51
|
Rana T. Unravelling of nitric oxide signalling: A potential biomarker with multifaceted complex mechanism associated with canine inflammatory bowel disease (IBD). Anaerobe 2020; 66:102288. [PMID: 33132137 DOI: 10.1016/j.anaerobe.2020.102288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/14/2020] [Accepted: 10/22/2020] [Indexed: 01/16/2023]
Abstract
Inflammatory bowel disease (IBD) is an important chronic condition associated with the infection affecting the gastrointestinal tract (G.I.) in dogs. Several factors' viz. gastrointestinal tract lymphoid tissue (GALT), permeability defects, genetic, ischemic, biochemical, psychosomatic disorders, infectious and parasitic agents, dietary allergies, and adverse drug reactions are associated with inflammatory bowel disease. The most noticeable clinical signs are vomiting, diarrhea, changes in appetite, weight loss, anorexia, ascites and peripheral edema. Nitric oxide (NO), a pleiotropic free radical messenger molecule plays an immense role in playing mucosal inflammation in the intestine through activation of NO synthase enzyme (iNOS). The complex mechanism associated with inflammation in the G.I. tract is also correlated with the expression of iNOS, enzymatic activity, and NO production. NO exerts a beneficial role in maintaining epithelial permeability as well as the immune response in acute colitis. But the excessive production of NO causes adverse effects. In the review, the author suggests that a complex phenomenon is associated with competing biochemical pathways triggered by NO through the regulation of mucosal inflammation in inflammatory bowel disease. This review is a unique compilation about the role of NO in the pathogenesis of inflammatory bowel disease of the dogs.
Collapse
Affiliation(s)
- Tanmoy Rana
- Department of Veterinary Clinical Complex (V.M.E.J.), West Bengal University of Animal & Fishery Sciences, Kolkata, India.
| |
Collapse
|
52
|
Nijakowski K, Surdacka A. Salivary Biomarkers for Diagnosis of Inflammatory Bowel Diseases: A Systematic Review. Int J Mol Sci 2020; 21:ijms21207477. [PMID: 33050496 PMCID: PMC7589027 DOI: 10.3390/ijms21207477] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Saliva as a biological fluid has a remarkable potential in the non-invasive diagnostics of several systemic disorders. Inflammatory bowel diseases are chronic inflammatory disorders of the gastrointestinal tract. This systematic review was designed to answer the question “Are salivary biomarkers reliable for the diagnosis of inflammatory bowel diseases?”. Following the inclusion and exclusion criteria, eleven studies were included (according to PRISMA statement guidelines). Due to their heterogeneity, the potential salivary markers for IBD were divided into four groups: oxidative status markers, inflammatory cytokines, microRNAs and other biomarkers. Active CD patients manifest decreased activity of antioxidants (e.g., glutathione, catalase) and increased lipid peroxidation. Therefore, malondialdehyde seems to be a good diagnostic marker of CD. Moreover, elevated concentrations of proinflammatory cytokines (such as interleukin 1β, interleukin 6 or tumour necrosis factor α) are associated with the activity of IBD. Additionaly, selected miRNAs are altered in saliva (overexpressed miR-101 in CD; overexpressed miR-21, miR-31, miR-142-3p and underexpressed miR-142-5p in UC). Among other salivary biomarkers, exosomal PSMA7, α-amylase and calprotectin are detected. In conclusion, saliva contains several biomarkers which can be used credibly for the early diagnosis and regular monitoring of IBD. However, further investigations are necessary to validate these findings, as well as to identify new reliable salivary biomarkers.
Collapse
|
53
|
Dong N, Xue C, Zhang L, Zhang T, Wang C, Bi C, Shan A. Oleanolic acid enhances tight junctions and ameliorates inflammation in Salmonella typhimurium-induced diarrhea in mice via the TLR4/NF-κB and MAPK pathway. Food Funct 2020; 11:1122-1132. [PMID: 31825448 DOI: 10.1039/c9fo01718f] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Salmonella typhimurium (S.T) is a common cause of acute, self-limiting food-borne diarrhea with severe intestinal inflammation and intestinal barrier damage. Oleanolic acid (OA), isolated from almost 2000 plant species, has been shown to have anti-inflammatory roles. The purpose of this study was to investigate the potential protective effects of OA on S.T-induced diarrhea and enteritis and to elucidate its anti-inflammatory mechanisms. A total of eighty BALB/c mice (4-week-old) were randomly divided into the control group (no S.T, no OA), the S.T group (S.T only), the S.T + OA group (S.T plus 100 mg kg-1 OA) and the OA group (100 mg kg-1 OA only). Compared with the S.T group, OA administration significantly reduced clinical symptoms and weight loss, and the severity of diarrhea and intestinal structural damage was significantly alleviated, which was confirmed by a decrease in the diarrhea index (DI) and jejunal histological damage. In addition, in the infected jejunum, OA maintained the expression and localization of occludin, claudin-1 and ZO-1 to protect the jejunal barrier, thereby maintaining the integrity of the gut barrier. Finally, OA treatment not only reduced the levels of COX-2 and iNOS but also inhibited the secretion of pro-inflammatory cytokines, such as IL-1β, IL-6 and TNF-α. Furthermore, western blotting results showed that OA treatment significantly inhibited IκB phosphorylation and degradation in intestinal tissues and the nuclear translocation of p65, and OA also decreased the level of TLR4 and the activation of the MAPK pathway. To summarise, OA can maintain the intestinal tight junction barrier and prevent diarrhea caused by S.T. as well as reduce intestinal inflammation through the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Na Dong
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
54
|
Tachibana M, Watanabe N, Koda Y, Oya Y, Kaminuma O, Katayama K, Fan Z, Sakurai F, Kawabata K, Hiroi T, Mizuguchi H. Ablation of IL-17A leads to severe colitis in IL-10-deficient mice: implications of myeloid-derived suppressor cells and NO production. Int Immunol 2020; 32:187-201. [PMID: 31755523 PMCID: PMC7067553 DOI: 10.1093/intimm/dxz076] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 11/20/2019] [Indexed: 12/18/2022] Open
Abstract
IL-10 is an immune regulatory cytokine and its genetic defect leads to gastrointestinal inflammation in humans and mice. Moreover, the IL-23/Th17 axis is known to be involved in these inflammatory disorders. IL-17A, a representative cytokine produced by Th17 cells, has an important role for the pathological process of inflammatory diseases. However, the precise function of IL-17A in inflammatory bowel disease (IBD) remains controversial. In this study, we evaluated the effect of IL-17A on colitis in IL-10-deficient (Il10−/−) mice. Mice lacking both IL-10 and IL-17A (Il10−/−Il17a−/−) suffered from fatal wasting and manifested more severe colitis compared with Il10−/−Il17a+/− mice. Moreover, we found that CD11b+Gr-1+ myeloid-derived suppressor cells (MDSCs) accumulated in the bone marrow, spleen and peripheral blood of Il10−/−Il17a−/− mice. These MDSCs highly expressed inducible nitric oxide synthase (iNOS) (Nos2) and suppressed the T-cell response in vitro in a NOS-dependent manner. In correlation with these effects, the concentration of nitric oxide was elevated in the serum of Il10−/−Il17a−/− mice. Surprisingly, the severe colitis observed in Il10−/−Il17a−/− mice was ameliorated in Il10−/−Il17a−/−Nos2−/− mice. Our findings suggest that IL-17A plays suppressive roles against spontaneous colitis in Il10−/− mice in an iNOS-dependent manner and inhibits MDSC differentiation and/or proliferation.
