing activin A in the SCID mouse model appears to be mediated by an interaction between activin/follistatin and other local cells indirectly affecting RASF because activin A displayed certain pro-inflammatory effects on RASF. Activin A even inhibits production and release of follistatin in RASF and therefore prevents itself from being blocked by its inhibitory binding protein follistatin in the local inflammatory joint environment. Electronic supplementary material The online version of this article (10.1186/s13075-019-1926-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Magnus Diller
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Klaus Frommer
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Berno Dankbar
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Ingo Tarner
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Marie-Lisa Hülser
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Lali Tsiklauri
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Rebecca Hasseli
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Michael Sauerbier
- Department of Plastic, Hand and Reconstructive Surgery, BGU Frankfurt, Frankfurt, Germany
| | - Thomas Pap
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Stefan Rehart
- Department of Orthopaedics and Trauma Surgery, Agaplesion Markus Hospital, Frankfurt, Germany
| | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany
| | - Elena Neumann
- Department of Rheumatology and Clinical Immunology, Justus Liebig University Giessen, Campus Kerckhoff, Bad Nauheim, Benekestr: 2-8, 61231, Bad Nauheim, Germany.
| |
Collapse
|
22
|
Laudisi F, Di Fusco D, Dinallo V, Stolfi C, Di Grazia A, Marafini I, Colantoni A, Ortenzi A, Alteri C, Guerrieri F, Mavilio M, Ceccherini-Silberstein F, Federici M, MacDonald TT, Monteleone I, Monteleone G. The Food Additive Maltodextrin Promotes Endoplasmic Reticulum Stress-Driven Mucus Depletion and Exacerbates Intestinal Inflammation. Cell Mol Gastroenterol Hepatol 2018; 7:457-473. [PMID: 30765332 PMCID: PMC6369223 DOI: 10.1016/j.jcmgh.2018.09.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/27/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Food additives, such as emulsifiers, stabilizers, or bulking agents, are present in the Western diet and their consumption is increasing. However, little is known about their potential effects on intestinal homeostasis. In this study we examined the effect of some of these food additives on gut inflammation. METHODS Mice were given drinking water containing maltodextrin (MDX), propylene glycol, or animal gelatin, and then challenged with dextran sulfate sodium or indomethacin. In parallel, mice fed a MDX-enriched diet were given the endoplasmic reticulum (ER) stress inhibitor tauroursodeoxycholic acid (TUDCA). Transcriptomic analysis, real-time polymerase chain reaction, mucin-2 expression, phosphorylated p38 mitogen-activated protein (MAP) kinase quantification, and H&E staining was performed on colonic tissues. Mucosa-associated microbiota composition was characterized by 16S ribosomal RNA sequencing. For the in vitro experiments, murine intestinal crypts and the human mucus-secreting HT29-methotrexate treated cell line were stimulated with MDX in the presence or absence of TUDCA or a p38 MAP kinase inhibitor. RESULTS Diets enriched in MDX, but not propylene glycol or animal gelatin, exacerbated intestinal inflammation in both models. Analysis of the mechanisms underlying the detrimental effect of MDX showed up-regulation of inositol requiring protein 1β, a sensor of ER stress, in goblet cells, and a reduction of mucin-2 expression with no significant change in mucosa-associated microbiota. Stimulation of murine intestinal crypts and HT29-methotrexate treated cell line cells with MDX induced inositol requiring protein 1β via a p38 MAP kinase-dependent mechanism. Treatment of mice with TUDCA prevented mucin-2 depletion and attenuated colitis in MDX-fed mice. CONCLUSIONS MDX increases ER stress in gut epithelial cells with the downstream effect of reducing mucus production and enhancing colitis susceptibility.
Collapse
Affiliation(s)
- Federica Laudisi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Davide Di Fusco
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Vincenzo Dinallo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Antonio Di Grazia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Irene Marafini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alfredo Colantoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angela Ortenzi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Alteri
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Guerrieri
- Center for Life NanoScience at Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Maria Mavilio
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy,Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy
| | - Thomas Thornton MacDonald
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Whitechapel, London, United Kingdom
| | - Ivan Monteleone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy,Correspondence Address correspondence to: Giovanni Monteleone, MD, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy. fax: (39) 06-72596391.
| |
Collapse
|
23
|
Sedda S, Franzè E, Bevivino G, Di Giovangiulio M, Rizzo A, Colantoni A, Ortenzi A, Grasso E, Giannelli M, Sica GS, Fantini MC, Monteleone G. Reciprocal Regulation Between Smad7 and Sirt1 in the Gut. Front Immunol 2018; 9:1854. [PMID: 30147698 PMCID: PMC6097015 DOI: 10.3389/fimmu.2018.01854] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022] Open
Abstract
In inflammatory bowel disease (IBD) mucosa, there is over-expression of Smad7, an intracellular inhibitor of the suppressive cytokine transforming growth factor-β1, due to post-transcriptional mechanisms that enhance Smad7 acetylation status thus preventing ubiquitination-mediated proteosomal degradation of the protein. IBD-related inflammation is also marked by defective expression of Sirt1, a class III NAD+-dependent deacetylase, which promotes ubiquitination-mediated proteosomal degradation of various intracellular proteins and triggers anti-inflammatory signals. The aim of our study was to determine whether, in IBD, there is a reciprocal regulation between Smad7 and Sirt1. Smad7 and Sirt1 were examined in mucosal samples of IBD patients and normal controls by Western blotting and immunohistochemistry, and Sirt1 activity was assessed by a fluorimetric assay. To determine whether Smad7 is regulated by Sirt1, normal or IBD lamina propria mononuclear cells (LPMC) were cultured with either Sirt1 inhibitor (Ex527) or activator (Cay10591), respectively. To determine whether Smad7 controls Sirt1 expression, ex vivo organ cultures of IBD mucosal explants were treated with Smad7 sense or antisense oligonucleotide. Moreover, Sirt1 expression was evaluated in LPMC isolated from Smad7-transgenic mice given dextran sulfate sodium (DSS). Upregulation of Smad7 was seen in both the epithelial and lamina propria compartments of IBD patients and this associated with reduced expression and activity of Sirt1. Activation of Sirt1 in IBD LPMC with Cay10591 reduced acetylation and enhanced ubiquitination-driven proteasomal-mediated degradation of Smad7, while inhibition of Sirt1 activation in normal LPMC with Ex527 increased Smad7 expression. Knockdown of Smad7 in IBD mucosal explants enhanced Sirt1 expression, thus suggesting a negative effect of Smad7 on Sirt1 induction. Consistently, mucosal T cells of Smad7-transgenic mice contained reduced levels of Sirt1, a defect that was amplified by induction of DSS colitis. The data suggest the existence of a reciprocal regulatory mechanism between Smad7 and Sirt1, which could contribute to amplify inflammatory signals in the gut.