Collapse
Affiliation(s)
- Masashi Tachibana
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan.,Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Nobumasa Watanabe
- Department of Genome Medicine, Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuzo Koda
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan
| | - Yukako Oya
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan
| | - Osamu Kaminuma
- Department of Genome Medicine, Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazufumi Katayama
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan
| | - Zifei Fan
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Takachika Hiroi
- Department of Genome Medicine, Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan.,Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| |
Collapse
|
55
|
Bas N, Kayar NA, Baba ZF, Avunduk MC, Haliloğlu S, Alptekin NÖ. Systemic treatment with alpha-tocopherol and/or sodium selenite decreases the progression of experimental periodontitis. Clin Oral Investig 2020; 25:2677-2688. [PMID: 32986166 DOI: 10.1007/s00784-020-03579-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 09/10/2020] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To investigate the effects of sodium selenite (Se) and/or α-tocopherol (αT) applications on the alveolar bone loss (ABL), the number of gingival collagen fibers, inducible nitric oxide synthase (iNOS)+ and CD95+ cell numbers, and serum cytokine concentrations in experimental periodontitis in rats. MATERIALS AND METHODS Forty Sprague Dawley rats were divided into four groups of ten as follows: group A: Se group, group B: αT group, group C: Se and αT combined group, and group D: control group (intraperitoneal (IP) saline injection applied). Using the image analysis method in the connective tissue under the connective epithelium, the numbers of iNOS, CD95 positive cells, and collagen fibers were counted. ELISA kits were used to test the concentrations of serum interleukin (IL)-1β, IL-6, and IL-4. RESULTS The combination of Se and αT (group C) suppressed ABL compared with the control group (group D) (P < 0.05). In group A (Se), the number of iNOS+ cells was smaller than in group D (P < 0.05). CONCLUSION Se has been concluded to inhibit inflammation of the gum due to iNOS. Se and αT can have a remarkable important role in preventing alveolar bone loss, and particularly in combination. CLINICAL RELEVANCE Se and/or αT application may be useful in preventing the destruction of periodontal tissue and treatment of periodontal disease.
Collapse
Affiliation(s)
- Nurgül Bas
- Kayseri Nimet Bayraktar Oral and Dental Health Center, Republic of Turkey Ministry of Health, Kayseri, Turkey
| | - Nezahat Arzu Kayar
- Department of Periodontology, Faculty of Dentistry, Akdeniz University, 07058, Antalya, Turkey.
| | - Z Füsun Baba
- Patology Laboratory, Acıbadem International Hospital, İstanbul, Turkey
| | - Mustafa Cihat Avunduk
- Department of Pathology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Seyfullah Haliloğlu
- Department of Biochemistry, Faculty of Veterinary, Selcuk University, Konya, Turkey
| | - Nilgün Özlem Alptekin
- Department of Periodontology, Faculty of Dentistry, Başkent University, Ankara, Turkey
| |
Collapse
|
56
|
Zhang S, Cho WJ, Jin AT, Kok LY, Shi Y, Heller DE, Lee YAL, Zhou Y, Xie X, Korzenik JR, Lennerz JK, Traverso G. Heparin-Coated Albumin Nanoparticles for Drug Combination in Targeting Inflamed Intestine. Adv Healthc Mater 2020; 9:e2000536. [PMID: 32597571 PMCID: PMC7482138 DOI: 10.1002/adhm.202000536] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/12/2020] [Indexed: 12/18/2022]
Abstract
Targeting areas of inflammation offers potential therapeutic and diagnostic benefits by maximizing drug and imaging marker on-target effects while minimizing systemic exposure that can be associated with adverse side effects. This strategy is particularly beneficial in the management of inflammatory bowel disease (IBD). Here an inflammation-targeting (IT) approach based on heparin-coated human serum albumin nanoparticles (HEP-HSA NPs) that utilize the increased intestinal permeability and changes in electrostatic interaction at the site of intestinal inflammation is described. Using small-molecule and biologic drugs as a model for drug combination, the HEP-HSA NPs demonstrate the capacity to load both drugs simultaneously; the dual-drug loaded HEP-HSA NPs exhibit a higher anti-inflammatory effect than both of the single-drug loaded NPs in vitro and selectively bind to inflamed intestine after enema administration in vivo in a murine model of colitis. Importantly, analyses of the physicochemical characteristics and targeting capacities of these NPs indicate that HEP coating modulates NP binding to the inflamed intestine, providing a foundation for future IT-NP formulation development.
Collapse
Affiliation(s)
- Sufeng Zhang
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Won Joon Cho
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Amy T. Jin
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lie Yun Kok
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yunhua Shi
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David E. Heller
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Young-Ah Lucy Lee
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yixuan Zhou
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xi Xie
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua R. Korzenik
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jochen K. Lennerz
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Traverso
- Dr. S. Zhang, A. T. Jin, Prof. J. R. Korzenik, Prof. G. Traverso Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA. Dr. S. Zhang, W. J. Woo, A. T. Jin, L. Y. Kok, Dr. Y. Shi, D. E. Heller, Y.-A. L. Lee, Y. Zhou, Dr. X. Xie Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Dr. S. Zhang, Prof. J. R. Korzenik, Prof. J. K. Lennerz, Prof. G. Traverso Harvard Medical School, Boston, MA 02115, USA. Prof. J. K. Lennerz Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA. Prof. G. Traverso Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
57
|
Abdelhafez OH, Ali TFS, Fahim JR, Desoukey SY, Ahmed S, Behery FA, Kamel MS, Gulder TAM, Abdelmohsen UR. Anti-Inflammatory Potential of Green Synthesized Silver Nanoparticles of the Soft Coral Nephthea Sp. Supported by Metabolomics Analysis and Docking Studies. Int J Nanomedicine 2020; 15:5345-5360. [PMID: 32801693 PMCID: PMC7395700 DOI: 10.2147/ijn.s239513] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/07/2020] [Indexed: 12/31/2022] Open
Abstract
Background Soft corals have been endorsed as a plentiful source of bioactive compounds with promising anti-inflammatory activities; therefore, exploring their potential as source of anti-inflammatory metabolites has stimulated a growing research interest. Purpose To investigate the anti-inflammatory potential of the soft coral, Nephthea sp., in its bulk and silver nanostructure. Metabolomics analysis of Nephthea sp., followed by molecular docking studies, was also conducted in order to explore and predict the secondary metabolites that might provide its inhibitory actions on inflammation. Materials and Methods The petroleum ether and ethyl acetate fractions were used to synthesize silver nanoparticles. The prepared silver nanoparticles were characterized through UV-vis spectrophotometric, transmission electron microscopy (TEM) and Fourier-transform infrared spectroscopy (FTIR) analyses. Testing for the anti-inflammatory activity was performed against COX-1 and COX-2. Furthermore, liquid chromatography–mass spectrometry (LC–MS) based metabolomics analysis and molecular docking were also applied. Results A variety of secondary metabolites were identified, among them, sesquiterpenes were found to prevail. The petroleum ether and acetone fractions of Nephthea sp. showed the highest COX-2 inhibitory activities, possibly attributable to their substantial contents of terpenoids. Additionally, the green synthesized silver nanoparticles of both the petroleum ether and ethyl acetate fractions of Nephthea sp. demonstrated higher anti-COX-2 properties. Conclusion The obtained results showed the effectiveness of non-targeted metabolomics technique in metabolic profiling of Nephthea sp., helping the search for new bioactive metabolites in future chemical studies on this soft coral. The interesting anti-inflammatory potential of the tested extracts and their nanoparticles could also be relevant to the development of new, effective anti-inflammatory agents.
Collapse
Affiliation(s)
| | - Taha Farouk Shehata Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - John Refaat Fahim
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Samar Yehia Desoukey
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Safwat Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Fathy A Behery
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.,Department of Pharmaceutical Sciences, College of Pharmacy, Riyadh Elm University, Riyadh 11681, Saudi Arabia
| | - Mohamed Salah Kamel
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Tobias A M Gulder
- Chair of Technical Biochemistry, Department of Chemistry and Food Chemistry, Technical University of Dresden, Dresden 01069, Germany
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia 61111, Egypt.,Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| |
Collapse
|
58
|
Yu Y, Zhang G, Han T, Huang H. Analysis of the pharmacological mechanism of Banxia Xiexin decoction in treating depression and ulcerative colitis based on a biological network module. BMC Complement Med Ther 2020; 20:199. [PMID: 32600331 PMCID: PMC7325019 DOI: 10.1186/s12906-020-02988-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/16/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The network pharmacology method was used to predict the active components of Banxia Xiexin decoction, its targets and the key signalling pathways that are activated in the treatment of depression and ulcerative colitis to explore the common mechanism. METHODS The active components and targets of Banxia Xiexin decoction were obtained by searching the ETCM,TCMSP and TCMIP database. The disease targets of depression and ulcerative colitis were obtained by combining the following the DisGeNET, OMIM,Drugbank,CTD and PharmGKB disease databases. The drug and disease target genes were obtained from the intersection of the herbal medicine targets and the disease targets and were imported into the STRING platform for the analysis of PPI network. The network modules were constructed using Cytoscape software. An analysis of the functional annotations of GO terms and KEGG signalling pathways was performed for each network module. Then, the tissue distribution, sub-cellular distribution and protein attributes of the key targets in the pathway were analysed by the BioGPS, Genecards and DisGeNET databases. RESULTS The mechanism of Banxia Xiexin Decoction in the treatment of depression and ulcerative colitis is related to drug reaction, steroid metabolism, lipid metabolism, inflammatory response, oxidative stress response, cell response to lipopolysaccharide, insulin secretion regulation, estradiol response and other biological functions, mainly through the regulation of 5-hydroxytryptamine synaptic, arachidonic acid metabolism, HIF-1 signaling pathway and NF-kappa B signaling pathway can achieve the effect of same treatment for different diseases. CONCLUSIONS The mechanism of Banxia Xiexin Decoction in treating different diseases involves direct or indirect correlation of multiple signal pathways, mainly involved in drug metabolism and lipid metabolism, but also through comprehensive intervention of the body's nervous system, immune system, digestive system and other systems. The effective components of Banxia Xiexin Decoction are mainly act on eight key target proteins (such as ALB, IL6, VEGFA, TNF, PTGS2, MAPK1, STAT3, EGFR) to carry out multi-target effect mechanism, biological mechanism of treating different diseases with the same treatment, and related mechanism of overall treatment, which provide theoretical reference for further research on the material basis and mechanism of Banxiaxiexin decoction on antidepressant and prevention and treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Ying Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355 China
| | - Gong Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355 China
| | - Tao Han
- Graduate Office of Shandong University of Traditional Chinese Medicine, Jinan, 250355 China
| | - Hailiang Huang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355 China
| |
Collapse
|
59
|
Kamalian A, Sohrabi Asl M, Dolatshahi M, Afshari K, Shamshiri S, Momeni Roudsari N, Momtaz S, Rahimi R, Abdollahi M, Abdolghaffari AH. Interventions of natural and synthetic agents in inflammatory bowel disease, modulation of nitric oxide pathways. World J Gastroenterol 2020; 26:3365-3400. [PMID: 32655263 PMCID: PMC7327787 DOI: 10.3748/wjg.v26.i24.3365] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/09/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) refers to a group of disorders characterized by chronic inflammation of the gastrointestinal (GI) tract. The elevated levels of nitric oxide (NO) in serum and affected tissues; mainly synthesized by the inducible nitric oxide synthase (iNOS) enzyme; can exacerbate GI inflammation and is one of the major biomarkers of GI inflammation. Various natural and synthetic agents are able to ameliorate GI inflammation and decrease iNOS expression to the extent comparable with some IBD drugs. Thereby, the purpose of this study was to gather a list of natural or synthetic mediators capable of modulating IBD through the NO pathway. Electronic databases including Google Scholar and PubMed were searched from 1980 to May 2018. We found that polyphenols and particularly flavonoids are able to markedly attenuate NO production and iNOS expression through the nuclear factor κB (NF-κB) and JAK/STAT signaling pathways. Prebiotics and probiotics can also alter the GI microbiota and reduce NO expression in IBD models through a broad array of mechanisms. A number of synthetic molecules have been found to suppress NO expression either dependent on the NF-κB signaling pathway (i.e., dexamethasone, pioglitazone, tropisetron) or independent from this pathway (i.e., nicotine, prednisolone, celecoxib, β-adrenoceptor antagonists). Co-administration of natural and synthetic agents can affect the tissue level of NO and may improve IBD symptoms mainly by modulating the Toll like receptor-4 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Aida Kamalian
- Department of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Masoud Sohrabi Asl
- Department of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mahsa Dolatshahi
- Department of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Khashayar Afshari
- Department of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Shiva Shamshiri
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran 1417614411, Iran
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network, Tehran 1417614411, Iran
| | - Roja Rahimi
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran 1417614411, Iran
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network, Tehran 1417614411, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
60
|
Besednova NN, Zaporozhets TS, Kuznetsova TA, Makarenkova ID, Kryzhanovsky SP, Fedyanina LN, Ermakova SP. Extracts and Marine Algae Polysaccharides in Therapy and Prevention of Inflammatory Diseases of the Intestine. Mar Drugs 2020; 18:E289. [PMID: 32486405 PMCID: PMC7345783 DOI: 10.3390/md18060289] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a serious public health problem worldwide. Current therapeutic strategies that use anti-inflammatory drugs, immunosuppressants, and biological treatments are often ineffective and have adverse health effects. In this regard, the use of natural compounds aimed at key pathogenic therapeutic targets in IBD attracts universal attention. Seaweed is a valuable source of structurally diverse biologically active compounds. The materials presented in the review indicate that seaweed extracts and polysaccharides are effective candidates for the development of drugs, biological food additives, and functional nutrition products for the treatment and prevention of IBD. The structural features of algal polysaccharides provide the possibility of exposure to therapeutic targets of IBD, including proinflammatory cytokines, chemokines, adhesion molecules, nuclear factor NF-kB, intestinal epithelial cells, reactive oxygen and nitrogen. Further study of the relationship between the effect of polysaccharides from different types of algae, with different structure and molecular weights on immune and epithelial cells, intestinal microorganisms will contribute to a deeper understanding of their mechanisms and will help in the development of drugs, dietary supplements, functional foods for the treatment of patients with IBD.