Collapse
Affiliation(s)
- Silvia Sedda
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Franzè
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Gerolamo Bevivino
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Angelamaria Rizzo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alfredo Colantoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angela Ortenzi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Enrico Grasso
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Mario Giannelli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe S Sica
- Department of Surgery, University of Rome Tor Vergata, Rome, Italy
| | | | | |
Collapse
|
24
|
Activin in acute pancreatitis: Potential risk-stratifying marker and novel therapeutic target. Sci Rep 2017; 7:12786. [PMID: 28986573 PMCID: PMC5630611 DOI: 10.1038/s41598-017-13000-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Acute Pancreatitis is a substantial health care challenge with increasing incidence. Patients who develop severe disease have considerable mortality. Currently, no reliable predictive marker to identify patients at risk for severe disease exists. Treatment is limited to rehydration and supporting care suggesting an urgent need to develop novel approaches to improve standard care. Activin is a critical modulator of inflammatory responses, but has not been assessed in pancreatitis. Here, we demonstrate that serum activin is elevated and strongly correlates with disease severity in two established murine models of acute pancreatitis induced by either cerulein or IL-12 + IL-18. Furthermore, in mice, inhibition of activin conveys survival benefits in pancreatitis. In addition, serum activin levels were measured from a retrospective clinical cohort of pancreatitis patients and high activin levels in patients at admission are predictive of worse outcomes, indicated by longer overall hospital and intensive care unit stays. Taken together, activin is a novel candidate as a clinical marker to identify those acute pancreatitis patients with severe disease who would benefit from aggressive treatment and activin may be a therapeutic target in severe acute pancreatitis.
Collapse
|
25
|
Kawai S, Iijima H, Shinzaki S, Hiyama S, Yamaguchi T, Araki M, Iwatani S, Shiraishi E, Mukai A, Inoue T, Hayashi Y, Tsujii M, Motooka D, Nakamura S, Iida T, Takehara T. Indigo Naturalis ameliorates murine dextran sodium sulfate-induced colitis via aryl hydrocarbon receptor activation. J Gastroenterol 2017; 52:904-919. [PMID: 27900483 DOI: 10.1007/s00535-016-1292-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Indigo Naturalis (IN) is used as a traditional herbal medicine for ulcerative colitis (UC). However, the mechanisms of action of IN have not been clarified. We aimed to evaluate the efficacy of IN for ameliorating colonic inflammation. We further investigated the mechanisms of action of IN. METHODS Colitis severity was assessed in dextran sodium sulfate-induced colitis and trinitrobenzene sulfonic acid-induced colitis models with or without the oral administration of IN or indigo, which is a known major component of IN. Colonic lamina propria (LP) mononuclear cells isolated from IN-treated mice were analyzed with quantitative reverse transcription polymerase chain reaction (qRT-PCR) and flow cytometry. LP and splenic mononuclear cells cultured in vitro with IN or indigo were also analyzed. The role of the candidate receptor for indigo, the aryl hydrocarbon receptor (AhR), was analyzed using Ahr-deficient mice. RESULTS Colitis severity was significantly ameliorated in the IN and indigo treatment groups compared with the control group. The mRNA expression levels of interleukin (Il)-10 and Il-22 in the LP lymphocytes were increased by IN treatment. The treatment of splenocytes with IN or indigo increased the expression of anti-inflammatory cytokines and resulted in the expansion of IL-10-producing CD4+ T cells and IL-22-producing CD3-RORγt+ cells, but not CD4+Foxp3+ regulatory T cells. The amelioration of colitis by IN or indigo was abrogated in Ahr-deficient mice, in association with diminished regulatory cytokine production. CONCLUSIONS IN and indigo ameliorated murine colitis through AhR signaling activation, suggesting that AhR could be a promising therapeutic target for UC.
Collapse
MESH Headings
- Animals
- CD3 Complex/metabolism
- CD4 Lymphocyte Count
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Colitis/chemically induced
- Colitis/drug therapy
- Colitis/pathology
- Dextran Sulfate
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Forkhead Transcription Factors/metabolism
- Gene Expression/drug effects
- Indigo Carmine/pharmacology
- Indigo Carmine/therapeutic use
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Interleukin-2 Receptor alpha Subunit/metabolism
- Interleukins/genetics
- Interleukins/metabolism
- Intestinal Mucosa/cytology
- Leukocytes, Mononuclear/metabolism
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- RNA, Messenger/metabolism
- Receptors, Aryl Hydrocarbon/deficiency
- Receptors, Aryl Hydrocarbon/drug effects
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Severity of Illness Index
- Spleen/cytology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/metabolism
- Trinitrobenzenesulfonic Acid
- Interleukin-22
Collapse
Affiliation(s)
- Shoichiro Kawai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Hiyama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Inflammatory Bowel Disease, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshio Yamaguchi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Manabu Araki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shuko Iwatani
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eri Shiraishi
- Department of Gastroenterology and Hepatology, Sumitomo Hospital, Osaka, Japan
| | - Akira Mukai
- Department of Gastroenterology and Hepatology, Sumitomo Hospital, Osaka, Japan
| | - Takahiro Inoue
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshito Hayashi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiko Tsujii
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Gastroenterology and Hepatology, Higashiosaka City General Hospital, Higashiosaka, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tetsuya Iida
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
26
|
Staudacher JJ, Bauer J, Jana A, Tian J, Carroll T, Mancinelli G, Özden Ö, Krett N, Guzman G, Kerr D, Grippo P, Jung B. Activin signaling is an essential component of the TGF-β induced pro-metastatic phenotype in colorectal cancer. Sci Rep 2017; 7:5569. [PMID: 28717230 PMCID: PMC5514149 DOI: 10.1038/s41598-017-05907-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/05/2017] [Indexed: 01/09/2023] Open
Abstract
Advanced colorectal cancer (CRC) remains a critical health care challenge worldwide. Various TGF-β superfamily members are important in colorectal cancer metastasis, but their signaling effects and predictive value have only been assessed in isolation. Here, we examine cross-regulation and combined functions of the two most prominent TGF-β superfamily members activin and TGF-β in advanced colorectal cancer. In two clinical cohorts we observed by immune-based assay that combined serum and tissue activin and TGF-β ligand levels predicts outcome in CRC patients and is superior to single ligand assessment. While TGF-β growth suppression is independent of activin, TGF-β treatment leads to increased activin secretion in colon cancer cells and TGF-β induced cellular migration is dependent on activin, indicating pathway cross-regulation and functional interaction in vitro. mRNA expression of activin and TGF-β pathway members were queried in silico using the TCGA data set. Coordinated ligand and receptor expression is common in solid tumors for activin and TGF-β pathway members. In conclusion, activin and TGF-β are strongly connected signaling pathways that are important in advanced CRC. Assessing activin and TGF-β signaling as a unit yields important insights applicable to future diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Jonas J Staudacher
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jessica Bauer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Arundhati Jana
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jun Tian
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Timothy Carroll
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Georgina Mancinelli
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Özkan Özden
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Nancy Krett
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - David Kerr
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Paul Grippo
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Barbara Jung
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
27
|
Follistatin and the Breast Implant Capsule. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2017; 5:e1258. [PMID: 28458972 PMCID: PMC5404443 DOI: 10.1097/gox.0000000000001258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 01/11/2017] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Breast capsular contracture remains an elusive problem faced by plastic surgeons and is the leading long-term complication after breast implantation. Follistatin (Fst) is a protein with known anti-inflammatory and antifibrotic properties and has the potential to limit the severity of diseases associated with inflammation and fibrosis such as capsular contracture. The aim of this study was to examine the effect of Fst288 on capsular fibrosis around silicone implants in a mouse model. Methods: BALB/c mice were implanted subcutaneously with untreated silicone implants (baseline control). In the experimental group, immediately after silicone implant insertion, the implant pocket received either a single injection of 1 µg Fst288 or normal saline (internal control). The animals were killed at 3, 5, 7, 14, 28, and 90 days after surgery, and serum, implants, and the surrounding tissue were removed for histological and immunohistochemical analyses. Results: Fst288 treatment resulted in significant decreases in capsule thickness at 28 days (P < 0.05) and 3 months (P < 0.001), decreased collagen production at 14 days (P < 0.05) and 3 months (P < 0.01), decreased angiogenesis at 3 months (P < 0.001), decreased α-smooth muscle actin levels at 3 months (P < 0.05), and a decrease in the number of CD45+ cells at days 5 (P < 0.05) and 7 (P < 0.01), respectively, when compared with control implants. Conclusions: A single injection of Fst288 at the time of silicone implant insertion into the mice results in a significant reduction in pericapsular inflammation and capsular fibrosis.