Collapse
Affiliation(s)
- Natalya N. Besednova
- Somov Institute of Epidemiology and Microbiology, Vladivostok 690087, Russia; (N.N.B.); (T.A.K.); (I.D.M.)
| | - Tatyana S. Zaporozhets
- Somov Institute of Epidemiology and Microbiology, Vladivostok 690087, Russia; (N.N.B.); (T.A.K.); (I.D.M.)
| | - Tatyana A. Kuznetsova
- Somov Institute of Epidemiology and Microbiology, Vladivostok 690087, Russia; (N.N.B.); (T.A.K.); (I.D.M.)
| | - Ilona D. Makarenkova
- Somov Institute of Epidemiology and Microbiology, Vladivostok 690087, Russia; (N.N.B.); (T.A.K.); (I.D.M.)
| | - Sergey P. Kryzhanovsky
- School of Biomedicine, Far Eastern Federal University, Vladivostok 690087, Russia; (S.P.K.); (L.N.F.)
| | - Lydmila N. Fedyanina
- School of Biomedicine, Far Eastern Federal University, Vladivostok 690087, Russia; (S.P.K.); (L.N.F.)
| | - Svetlana P. Ermakova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, FEB RAS, Vladivostok 690022, Russia;
| |
Collapse
|
61
|
Li K, Sun S, Kageyama M, Xiao L, Xing G, Gao R, You F, Fu X, Zhang Z. Evaluation of the Immunomodulatory and Anti-Inflammatory Activities of Honey Bee Larva Powder. J Med Food 2020; 23:772-782. [PMID: 32456573 DOI: 10.1089/jmf.2019.4554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Honey bee larva powder (HLP) has traditionally been used as a daily supplement and tonic for health promotion with an uncertain scientific basis. In this study, B16-F10 tumor-bearing mice were established to evaluate the immunomodulatory activity of HLP. The proliferation and apoptosis assays were performed to evaluate the anti-inflammatory activity of honey bee larva extract (HLE) in RAW 264.7 macrophage. The in vivo experimental results demonstrated that the oral administration of freeze-dried HLP (4 and 6 g/kg) significantly enhanced the spleen index, the percentage of CD4+cells, and the ratio of CD4+ and CD8+ T lymphocytes (CD4+/CD8+) in the peripheral blood compared with those in the tumor control mice. The in vitro studies demonstrated the potent immunomodulatory activities of HLE through the induction of RAW 264.7 macrophage proliferation and the mitigation of doxorubicin (DOX)-induced toxicity. HLE also exhibits anti-inflammatory activity by decreasing the production of nitric oxide (NO) and the cytokine level of interleukin-6 (IL-6) in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage. The present study provides important scientific evidence for the immunomodulatory and anti-inflammatory activities of HLP and HLE.
Collapse
Affiliation(s)
- Kejuan Li
- College of Life Science, Sichuan Normal University, Sichuan, China
| | - Shuang Sun
- Shengshi Rongen Bio-tech Co., Ltd., Shanghai, China
| | | | - Long Xiao
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Guoqing Xing
- Nature Industry of Tsukuba Co., Ltd., Ibaraki, Japan
| | - Ran Gao
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Zhenya Zhang
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
62
|
Decara J, Rivera P, López-Gambero AJ, Serrano A, Pavón FJ, Baixeras E, Rodríguez de Fonseca F, Suárez J. Peroxisome Proliferator-Activated Receptors: Experimental Targeting for the Treatment of Inflammatory Bowel Diseases. Front Pharmacol 2020; 11:730. [PMID: 32536865 PMCID: PMC7266982 DOI: 10.3389/fphar.2020.00730] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptor proteins that promote ligand-dependent transcription of target genes that regulate energy production, lipid metabolism, and inflammation. The PPAR superfamily comprises three subtypes, PPARα, PPARγ, and PPARβ/δ, with differential tissue distributions. In addition to their different roles in the regulation of energy balance and carbohydrate and lipid metabolism, an emerging function of PPARs includes normal homeostasis of intestinal tissue. PPARα activation represses NF-κB signaling, which decreases the inflammatory cytokine production by different cell types, while PPARγ ligands can inhibit activation of macrophages and the production of inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and Il-1β. In this regard, the anti-inflammatory responses induced by PPAR activation might restore physiopathological imbalances associated with inflammatory bowel diseases (IBD). Thus, PPARs and their ligands have important therapeutic potential. This review briefly discusses the roles of PPARs in the physiopathology and therapies of the most important IBDs, ulcerative colitis (UC), and Crohn's disease (CD), as well some new experimental compounds with PPAR activity as promising drugs for IBD treatment.