Collapse
|
28
|
Kadiombo AT, Maeshima A, Kayakabe K, Ikeuchi H, Sakairi T, Kaneko Y, Hiromura K, Nojima Y. Involvement of infiltrating macrophage-derived activin A in the progression of renal damage in MRL-lpr mice. Am J Physiol Renal Physiol 2017; 312:F297-F304. [DOI: 10.1152/ajprenal.00191.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 11/15/2016] [Accepted: 11/20/2016] [Indexed: 01/12/2023] Open
Abstract
Lupus nephritis is a life-threatening complication of systemic lupus erythematosus (SLE). Various growth factors, cytokines, and chemokines are implicated in the development of SLE. However, the pathophysiological processes involved in the development of lupus nephritis still remain unclear. In this study, we examined the involvement of activin A, a member of the transforming growth factor β (TGF-β) superfamily, in the progression of renal damage in lupus-prone MRL- lpr mice. Activin A was not expressed in the kidneys of control MRL-MpJ mice but was detectable in perivascular infiltrating cluster of differentiation 68 (CD68)-positive cells in the kidneys of MRL- lpr mice. Urinary activin A, which was also absent in MRL-MpJ mice, was detectable in MRL- lpr mice from 16 wk onward. Urinary activin A levels were significantly correlated with the number of perivascular inflammatory cell layers, the number of crescentic glomeruli, and the percentage of Elastica van Gieson (EVG)-positive fibrotic areas, but not with urinary protein levels or serum activin A. When activin action was blocked in vivo by the intraperitoneal administration of an activin antagonist, follistatin, the number of crescentic glomeruli, percentage of EVG-positive fibrotic areas, CD68-positive cell infiltration, and proteinuria were significantly reduced in a dose-dependent manner. These data suggest that infiltrating macrophage-derived activin A is involved in the progression of renal damage in MRL- lpr mice.
Collapse
Affiliation(s)
| | - Akito Maeshima
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Ken Kayakabe
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Hidekazu Ikeuchi
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Toru Sakairi
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Yoriaki Kaneko
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Keiju Hiromura
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Yoshihisa Nojima
- Department of Medicine and Clinical Science, Gunma University, Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
29
|
Jung B, Staudacher JJ, Beauchamp D. Transforming Growth Factor β Superfamily Signaling in Development of Colorectal Cancer. Gastroenterology 2017; 152:36-52. [PMID: 27773809 PMCID: PMC5550896 DOI: 10.1053/j.gastro.2016.10.015] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Transforming growth factor (TGF)-β cytokines signal via a complex network of pathways to regulate proliferation, differentiation, adhesion, migration, and other functions in many cell types. A high percentage of colorectal tumors contain mutations that disrupt TGF-β family member signaling. We review how TGF-β family member signaling is altered during development of colorectal cancer, models of study, interaction of pathways, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Barbara Jung
- University of Illinois at Chicago, Chicago, Illinois.
| | | | | |
Collapse
|
30
|
Refaat B, El-Shemi AG, Mohamed AM, Kensara OA, Ahmad J, Idris S. Activins and their related proteins in colon carcinogenesis: insights from early and advanced azoxymethane rat models of colon cancer. BMC Cancer 2016; 16:879. [PMID: 27835986 PMCID: PMC5106801 DOI: 10.1186/s12885-016-2914-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/27/2016] [Indexed: 02/07/2023] Open
Abstract
Background Activin-A may exert pro- or anti-tumorigenic activities depending on cellular context. However, little is known about its role, or the other mature activin proteins, in colorectal carcinoma (CRC). This study measured the expression of activin βA- & βB-subunits, activin type IIA & IIB receptors, smads 2/3/4/6/7 and follistatin in CRC induced by azoxymethane (AOM) in rats. The results were compared with controls and disseminated according to the characteristics of histopathological lesions. Methods Eighty male Wistar rats were allocated into 20 controls and the remaining were equally divided between short ‘S-AOM’ (15 weeks) and long ‘L-AOM’ (35 weeks) groups following injecting AOM for 2 weeks. Subsequent to gross and histopathological examinations and digital image analysis, the expression of all molecules was measured by immunohistochemistry and quantitative RT-PCR. Activin-A, activin-B, activin-AB and follistatin were measured by ELISA in serum and colon tissue homogenates. Results Colonic pre-neoplastic and cancerous lesions were identified in both AOM groups and their numbers and sizes were significantly (P < 0.05) greater in the L-AOM group. All the molecules were expressed in normal colonic epithelial cells. There was a significantly (P < 0.05) greater expression of βA-subunit, IIB receptor and follistatin in both pre-neoplastic and cancerous tissues. Oppositely, a significant (P < 0.05) decrease in the remaining molecules was detected in both AOM groups. Metastatic lesions were only observed within the L-AOM group and were associated with the most significant alterations of all molecules. Significantly higher concentrations of activin-A and follistatin and lower activin-AB were also detected in both groups of AOM. Tissue and serum concentrations of activin-A and follistatin correlated positively, while tissue activin-AB inversely, and significantly with the numbers and sizes of colonic lesions. Conclusions Normal rat colon epithelial cells are capable of synthesising, controlling as well as responding to activins in a paracrine/autocrine manner. Colonic activin systems are pathologically altered during tumorigenesis and appear to be time and lesion-dependent. Activins could also be potential sensitive markers and/or molecular targets for the diagnosis and/or treatment of CRC. Further studies are required to illustrate the clinical value of activins and their related proteins in colon cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2914-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bassem Refaat
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-Abdeyah, PO Box 7607, Makkah, Kingdom of Saudi Arabia.