Collapse
Affiliation(s)
- Juan Decara
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Patricia Rivera
- Departamento de Endocrinología, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Antonio Jesús López-Gambero
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Antonia Serrano
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Francisco Javier Pavón
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) and UGC del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Elena Baixeras
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Málaga, IBIMA, Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Juan Suárez
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
63
|
Suyasa IK, Wiradewi Lestari AA. Low expression of vascular endothelial growth factor and high serum level of cyclic guanine monophosphate as the risk factors of femoral head osteonecrosis in alcohol-exposed Wistar rat. Chin J Traumatol 2020; 23:107-112. [PMID: 31980236 PMCID: PMC7156883 DOI: 10.1016/j.cjtee.2019.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Severe damage to the femoral head in patients with osteonecrosis has a high impact on morbidity. Despite early diagnosis, the treatment outcome is still unsatisfactory. This study aimed to explore the expression of vascular endothelial growth factor (VEGF) and cyclic guanine monophosphate (cGMP) serum level as the risk factors of femoral head osteonecrosis in alcohol-exposed Wistar rats. METHODS This was an experimental study using randomized post-test only control group design, with samples using 10-14 weeks Wistar male rats. Rats were then divided into 6 groups: 3 groups without intervention, and 3 groups with intervention using 40% alcohol given perorally. Each one group from intervention and control group was euthanized by the end of the week for 3 consecutive weeks. Proximal femurs were examined under microscope for osteonecrosis, immunohistochemically for VEGF, and blood serum for cGMP levels. RESULTS VEGF expression in the femoral head of alcohol-exposed Wistar rats was lower than those not exposed to alcohol (p < 0.005). Blood serum cGMP levels of alcohol-exposed Wistar rats were higher than those not exposed to alcohol (p < 0.005). The number of necrotic osteocytes in the femoral head of Wistar rats exposed to alcohol was greater than those not exposed to alcohol (p < 0.005). There are significant differences between VEGF, cGMP levels, and number of necrotic osteocytes in the control group and treatment at 1st, 2nd, and 3rd week (p < 0.005). CONCLUSIONS Based on the result of this study, VEGF and cGMP may be considered as diagnostic biomarkers for alcohol-induced femoral head osteonecrosis.
Collapse
Affiliation(s)
- I Ketut Suyasa
- Department of Orthopedic and Traumatology, Faculty of Medicine, Udayana University, Bali, Indonesia
| | - Anak Agung Wiradewi Lestari
- Department of Clinical Pathology, Faculty of Medicine, Udayana University, Bali, Indonesia,Corresponding author.
| |
Collapse
|
64
|
Gliclazide attenuates acetic acid-induced colitis via the modulation of PPARγ, NF-κB and MAPK signaling pathways. Toxicol Appl Pharmacol 2020; 391:114919. [PMID: 32045587 DOI: 10.1016/j.taap.2020.114919] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/11/2022]
Abstract
Ulcerative Colitis is a universal autoimmune disease with high incidence rates worldwide. It is characterized by the existence of many other concurrent immune-associated ailments, including diabetes. The used strategies for the management of this highly costing and complicated disease face great challenges. Therefore, the urge for new medication with fewer side effects and high efficacy is growing. The peroxisome proliferator-activated receptor-gamma (PPARγ) and nuclear factor Kappa-B (NF-κB) can be considered as crucial targets for the treatment of ulcerative colitis. Several studies reported the antioxidants, anti-inflammatory, and antiapoptotic actions of gliclazide and evaluated its cardioprotective and renoprotective effects. However, its impact on ulcerative colitis has never been investigated. This study delineated the effect of gliclazide administration on ulcerative colitis induced by acetic acid in rats and the underlying molecular mechanisms. Gliclazide (10 mg/kg; p.o) prominently decreased colon tissue injury as assessed by the histopathological analysis as well as myeloperoxidase, and intercellular adhesion molecule-1 levels. Gliclazide significantly alleviated the proinflammatory mediator, IL-6, promoted the anti-inflammatory cytokine, IL-10 and, withheld oxidative stress in the injured colon tissues. The protective effect of gliclazide was mediated through the upregulation of PPARγ and downregulation of NF-κB expression. The diminution of ulcerative colitis was also accompanied by an inhibition of the elevated activity and expression of mitogen-activated protein kinases and caspase-3 as assessed by Western blot and immunohistochemistry, respectively. Our findings spotlight, for the first time, the potential of the antidiabetic agent, gliclazide, to attenuate the experimentally induced ulcerative colitis. Therefore, gliclazide might be a propitious agent for the management of ulcerative colitis in diabetic patients.
Collapse
|
65
|
Stettner N, Rosen C, Bernshtein B, Gur-Cohen S, Frug J, Silberman A, Sarver A, Carmel-Neiderman NN, Eilam R, Biton I, Pevsner-Fischer M, Zmora N, Brandis A, Bahar Halpern K, Mazkereth R, di Bernardo D, Brunetti-Pierri N, Premkumar MH, Dank G, Nagamani SCS, Jung S, Harmelin A, Erez A. Induction of Nitric-Oxide Metabolism in Enterocytes Alleviates Colitis and Inflammation-Associated Colon Cancer. Cell Rep 2019; 23:1962-1976. [PMID: 29768197 PMCID: PMC5976577 DOI: 10.1016/j.celrep.2018.04.053] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 02/05/2018] [Accepted: 04/12/2018] [Indexed: 12/30/2022] Open
Abstract
Nitric oxide (NO) plays an established role in numerous physiological and pathological processes, but the specific cellular sources of NO in disease pathogenesis remain unclear, preventing the implementation of NO-related therapy. Argininosuccinate lyase (ASL) is the only enzyme able to produce arginine, the substrate for NO generation by nitric oxide synthase (NOS) isoforms. Here, we generated cell-specific conditional ASL knockout mice in combination with genetic and chemical colitis models. We demonstrate that NO derived from enterocytes alleviates colitis by decreasing macrophage infiltration and tissue damage, whereas immune cell-derived NO is associated with macrophage activation, resulting in increased severity of inflammation. We find that induction of endogenous NO production by enterocytes with supplements that upregulate ASL expression and complement its substrates results in improved epithelial integrity and alleviation of colitis and of inflammation-associated colon cancer.
Collapse
Affiliation(s)
- Noa Stettner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel; Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel; Koret School of Veterinary Medicine, Hebrew University, Rehovot, Israel
| | - Chava Rosen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel; The Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel
| | - Biana Bernshtein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shiri Gur-Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Frug
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alona Sarver
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Inbal Biton
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | - Niv Zmora
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Department of Biological Services, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ram Mazkereth
- The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy; Department of Chemical, Materials and Industrial Engineering, Federico II University, Naples, Italy
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy; Department of Translational Medicine, Federico II University, Naples, Italy
| | - Muralidhar H Premkumar
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Gillian Dank
- Koret School of Veterinary Medicine, Hebrew University, Rehovot, Israel
| | - Sandesh C S Nagamani
- Texas Children's Hospital, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
66
|
Mendes C, Rocha J, Direito R, Fernandes A, Sepodes B, Figueira ME, Ribeiro MH. Anti-inflammatory activity of grapefruit juice in an in vivo model of ulcerative colitis: Comparability studies of unprocessed and bioprocessed juices. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
67
|
Lakshmanan J, Zhang B, Wright K, Motameni AT, Jaganathan V, Schultz DJ, Klinge CM, Harbrecht BG. Tender coconut water suppresses hepatic inflammation by activating AKT and JNK signaling pathways in an in vitro model of sepsis. J Funct Foods 2019; 64. [PMID: 32863888 DOI: 10.1016/j.jff.2019.103637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tender coconut water (TCW) is a natural plant product rich in phytochemicals and protects against toxic liver injury. However, the mechanism by which TCW inhibits inflammation and tissue damage is unknown. We examined the effect of TCW on primary rat hepatocyte viability, cytokine-induced gene expression and proinflammatory signaling in an in vitro model of sepsis. We observed that TCW improved hepatocyte viability and protected hepatocytes against cytokine-mediated cell death. TCW suppressed IL-1β-mediated increases in Nos2, Tnf, and Il6 mRNA and increased heme oxygenase 1 (HMOX1) protein. TCW inhibited iNOS expression through activation of AKT and JNK pathways since inhibition of PI3K and JNK signaling reduced TCW's effect on iNOS protein expression and activity. These results demonstrate that TCW reduces proinflammatory gene expression and hepatocyte injury produced by elevated inflammatory cytokines and nitric oxide production.