| | - Adel Galal El-Shemi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-Abdeyah, PO Box 7607, Makkah, Kingdom of Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amr Mohamed Mohamed
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-Abdeyah, PO Box 7607, Makkah, Kingdom of Saudi Arabia.,Clinical Laboratory Diagnosis, Department of Animal Medicine, Faculty of Veterinary Medicine, Assiut University, 71526, Assiut, Egypt
| | - Osama Adnan Kensara
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-Abdeyah, PO Box 7607, Makkah, Kingdom of Saudi Arabia
| | - Jawwad Ahmad
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-Abdeyah, PO Box 7607, Makkah, Kingdom of Saudi Arabia
| | - Shakir Idris
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al-Abdeyah, PO Box 7607, Makkah, Kingdom of Saudi Arabia
| |
Collapse
|
31
|
Namwanje M, Brown CW. Activins and Inhibins: Roles in Development, Physiology, and Disease. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a021881. [PMID: 27328872 DOI: 10.1101/cshperspect.a021881] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Since their original discovery as regulators of follicle-stimulating hormone (FSH) secretion and erythropoiesis, the TGF-β family members activin and inhibin have been shown to participate in a variety of biological processes, from the earliest stages of embryonic development to highly specialized functions in terminally differentiated cells and tissues. Herein, we present the history, structures, signaling mechanisms, regulation, and biological processes in which activins and inhibins participate, including several recently discovered biological activities and functional antagonists. The potential therapeutic relevance of these advances is also discussed.
Collapse
Affiliation(s)
- Maria Namwanje
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Chester W Brown
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030 Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030 Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
32
|
Hardy CL, Rolland JM, O'Hehir RE. The immunoregulatory and fibrotic roles of activin A in allergic asthma. Clin Exp Allergy 2016; 45:1510-22. [PMID: 25962695 PMCID: PMC4687413 DOI: 10.1111/cea.12561] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activin A, a member of the TGF-β superfamily of cytokines, was originally identified as an inducer of follicle stimulating hormone release, but has since been ascribed roles in normal physiological processes, as an immunoregulatory cytokine and as a driver of fibrosis. In the last 10–15 years, it has also become abundantly clear that activin A plays an important role in the regulation of asthmatic inflammation and airway remodelling. This review provides a brief introduction to the activin A/TGF-β superfamily, focussing on the regulation of receptors and signalling pathways. We examine the contradictory evidence for generalized pro- vs. anti-inflammatory effects of activin A in inflammation, before appraising its role in asthmatic inflammation and airway remodelling specifically by evaluating data from both murine models and clinical studies. We identify key issues to be addressed, paving the way for safe exploitation of modulation of activin A function for treatment of allergic asthma and other inflammatory lung diseases.
Collapse
Affiliation(s)
- C L Hardy
- Department of Allergy, Immunology & Respiratory Medicine, Monash University and The Alfred Hospital, Melbourne, Vic., Australia.,Department of Immunology, Monash University, Melbourne, Vic., 3004, Australia
| | - J M Rolland
- Department of Allergy, Immunology & Respiratory Medicine, Monash University and The Alfred Hospital, Melbourne, Vic., Australia.,Department of Immunology, Monash University, Melbourne, Vic., 3004, Australia
| | - R E O'Hehir
- Department of Allergy, Immunology & Respiratory Medicine, Monash University and The Alfred Hospital, Melbourne, Vic., Australia.,Department of Immunology, Monash University, Melbourne, Vic., 3004, Australia
| |
Collapse
|
33
|
Fujii H, Shinzaki S, Iijima H, Wakamatsu K, Iwamoto C, Sobajima T, Kuwahara R, Hiyama S, Hayashi Y, Takamatsu S, Uozumi N, Kamada Y, Tsujii M, Taniguchi N, Takehara T, Miyoshi E. Core Fucosylation on T Cells, Required for Activation of T-Cell Receptor Signaling and Induction of Colitis in Mice, Is Increased in Patients With Inflammatory Bowel Disease. Gastroenterology 2016; 150:1620-1632. [PMID: 26965517 DOI: 10.1053/j.gastro.2016.03.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/27/2016] [Accepted: 03/01/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS Attachment of a fucose molecule to the innermost N-glycan in a glycoprotein (core fucosylation) regulates the activity of many growth factor receptors and adhesion molecules. The process is catalyzed by α1-6 fucosyltransferase (FUT8) and required for immune regulation, but it is not clear whether this process is dysregulated during disease pathogenesis. We investigated whether core fucosylation regulates T-cell activation and induction of colitis in mice, and is altered in patients with inflammatory bowel disease (IBD). METHODS Biopsy samples were collected from inflamed and noninflamed regions of intestine from patients (8 with Crohn's disease, 4 with ulcerative colitis, and 4 without IBD [controls]) at Osaka University Hospital. Colitis was induced in FUT8-deficient (Fut8(-/-)) mice and Fut8(+/+) littermates by administration of trinitrobenzene sulfonic acid. Intestinal tissues were collected and analyzed histologically. Immune cells were collected and analyzed by lectin flow cytometry, immunofluorescence, and reverse-transcription polymerase chain reaction, as well as for production of cytokines and levels of T-cell receptor (TCR) in lipid raft fractions. T-cell function was analyzed by intraperitoneal injection of CD4(+)CD62L(+) naïve T cells into RAG2-deficient mice. RESULTS Levels of core fucosylation were increased on T cells from mice with colitis, compared with mice without colitis, as well as on inflamed mucosa from patients with IBD, compared with their noninflamed tissues or tissues from control patients. Fut8(-/-) mice developed less-severe colitis than Fut8(+/+) mice, and T cells from Fut8(-/-) mice produced lower levels of T-helper 1 and 2 cytokines. Adoptive transfer of Fut8(-/-) T cells to RAG2-deficient mice reduced the severity of colitis. Compared with CD4(+) T cells from Fut8(+/+) mice, those from Fut8(-/-) mice expressed similar levels of TCR and CD28, but these proteins did not contain core fucosylation. TCR complexes formed on CD4(+) T cells from Fut8(-/-) mice did not signal properly after activation and were not transported to lipid rafts. CONCLUSIONS Core fucosylation of the TCR is required for T-cell signaling and production of inflammatory cytokines and induction of colitis in mice. Levels of TCR core fucosylation are increased on T cells from intestinal tissues of patients with IBD; this process might be blocked as a therapeutic strategy.
Collapse
Affiliation(s)
- Hironobu Fujii
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kana Wakamatsu
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chizuru Iwamoto
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoaki Sobajima
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryusuke Kuwahara
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| | - Satoshi Hiyama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshito Hayashi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinji Takamatsu
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naofumi Uozumi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Kamada
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masahiko Tsujii
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
34
|
Vitamin K deficiency leads to exacerbation of murine dextran sulfate sodium-induced colitis. J Gastroenterol 2016; 51:346-56. [PMID: 26314836 DOI: 10.1007/s00535-015-1112-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 08/06/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) often exhibit vitamin K deficiency. Vitamin K has been shown to inhibit inflammation via interleukin (IL)-6 suppression. This study aimed to evaluate the effect of vitamin K in a murine model of colitis. METHODS Colitis was induced using dextran sulfate sodium (DSS) in mice fed either a vitamin K-deficient (K-def) or a vitamin K-supplemented (K-sup) diet. The clinical and histological severity of colitis was assessed, and levels of cytokine production from the spleen and colonic lamina propria were measured by enzyme-linked immunosorbent assay and quantitative real-time reverse transcription polymerase chain reaction. Cytokine expression levels in CD4(+), CD11b(+), and CD19(+) cells in the presence and absence of vitamin K [menatetrenone (MK-4)] were measured in vitro and apoptosis was determined by caspase 3/7 activity and Annexin V staining. RESULTS DSS administration resulted in significantly more severe body weight loss, shorter colon length, and higher histological scores in mice fed a K-def diet than those fed a K-sup diet. IL-6 expression in lamina propria mononuclear cells was significantly higher in the K-def group than in the K-sup group. IL-6 expression was significantly decreased in the presence of MK-4 in CD19(+) cells, but not in the CD4(+) and CD11b(+) subpopulations. Apoptotic cell population in CD19(+) cells was increased in the presence of MK-4 in vitro and in vivo. CONCLUSIONS Vitamin K exerts a protective effect against DSS colitis; this effect is associated with IL-6 downregulation. Vitamin K could be a potential treatment target for IBD.
Collapse
|
35
|
Shinzaki S, Ishii M, Fujii H, Iijima H, Wakamatsu K, Kawai S, Shiraishi E, Hiyama S, Inoue T, Hayashi Y, Kuwahara R, Takamatsu S, Kamada Y, Morii E, Tsujii M, Takehara T, Miyoshi E. N-Acetylglucosaminyltransferase V exacerbates murine colitis with macrophage dysfunction and enhances colitic tumorigenesis. J Gastroenterol 2016; 51:357-69. [PMID: 26349931 DOI: 10.1007/s00535-015-1119-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 08/20/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Oligosaccharide structures and their alterations have important roles in modulating intestinal inflammation. N-Acetylglucosaminyltransferase V (GnT-V) is involved in the biosynthesis of N-acetylglucosamine (GlcNAc) by β1,6-branching on N-glycans and is induced in various pathologic processes, such as inflammation and regeneration. GnT-V alters host immune responses by inhibiting the functions of CD4(+) T cells and macrophages. The present study aimed to clarify the role of GnT-V in intestinal inflammation using GnT-V transgenic mice. METHODS Colitis severity was compared between GnT-V transgenic mice and wild-type mice. β1,6-GlcNAc levels were investigated by phytohemagglutinin-L4 lectin blotting and flow cytometry. We investigated phagocytosis of macrophages by measuring the number of peritoneal-macrophage-ingested fluorescent latex beads by flow cytometry. Cytokine production in the culture supernatant of mononuclear cells from the spleen, mesenteric lymph nodes, and bone-marrow-derived macrophages was determined by enzyme-linked immunosorbent assay. Clodronate liposomes were intravenously injected to deplete macrophages in vivo. Chronic-colitis-associated tumorigenesis was assessed after 9 months of repeated administration of dextran sodium sulfate (DSS). RESULTS DSS-induced colitis and colitis induced by trinitrobenzene sulfonic acid were markedly exacerbated in GnT-V transgenic mice compared with wild-type mice. Production of interleukin-10 and phagocytosis of macrophages were significantly impaired in GnT-V transgenic mice compared with wild-type mice. Clodronate liposome treatment to deplete macrophages blocked the exacerbation of DSS-induced colitis and impairment of interleukin-10 production in GnT-V transgenic mice. Chronic-colitis-associated tumorigenesis was significantly increased in GnT-V transgenic mice. CONCLUSIONS Overexpression of GnT-V exacerbated murine experimental colitis by inducing macrophage dysfunction, thereby enhancing colorectal tumorigenesis.
Collapse
Affiliation(s)
- Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mayuko Ishii
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hironobu Fujii
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kana Wakamatsu
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shoichiro Kawai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eri Shiraishi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Hiyama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takahiro Inoue
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshito Hayashi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryusuke Kuwahara
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, 7-1, Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Shinji Takamatsu
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihiro Kamada
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiko Tsujii
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
36
|
de Kretser DM, Bensley JG, Phillips DJ, Levvey BJ, Snell GI, Lin E, Hedger MP, O’Hehir RE. Substantial Increases Occur in Serum Activins and Follistatin during Lung Transplantation. PLoS One 2016; 11:e0140948. [PMID: 26820896 PMCID: PMC4731072 DOI: 10.1371/journal.pone.0140948] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/03/2015] [Indexed: 01/08/2023] Open
Abstract
Background Lung transplantation exposes the donated lung to a period of anoxia. Re-establishing the circulation after ischemia stimulates inflammation causing organ damage. Since our published data established that activin A is a key pro-inflammatory cytokine, we assessed the roles of activin A and B, and their binding protein, follistatin, in patients undergoing lung transplantation. Methods Sera from 46 patients participating in a published study of remote ischemia conditioning in lung transplantation were used. Serum activin A and B, follistatin and 11 other cytokines were measured in samples taken immediately after anaesthesia induction, after remote ischemia conditioning or sham treatment undertaken just prior to allograft reperfusion and during the subsequent 24 hours. Results Substantial increases in serum activin A, B and follistatin occurred after the baseline sample, taken before anaesthesia induction and peaked immediately after the remote ischemia conditioning/sham treatment. The levels remained elevated 15 minutes after lung transplantation declining thereafter reaching baseline 2 hours post-transplant. Activin B and follistatin concentrations were lower in patients receiving remote ischemia conditioning compared to sham treated patients but the magnitude of the decrease did not correlate with early transplant outcomes. Conclusions We propose that the increases in the serum activin A, B and follistatin result from a combination of factors; the acute phase response, the reperfusion response and the use of heparin-based anti-coagulants.