Collapse
Affiliation(s)
- Jaganathan Lakshmanan
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, School of Medicine, Louisville, KY 40202
| | - Baochun Zhang
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, School of Medicine, Louisville, KY 40202
| | - Kalen Wright
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, School of Medicine, Louisville, KY 40202
| | - Amierreza T Motameni
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, School of Medicine, Louisville, KY 40202
| | - Vaitheesh Jaganathan
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, School of Medicine, Louisville, KY 40202
| | - David J Schultz
- Department of Biology, University of Louisville, School of Medicine, Louisville, KY 40202
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville, School of Medicine, Louisville, KY 40202
| | - Brian G Harbrecht
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, School of Medicine, Louisville, KY 40202
| |
Collapse
|
68
|
Giménez-Gómez P, Pérez-Hernández M, O'Shea E, Caso JR, Martín-Hernandez D, Cervera LA, Centelles MLGL, Gutiérrez-Lopez MD, Colado MI. Changes in brain kynurenine levels via gut microbiota and gut-barrier disruption induced by chronic ethanol exposure in mice. FASEB J 2019; 33:12900-12914. [PMID: 31509716 DOI: 10.1096/fj.201900491rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammatory processes have been shown to modify tryptophan (Trp) metabolism. Gut microbiota appears to play a significant role in the induction of peripheral and central inflammation. Ethanol (EtOH) exposure alters gut permeability, but its effects on Trp metabolism and the involvement of gut microbiota have not been studied. We analyzed several parameters of gut-barrier and of peripheral and central Trp metabolism following 2 different EtOH consumption patterns in mice, the binge model, drinking in the dark (DID), and the chronic intermittent (CI) consumption paradigm. Antibiotic treatment was used to evaluate gut microbiota involvement in the CI model. Mice exposed to CI EtOH intake, but not DID, show bacterial translocation and increased plasma LPS immediately after EtOH removal. Gut-barrier permeability to FITC-dextran is increased by CI, and, furthermore, intestinal epithelial tight-junction (TJ) disruption is observed (decreased expression of zonula occludens 1 and occludin) associated with increased matrix metalloproteinase (MMP)-9 activity and iNOS expression. CI EtOH, but not DID, increases kynurenine (Kyn) levels in plasma and limbic forebrain. Intestinal bacterial decontamination prevents the LPS increase but not the permeability to FITC-dextran, TJ disruption, or the increase in MMP-9 activity and iNOS expression. Although plasma Kyn levels are not affected by antibiotic treatment, the elevation of Kyn in brain is prevented, pointing to an involvement of microbiota in CI EtOH-induced changes in brain Trp metabolism. Additionally, CI EtOH produces depressive-like symptoms of anhedonia, which are prevented by the antibiotic treatment thus pointing to an association between anhedonia and the increase in brain Kyn and to the involvement of gut microbiota.-Giménez-Gómez, P., Pérez-Hernández, M., O'Shea, E., Caso, J. R., Martín-Hernández, D., Cervera, L. A., Centelles. M. L. G.-L., Gutiérrez-Lopez, M. D., Colado, M. I. Changes in brain kynurenine levels via gut microbiota and gut-barrier disruption induced by chronic ethanol exposure in mice.
Collapse
Affiliation(s)
- Pablo Giménez-Gómez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - David Martín-Hernandez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Salud Mental (CIBERSAM), Madrid, Spain
| | - Luis Alou Cervera
- Área de Microbiología, Departamento de Medicina, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | | | - María Dolores Gutiérrez-Lopez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Maria Isabel Colado
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain.,Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain.,Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
69
|
Organometallic Compounds and Metal Complexes in Current and Future Treatments of Inflammatory Bowel Disease and Colorectal Cancer-a Critical Review. Biomolecules 2019; 9:biom9090398. [PMID: 31443436 PMCID: PMC6770552 DOI: 10.3390/biom9090398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/04/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
In recent years, there has been a significant increase in the clinical use of organometallic compounds and metal complexes for therapeutic purposes including treatment of inflammatory bowel diseases (IBD). Their action is based on the inhibition of the inflow of pro-inflammatory cytokines, the elimination of free radicals or the modulation of intestinal microbiota. In addition, these compounds are intended for use in the diagnosis and treatment of colorectal cancer (CRC) which is often a consequence of IBD. The aim of this study is to critically discuss recent findings on the use of organometallic compounds and metal complexes in the treatment of IBD and CRC and suggest future trends in drug design.
Collapse
|
70
|
Reduction of inflammation and colon injury by a Pennyroyal phenolic extract in experimental inflammatory bowel disease in mice. Biomed Pharmacother 2019; 118:109351. [PMID: 31545244 DOI: 10.1016/j.biopha.2019.109351] [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: 05/01/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Little is known about the pharmacological effects of the phenolic compounds of Pennyroyal (Mentha pulegium). This Mediterranean aromatic plant, used as a gastronomic spice and as food preservative by the food industry has been studied mainly due to its essential oil antibacterial properties, composed primarily by monoterpenes. With this work, we aimed to evaluate the effects of a phenolic extract of pennyroyal in the impairment of inflammatory processes in Inflammatory Bowel Diseases (IBD) and in the potential inhibition of progression to colorectal cancer (CRC). METHODS To that purpose, we evaluated the effect of pennyroyal extract administration in a model of TNBS-induced colitis in mice and further determined its effect on human colon carcinoma cell proliferation and invasion. RESULTS The phenolic extract of pennyroyal exhibited antioxidant properties in in vitro assays and administration of the extract in a rat model of carrageenan-induced paw oedema led to significant anti-inflammatory effects. Further results evidenced a beneficial effect of the phenolic extract in the attenuation of experimental colitis and a potential antiproliferative effect on cultured colon cancer cells, effects not previously described, to our knowledge. A reduction in several markers of colon inflammation was observed following administration of the extract to colitis-induced mice, including functional and histological indicators. A successful inhibition of cancer cell invasion and proliferation was also observed in in vitro studies with HT-29 cells. Furthermore, the extract also led to a reduced expression of iNOS/COX-2 in the colon of colitis-induced mice, both being crucial mediators of intestinal inflammation. CONCLUSIONS Taking into consideration the central role of inflammation in the pathophysiology of CRC and the recognised connection between inflammatory events and cancer, these results enlighten the relevance of the phenolic constituents of pennyroyal as important pharmacological sources in the investigation of new treatment options for patients with inflammatory bowel diseases.
Collapse
|
71
|
Protective effect of methylene blue on TNBS-induced colitis in rats mediated through the modulation of inflammatory and apoptotic signalling pathways. Arch Toxicol 2019; 93:2927-2942. [DOI: 10.1007/s00204-019-02548-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/14/2019] [Indexed: 01/16/2023]
|
72
|
Nitroso-oxidative stress after activation of 5-HT4 receptors under conditions of colitis in rats. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2019. [DOI: 10.2478/cipms-2019-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) plays an important role in the regulation of the functioning of the gastrointestinal tract, including that of the colon. The response of smooth muscles, blood vessels and colon mucosa (CM) to 5-HT is realized through the activation of various types of 5-HT receptors, in particular, 5-HT4 receptors, since the latter are identified on colon cells membranes (enterocytes, smooth muscles and endothelium). The aim of our study was to determine the effect of 5-НT4 receptors agonist (mosapride) on nitrogen (II) oxide production and lipid peroxidation in CM and colon muscle tissue (CMT) under the conditions of experimental ulcerative colitis (UC).
Collapse
|
73
|
Gallic acid protects rat liver mitochondria ex vivo from bisphenol A induced oxidative stress mediated damages. Toxicol Rep 2019; 6:578-589. [PMID: 31293903 PMCID: PMC6595240 DOI: 10.1016/j.toxrep.2019.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 06/08/2019] [Accepted: 06/15/2019] [Indexed: 01/19/2023] Open
Abstract
Bisphenol A induces oxidative stress mediated liver mitochondrial damage. Bisphenol A induced damage is being protected when mitochondria are co-incubated with gallic acid. Scanning electron microscopy of mitochondrial tomography supports the biochemical observations. Gallic acid may be used as future remedial measure for the protection of bisphenol A induced damages of liver mitochondria.
Humans are often exposed to bisphenol A (BPA), the monomer of polycarbonate plastics and epoxy resins, through BPA contaminated drinking water, beverages and foods, packaged in polycarbonate plastic bottles and cans coated with epoxy resins due to leaching. Several research groups have reported that BPA may cause damage of mitochondria in liver, kidney, heart and brain cells by inducing oxidative stress. The antioxidant efficacy of gallic acid (GA), a polyphenol compound obtained from plants, against different toxicants induced oxidative stress has been well established. The aim of the present study was to examine the protective efficacy of GA against BPA induced oxidative damages of the rat liver mitochondria ex vivo. In our study, we have found a significant decrease in the intactness of mitochondria; a significant increase (P ≤ 0.001) in the levels of lipid peroxidation end product (i.e. malondialdehyde) and protein carbonylation product; and also a significant decrease (P ≤ 0.001) in the reduced glutathione content; when mitochondria were incubated with BPA (160 μM/ml) only. These results indicate that BPA probably causes damage to the cellular macromolecules through oxidative stress. We have observed significant counteractions (P ≤ 0.001) against BPA induced alterations in mitochondrial intactness, lipid peroxidation and protein carbonylation products formation and reduced glutathione content when mitochondria were incubated with BPA and GA (20 μg/ml/ 40 μg/ml/ 80 μg/ml) in combination in a dose-dependent manner. Gallic acid also showed significant restorations (P ≤ 0.001) of the activities of antioxidant enzymes, Krebs cycle enzymes, respiratory chain enzymes and thiolase when mitochondria were incubated with BPA and dosage of GA (20 μg/ml/ 40 μg/ml/ 80 μg/ml) in combination compared to BPA incubated mitochondria. Furthermore, GA significantly (P ≤ 0.001) counteracted the BPA induced decrease in tryptophan and NADH auto-fluroscence levels in mitochondria. This result suggests that GA protects the mitochondria probably by reducing the oxidative stress. Besides, GA protects the mitochondrial surface from BPA induced oxidative damages as viewed under the scanning electron microscope. Considering all the results, it can be concluded that GA shows potent efficacy in protecting the rat liver mitochondria ex vivo from BPA induced oxidative stress mediated damages.
Collapse
|
74
|
Direito R, Rocha J, Lima A, Gonçalves MM, Duarte MP, Mateus V, Sousa C, Fernandes A, Pinto R, Boavida Ferreira R, Sepodes B, Figueira ME. Reduction of Inflammation and Colon Injury by a Spearmint Phenolic Extract in Experimental Bowel Disease in Mice. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E65. [PMID: 31174376 PMCID: PMC6630206 DOI: 10.3390/medicines6020065] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
Background: Inflammatory Bowel Diseases (IBD) encompasses both Crohn's Disease and Ulcerative Colitis, known to be connected to an enlarged risk for developing colorectal cancer (CRC). Spearmint (Mentha spicata L.) is a Mediterranean plant used as an aromatic agent, and studies have mainly focused on the essential oil suggesting an anti-inflammatory activity. This work aimed to perform a preliminary screening of the in vivo anti-inflammatory effects of a spearmint phenolic extract in an acute inflammation model, in a chronic inflammation model of colitis, and also study the effects in vitro on a colon cancer model. Methods: Spearmint extract was administered to rats of a paw oedema model (induced by carrageenan) and to mice from a TNBS-induced colitis model in parallel with studies using HT-29 CRC cells. Results: Administration of the extract led to reduced paw inflammation, reduction of colon injury and inflammation, with attenuation of histological markers, and reduction of iNOS expression. It repressed the in vitro movement of HT-29 cells in a wound healing assay. Conclusions: These findings suggest that spearmint extract exhibits acute and chronic anti-inflammatory activity and is able to inhibit migration of cancer cells, suggesting a potential role in the supplementary therapy of IBD patients.