Collapse
Affiliation(s)
- David M. de Kretser
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
- Hudson Institute of Medical Research, Clayton, Victoria, Australia
- * E-mail:
| | - Jonathan G. Bensley
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | | | - Bronwyn J. Levvey
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
- Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia
| | - Greg I. Snell
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
- Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Enjarn Lin
- Department of Anaesthesia and Perioperative Medicine, Alfred Hospital, Melbourne, Victoria, Australia
| | - Mark P. Hedger
- Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Robyn E. O’Hehir
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
- Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Shaffer AD, Feng B, La JH, Joyce SC, Gebhart GF. A novel role for follistatin in hypersensitivity following cystitis. Neurourol Urodyn 2015; 36:286-292. [PMID: 26713850 DOI: 10.1002/nau.22941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/23/2015] [Indexed: 11/09/2022]
Abstract
AIMS Previous studies have shown that the activin-binding protein follistatin reduces inflammation in several mouse models of colitis. To determine whether follistatin also has a beneficial effect following bladder inflammation, we induced cystitis in mice using cyclophosphamide (CYP) and examined the relationship between bladder hypersensitivity and bladder follistatin expression. METHODS Adult female C57BL/6 mice were treated with CYP (100 mg/kg) or vehicle (saline) three times over 5 days. Bladder hypersensitivity was assessed by recording the visceromotor response (VMR) to urinary bladder distension and in vitro single-fiber bladder afferent recording. Follistatin gene expression was measured using qRT-PCR. Immunohistochemistry was employed for further characterization. RESULTS Bladder hypersensitivity was established by day 6 and persisted to day 14 in CYP-treated mice. On day 14, hypersensitivity was accompanied by increases in follistatin gene expression in the bladder. Follistatin-like immunoreactivity colocalized with laminin, and the percentage of structures in the lamina propria that were follistatin-positive was increased in CYP-treated mice. Exogenous follistatin increased VMR and afferent responses to bladder distension in CYP- but not vehicle-treated mice. CONCLUSIONS Chronic bladder pain following CYP treatment is associated with increased follistatin expression in the bladder. These results suggest a novel, pro-nociceptive role for follistatin in cystitis, in contrast with its proposed therapeutic role in colitis. This protein has exciting potential as a biomarker and therapeutic target for bladder hypersensitivity. Neurourol. Urodynam. 36:286-292, 2017. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amber D Shaffer
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bin Feng
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jun-Ho La
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sonali C Joyce
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - G F Gebhart
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
38
|
Snell JN, Westall GP, Snell GI. The potential role of activin and follistatin in lung transplant dysfunction. Expert Rev Respir Med 2015; 9:697-701. [DOI: 10.1586/17476348.2015.1098537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Verhamme FM, Bracke KR, Joos GF, Brusselle GG. Transforming growth factor-β superfamily in obstructive lung diseases. more suspects than TGF-β alone. Am J Respir Cell Mol Biol 2015; 52:653-62. [PMID: 25396302 DOI: 10.1165/rcmb.2014-0282rt] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Asthma and chronic obstructive pulmonary disease are respiratory disorders and a major global health problem with increasing incidence and severity. Genes originally associated with lung development could be relevant in the pathogenesis of chronic obstructive pulmonary disease/asthma, owing to either an early-life origin of adult complex diseases or their dysregulation in adulthood upon exposure to environmental stressors (e.g., smoking). The transforming growth factor (TGF)-β superfamily is conserved through evolution and is involved in a range of biological processes, both during development and in adult tissue homeostasis. TGF-β1 has emerged as an important regulator of lung and immune system development. However, considerable evidence has been presented for a role of many of the other ligands of the TGF-β superfamily in lung pathology, including activins, bone morphogenetic proteins, and growth differentiation factors. In this review, we summarize the current knowledge on the mechanisms by which activin, bone morphogenetic protein, and growth differentiation factor signaling contribute to the pathogenesis of obstructive airway diseases.
Collapse
Affiliation(s)
- Fien M Verhamme
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | | | | |
Collapse
|
40
|
Holtan SG, Verneris MR, Schultz KR, Newell LF, Meyers G, He F, DeFor TE, Vercellotti GM, Slungaard A, MacMillan ML, Cooley SA, Blazar BR, Panoskaltsis-Mortari A, Weisdorf DJ. Circulating angiogenic factors associated with response and survival in patients with acute graft-versus-host disease: results from Blood and Marrow Transplant Clinical Trials Network 0302 and 0802. Biol Blood Marrow Transplant 2015; 21:1029-36. [PMID: 25759146 PMCID: PMC4426052 DOI: 10.1016/j.bbmt.2015.02.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/17/2015] [Indexed: 12/26/2022]
Abstract
Circulating angiogenic factors (AF) reflect tissue healing capacity, although some AF can also contribute to inflammation and are indicative of endothelial dysfunction. The AF milieu in acute graft-versus-host disease (aGVHD) has not been broadly characterized. We hypothesized that patients with abundant AF involved in repair/regeneration versus those mediating damage/inflammation would have improved outcomes. Circulating AF known predominantly for repair/regeneration (epidermal growth factor [EGF], fibroblast growth factor-1 and -2, heparin binding-EGF-like growth factor, and vascular endothelial growth factor-A [VEGF-A], -C, and -D) and for damage/inflammation (angiopoietin-2, endothelin-1, soluble endoglin [sEng], follistatin [FS], leptin, and placental growth factor [PlGF]) were measured in a discovery set of hematopoietic cell recipients with grade III and IV aGVHD and compared with controls, then validated in 2 aGVHD cohorts enrolled in Blood and Marrow Transplant Clinical Trials Network (BMT CTN) trials 0302 (n = 105, serum) and 0802 (n = 158, plasma) versus controls without aGVHD (n = 53, serum). Levels of EGF and VEGF-A were lower than in controls at the onset of aGVHD in both trials and higher with complete response to first-line aGVHD therapy in CTN 0802. FS and PlGF were elevated in aGVHD measured in either serum or plasma. At day 28 after initial aGVHD therapy, elevated FS was an independent negative prognostic factor for survival in both cohorts (hazard ratio, 9.3 in CTN 0302; 2.8 in CTN 0802). These data suggest that circulating AF are associated with clinical outcomes after aGVHD and, thus, may contribute to both pathogenesis and recovery.
Collapse
Affiliation(s)
- Shernan G Holtan
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota.
| | - Michael R Verneris
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Kirk R Schultz
- Division of Pediatric Hematology/Oncology/BMT, BC Children's Hospital, Vancouver, University of British Columbia; Blood and Marrow Transplant Clinical Trials Network, Rockville, Maryland
| | - Laura F Newell
- Blood and Marrow Transplant Program, Oregon Health & Science University, Portland, Oregon
| | - Gabrielle Meyers
- Blood and Marrow Transplant Program, Oregon Health & Science University, Portland, Oregon
| | - Fiona He
- School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Todd E DeFor
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Gregory M Vercellotti
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Arne Slungaard
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Sarah A Cooley
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Bruce R Blazar
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | | | - Daniel J Weisdorf
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota; Blood and Marrow Transplant Clinical Trials Network, Rockville, Maryland
| |
Collapse
|
41
|
Follistatin alleviates synovitis and articular cartilage degeneration induced by carrageenan. Int J Inflam 2014; 2014:959271. [PMID: 25574420 PMCID: PMC4276300 DOI: 10.1155/2014/959271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 11/06/2014] [Accepted: 11/19/2014] [Indexed: 12/23/2022] Open
Abstract
Activins are proinflammatory cytokines which belong to the TGFβ superfamily. Follistatin is an extracellular decoy receptor for activins. Since both activins and follistatin are expressed in articular cartilage, we hypothesized that activin-follistatin signaling participates in the process of joint inflammation and cartilage degeneration. To test this hypothesis, we examined the effects of follistatin in a carrageenan-induced mouse arthritis model. Synovitis induced by intra-articular injection of carrageenan was significantly alleviated by preinjection with follistatin. Macrophage infiltration into the synovial membrane was significantly reduced in the presence of follistatin. In addition, follistatin inhibited proteoglycan erosion induced by carrageenan in articular cartilage. These data indicate that activin-follistatin signaling is involved in joint inflammation and cartilage homeostasis. Our data suggest that follistatin can be a new therapeutic target for inflammation-induced articular cartilage degeneration.
Collapse
|
42
|
Mukai A, Iijima H, Hiyama S, Fujii H, Shinzaki S, Inoue T, Shiraishi E, Kawai S, Araki M, Hayashi Y, Kondo J, Mizushima T, Kanto T, Egawa S, Nishida T, Tsujii M, Takehara T. Regulation of anergy-related ubiquitin E3 ligase, GRAIL, in murine models of colitis and patients with Crohn's disease. J Gastroenterol 2014; 49:1524-35. [PMID: 24356810 DOI: 10.1007/s00535-013-0923-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 12/04/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Abrogating tolerance is a critical step in the pathogenesis of Crohn's disease (CD). T cell-anergy is one of the main mechanisms of tolerance and is regulated by the gene related to anergy in lymphocytes (GRAIL). This study investigated the expressions and regulation of GRAIL in CD and murine colitis models. METHODS Expressions of GRAIL mRNA and protein in CD4+ T cells were investigated in the peripheral blood and mucosal tissues of patients with CD, mice with dextran sodium salt (DSS)-induced colitis, and Il-10-deficient mice. MicroRNAs responsible for the regulation of GRAIL were examined by miRNA microarray. GRAIL-overexpressing T cells were intravenously injected in mice with DSS-induced colitis. RESULTS The GRAIL expression was higher in the lamina propria (LP) CD4+ T cells of CD patients than of the control subjects, while it was lower in the peripheral blood CD4+ T cells of the CD patients than of the control subjects. The GRAIL mRNA expression was lower, but the GRAIL protein expression was higher in the LP of colitic mice than that of non-colitic mice. The miRNA microarray identified miR-290-5p as an miRNA that inhibits expression of the GRAIL protein and that is highly expressed in the LP of non-colitic mice. GRAIL-expressing T cells expressed regulatory T cell markers and showed suppressive effects in murine DSS-induced colitis. CONCLUSIONS Our results show that expression of GRAIL is uniquely regulated by the specific miRNA in the intestinal mucosa, and suggest that GRAIL may associate with the pathophysiology of CD.
Collapse
Affiliation(s)
- Akira Mukai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 K1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Impaired resolution of inflammation in the Endoglin heterozygous mouse model of chronic colitis. Mediators Inflamm 2014; 2014:767185. [PMID: 25114380 PMCID: PMC4121192 DOI: 10.1155/2014/767185] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 12/16/2022] Open
Abstract
Endoglin is a coreceptor of the TGF-β superfamily predominantly expressed on the vascular endothelium and selective subsets of immune cells. We previously demonstrated that Endoglin heterozygous (Eng (+/-)) mice subjected to dextran sulfate sodium (DSS) developed persistent gut inflammation and pathological angiogenesis. We now report that colitic Eng (+/-) mice have low colonic levels of active TGF-β1, which was associated with reduced expression of thrombospondin-1, an angiostatic factor known to activate TGF-β1. We also demonstrate dysregulated expression of BMPER and follistatin, which are extracellular regulators of the TGF-β superfamily that modulate angiogenesis and inflammation. Heightened colonic levels of the neutrophil chemoattractant and proangiogenic factor, CXCL1, were also observed in DSS-treated Eng (+/-) mice. Interestingly, despite increased macrophage and neutrophil infiltration, a gut-specific reduction in expression of the key phagocytic respiratory burst enzymes, NADPH oxidase 2 (Nox-2) and myeloperoxidase, was seen in Eng (+/-) mice undergoing persistent inflammation. Taken together, these findings suggest that endoglin is required for TGF-β superfamily mediated resolution of inflammation and fully functional myeloid cells.
Collapse
|
44
|
Follistatin, an activin antagonist, ameliorates renal interstitial fibrosis in a rat model of unilateral ureteral obstruction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:376191. [PMID: 24883308 PMCID: PMC4026945 DOI: 10.1155/2014/376191] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/14/2014] [Indexed: 02/04/2023]
Abstract
Activin, a member of the TGF-β superfamily, regulates cell growth and differentiation in various cell types. Activin A acts as a negative regulator of renal development as well as tubular regeneration after renal injury. However, it remains unknown whether activin A is involved in renal fibrosis. To clarify this issue, we utilized a rat model of unilateral ureteral obstruction (UUO). The expression of activin A was significantly increased in the UUO kidneys compared to that in contralateral kidneys. Activin A was detected in glomerular mesangial cells and interstitial fibroblasts in normal kidneys. In UUO kidneys, activin A was abundantly expressed by interstitial α-SMA-positive myofibroblasts. Administration of recombinant follistatin, an activin antagonist, reduced the fibrotic area in the UUO kidneys. The number of proliferating cells in the interstitium, but not in the tubules, was significantly lower in the follistatin-treated kidneys. Expression of α-SMA, deposition of type I collagen and fibronectin, and CD68-positive macrophage infiltration were significantly suppressed in the follistatin-treated kidneys. These data suggest that activin A produced by interstitial fibroblasts acts as a potent profibrotic factor during renal fibrosis. Blockade of activin A action may be a novel approach for the prevention of renal fibrosis progression.
Collapse
|
45
|
Serum activin A and B levels predict outcome in patients with acute respiratory failure: a prospective cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R263. [PMID: 24172607 PMCID: PMC4057391 DOI: 10.1186/cc13093] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 09/30/2013] [Indexed: 12/31/2022]
Abstract
INTRODUCTION 30 day mortality in patients with Acute Respiratory Failure (ARF) is approximately 30%, defined as patients requiring ventilator support for more than 6 hours. Novel biomarkers are needed to predict patient outcomes and to guide potential future therapies. The activins A and B, members of the Transforming Growth Factor β family of proteins, and their binding protein, follistatin, have recently been shown to be important regulators of inflammation and fibrosis but no substantial data are available concerning their roles in ARF. METHODS Specific assays for activin A, B and follistatin were used and the results analyzed according to diagnostic groups as well as according to standard measures in intensive care. Multivariable logistic regression was used to create a model to predict death at 90 days and 12 months from the onset of the ARF. RESULTS Serum activin A and B were significantly elevated in most patients and in most of the diagnostic groups. Patients who had activin A and/or B concentrations above the reference maximum were significantly more likely to die in the 12 months following admission [either activin A or B above reference maximum: Positive Likelihood Ratio [LR+] 1.65 [95% CI 1.28-2.12, P = 0.00013]; both activin A and B above reference maximum: LR + 2.78 [95% CI 1.96-3.95, P < 0.00001]. The predictive model at 12 months had an overall accuracy of 80.2% [95% CI 76.6-83.3%]. CONCLUSIONS The measurement of activin A and B levels in these patients with ARF would have assisted in predicting those at greatest risk of death. Given the existing data from animal studies linking high activin A levels to significant inflammatory challenges, the results from this study suggest that approaches to modulate activin A and B bioactivity should be explored as potential therapeutic agents.
Collapse
|
46
|
Hedger MP, de Kretser DM. The activins and their binding protein, follistatin-Diagnostic and therapeutic targets in inflammatory disease and fibrosis. Cytokine Growth Factor Rev 2013; 24:285-95. [PMID: 23541927 DOI: 10.1016/j.cytogfr.2013.03.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/05/2013] [Indexed: 02/05/2023]
Abstract
The activins, as members of the transforming growth factor-β superfamily, are pleiotrophic regulators of cell development and function, including cells of the myeloid and lymphoid lineages. Clinical and animal studies have shown that activin levels increase in both acute and chronic inflammation, and are frequently indicators of disease severity. Moreover, inhibition of activin action can reduce inflammation, damage, fibrosis and morbidity/mortality in various disease models. Consequently, activin A and, more recently, activin B are emerging as important diagnostic tools and therapeutic targets in inflammatory and fibrotic diseases. Activin antagonists such as follistatin, an endogenous activin-binding protein, offer considerable promise as therapies in conditions as diverse as sepsis, liver fibrosis, acute lung injury, asthma, wound healing and ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- M P Hedger
- Monash Institute of Medical Research, Monash University, Melbourne, Victoria, Australia.
| | | |
Collapse
|
47
|
Activin, neutrophils, and inflammation: just coincidence? Semin Immunopathol 2013; 35:481-99. [PMID: 23385857 PMCID: PMC7101603 DOI: 10.1007/s00281-013-0365-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/17/2013] [Indexed: 01/18/2023]
Abstract
During the 26 years that have elapsed since its discovery, activin-A, a member of the transforming growth factor β super-family originally discovered from its capacity to stimulate follicle-stimulating hormone production by cultured pituitary gonadotropes, has been established as a key regulator of various fundamental biological processes, such as development, homeostasis, inflammation, and tissue remodeling. Deregulated expression of activin-A has been observed in several human diseases characterized by an immuno-inflammatory and/or tissue remodeling component in their pathophysiology. Various cell types have been recognized as sources of activin-A, and plentiful, occasionally contradicting, functions have been described mainly by in vitro studies. Not surprisingly, both harmful and protective roles have been postulated for activin-A in the context of several disorders. Recent findings have further expanded the functional repertoire of this molecule demonstrating that its ectopic overexpression in mouse airways can cause pathology that simulates faithfully human acute respiratory distress syndrome, a disorder characterized by strong involvement of neutrophils. This finding when considered together with the recent discovery that neutrophils constitute an important source of activin-A in vivo and earlier observations of upregulated activin-A expression in diseases characterized by strong activation of neutrophils may collectively imply a more intimate link between activin-A expression and neutrophil reactivity. In this review, we provide an outline of the functional repertoire of activin-A and suggest that this growth factor functions as a guardian of homeostasis, a modulator of immunity and an orchestrator of tissue repair activities. In this context, a relationship between activin-A and neutrophils may be anything but coincidental.
Collapse
|
48
|
|
49
|
de Kretser DM, O'Hehir RE, Hardy CL, Hedger MP. The roles of activin A and its binding protein, follistatin, in inflammation and tissue repair. Mol Cell Endocrinol 2012; 359:101-6. [PMID: 22037168 DOI: 10.1016/j.mce.2011.10.009] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 10/06/2011] [Accepted: 10/13/2011] [Indexed: 01/15/2023]
Abstract
Activin A, a member of the transforming growth factor-β superfamily of cytokines, is a critical controller of inflammation, immunity and fibrosis. It is rapidly released into the blood following a lipopolysaccharide challenge in experimental animals, through activation of the Toll-like receptor 4 signalling pathway. Blocking activin action by pre-treatment with its binding protein, follistatin, modifies the inflammatory cytokine cascade, and reduces the severity of the subsequent inflammatory response and mortality. Likewise, high serum levels of activin A are predictive of death in patients with septicaemia. However, activin A has complex immunomodulatory actions. It is produced by inflammatory macrophages, but can regulate either pro- or anti-inflammatory responses in these cells, depending on their prior activation status. Activin A is also produced by Th2 cells, and stimulates antibody production by B cells and the development of regulatory T cells. Production of activin A during inflammatory responses stimulates fibrosis and tissue remodelling, and follistatin inhibits these actions of activin A. The modulation of activin by follistatin may represent an important therapeutic target for the modulation and amelioration of inflammatory and fibrotic disorders.
Collapse
Affiliation(s)
- David M de Kretser
- Monash Institute of Medical Research and the Department of Immunology and Pathology, Monash University, Clayton Victoria 3800, Australia.
| | | | | | | |
Collapse
|
50
|
Wu H, Chen Y, Winnall WR, Phillips DJ, Hedger MP. Acute regulation of activin A and its binding protein, follistatin, in serum and tissues following lipopolysaccharide treatment of adult male mice. Am J Physiol Regul Integr Comp Physiol 2012; 303:R665-75. [PMID: 22855279 DOI: 10.1152/ajpregu.00478.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Activin A, a member of the transforming growth factor-β family, increases in the circulation within 1 h after administration of bacterial LPS. To clarify the origins of this rapid increase, the distribution of activin A and its binding protein, follistatin, and their production following LPS treatment, were assessed in adult male mice. In untreated mice, activin A was detectable in all 23 tissues examined, with highest mRNA expression (as measured by quantitative RT-PCR) was found in the liver, and the largest concentration of activin A protein (by ELISA) was found in the bone marrow. Likewise, follistatin mRNA and protein were present in all tissues, with highest expression in the vas deferens. Activin A and follistatin mRNA did not increase significantly in any tissue within the first hour after LPS, but activin A protein decreased by 35% in the bone marrow and increased 5-fold in the lung. No significant changes were observed in any other tissue. Activin A reached a peak in the circulation 1 h following LPS, and then declined. Cycloheximide, an inhibitor of protein translation, reduced this increase of activin A by more than 50%. Actinomycin D, an inhibitor of mRNA transcription, had no effect. Circulating follistatin did not increase until 4 h after LPS and was not affected by either inhibitor. These data indicate that the rapid increase in circulating activin A during LPS-induced inflammation is regulated at the posttranscriptional level, apparently from newly translated and stored protein, and implicate bone marrow-derived cells, and, in particular, neutrophils, as a significant source of this preformed activin A.
Collapse
Affiliation(s)
- Hui Wu
- Monash Institute of Medical Research, Monash University, Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|