Collapse
Affiliation(s)
- Rosa Direito
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - João Rocha
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Ana Lima
- Disease & Stress Biology Group, LEAF, Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisbon, Portugal.
| | - Maria Margarida Gonçalves
- Unidade de Biotecnologia Ambiental, Universidade Nova de Lisboa, Quinta da Torre, 2829-516 Monte da Caparica, Portugal.
| | - Maria Paula Duarte
- Unidade de Biotecnologia Ambiental (UBiA), Grupo de Disciplinas da Ecologia da Hidrosfera, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal.
| | - Vanessa Mateus
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
- H&TRC-Health and Technology Research Center, ESTeSL-Lisbon School of Health Technology, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal.
| | - Catarina Sousa
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Adelaide Fernandes
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Rui Pinto
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
- Joaquim Chaves Saúde, Dr Joaquim Chaves Lab Analises Clínicas, 1495-068 Miraflores-Algés, Portugal.
| | - Ricardo Boavida Ferreira
- Disease & Stress Biology Group, LEAF, Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisbon, Portugal.
| | - Bruno Sepodes
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Maria-Eduardo Figueira
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| |
Collapse
|
75
|
Burge K, Gunasekaran A, Eckert J, Chaaban H. Curcumin and Intestinal Inflammatory Diseases: Molecular Mechanisms of Protection. Int J Mol Sci 2019; 20:ijms20081912. [PMID: 31003422 PMCID: PMC6514688 DOI: 10.3390/ijms20081912] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Intestinal inflammatory diseases, such as Crohn’s disease, ulcerative colitis, and necrotizing enterocolitis, are becoming increasingly prevalent. While knowledge of the pathogenesis of these related diseases is currently incomplete, each of these conditions is thought to involve a dysfunctional, or overstated, host immunological response to both bacteria and dietary antigens, resulting in unchecked intestinal inflammation and, often, alterations in the intestinal microbiome. This inflammation can result in an impaired intestinal barrier allowing for bacterial translocation, potentially resulting in systemic inflammation and, in severe cases, sepsis. Chronic inflammation of this nature, in the case of inflammatory bowel disease, can even spur cancer growth in the longer-term. Recent research has indicated certain natural products with anti-inflammatory properties, such as curcumin, can help tame the inflammation involved in intestinal inflammatory diseases, thus improving intestinal barrier function, and potentially, clinical outcomes. In this review, we explore the potential therapeutic properties of curcumin on intestinal inflammatory diseases, including its antimicrobial and immunomodulatory properties, as well as its potential to alter the intestinal microbiome. Curcumin may play a significant role in intestinal inflammatory disease treatment in the future, particularly as an adjuvant therapy.
Collapse
Affiliation(s)
- Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Aarthi Gunasekaran
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| | - Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, 1200 North Everett Drive, ETNP7504, Oklahoma City, OK 73104, USA.
| |
Collapse
|
76
|
Xia T, Zhang J, Han L, Jin Z, Wang J, Li X, Man S, Liu C, Gao W. Protective effect of magnolol on oxaliplatin-induced intestinal injury in mice. Phytother Res 2019; 33:1161-1172. [PMID: 30868668 DOI: 10.1002/ptr.6311] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/01/2019] [Accepted: 01/19/2019] [Indexed: 02/06/2023]
Abstract
Oxaliplatin (OXL) is the first line treatment therapy for gastrointestinal (GI) cancers and often combines with other chemotherapy. However, few reports have studied on its GI toxicity. Magnolol (MG), one of the mainly active constituents in Magnolia, has been reported to treat digestive diseases. Therefore, the purpose of this study is to evaluate the intestinal protective effect of MG in OXL treatment group. OXL administration mice showed body weight loss, diarrhea, and intestinal damage characterized by the shortening of villi and destruction of intestinal crypts, as well as the colon length change. MG significantly reduced body weight loss, alleviated diarrhea, reversed histopathological changes, and prevented colon length reduction. Oxidative stress and inflammation were activated after OXL, and these responses were repressed by MG through increasing the activities of superoxide dismutase, glutathione peroxidase, and glutathione, decreasing level of nuclear factor of kappa b and downregulating the following pro-inflammatory cytokines. Although the expression of tight junction protein occludin and numbers of proliferative crypt cells were reduced on ileum and colon after OXL, MG administration promoted these expressions. The fecal gut microbiota composition disturbed by OXL was significantly reversed by MG. Thus, MG could prevent the development and progression of mucositis induced by oxaliplatin through multipathway.
Collapse
Affiliation(s)
- Ting Xia
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jingze Zhang
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Liying Han
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Zhaoxiang Jin
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Juan Wang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xia Li
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
77
|
Roy S, Rizvi ZA, Awasthi A. Metabolic Checkpoints in Differentiation of Helper T Cells in Tissue Inflammation. Front Immunol 2019; 9:3036. [PMID: 30692989 PMCID: PMC6340303 DOI: 10.3389/fimmu.2018.03036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/07/2018] [Indexed: 12/20/2022] Open
Abstract
Naïve CD4+ T cell differentiate into effector and regulatory subsets of helper T (Th) cells in various pathophysiological conditions and modulate tissue inflammation in autoimmune diseases. While cytokines play a key role in determining the fate of Th cells differentiation, metabolites, and metabolic pathways profoundly influence Th cells fate and their functions. Emerging literature suggests that interplay between metabolic pathways and cytokines potentiates T cell differentiation and functions in tissue inflammation in autoimmune diseases. Metabolic pathways, which are essential for the differentiation and functions of Th cell subsets, are regulated by cytokines, nutrients, growth factors, local oxygen levels, co-activation receptors, and metabolites. Dysregulation of metabolic pathways not only alters metabolic regulators in Th cells but also affect the outcome of tissue inflammation in autoimmune and allergic diseases. Understanding the modulation of metabolic pathways during T cells differentiation may potentially lead to a therapeutic strategy for immune-modulation of autoimmune and allergic diseases. In this review, we summarize the role of metabolic checkpoints and their crosstalk with different master transcription factors and signaling molecules in differentiation and function of Th subsets, which may potentially unravel novel therapeutic interventions for tissue inflammation and autoimmune disorders.
Collapse
Affiliation(s)
- Suyasha Roy
- Immuno-Biology Lab, Translational Health Science and Technology Institute, Faridabad, India
| | - Zaigham Abbas Rizvi
- Immuno-Biology Lab, Translational Health Science and Technology Institute, Faridabad, India
| | - Amit Awasthi
- Immuno-Biology Lab, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
78
|
Litvinova EA, Achasova KM, Borisova MA, Zhenilo SV, Prokhortchouk EB, Kozhevnikova EN. Role of the Kaiso gene in the development of inflammation in Mucin-2 defcient mice. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj18.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The number of people with inflammatory bowel disease (IBD) is constantly increasing worldwide. The main factors that have effects on the etiology of the disease are genetic, environmental and immunological. However, the mechanism of disease development and effective treatment of IBD have not yet been found. Animal models help address these problems. The most popular model is considered to be transgenic models in which individual genes are knocked out. One of such models for the study of IBD are mice with a null mutation of theMuc2gene encoding the Mucin-2 protein, which is involved in the formation of a protective mucin layer in the small and large intestine. Some of transcription factors that change the expression of intestinal genes are involved in the development of IBD and colorectal cancer. One of such transcription factors is “zinc fnger” domain-containing protein Kaiso which is able to bind to methylated DNA. In this study, we assessed the role of Kaiso in the development of intestinal inflammation using the experimental model of C57BL/6Muc2-/-Kaiso-/-. We have shown that mice with impaired intestinal barrier function that develop processes similar to human IBD also develop inflammatory responses, such as increased expression ofIl1,TnfandIl17agenes. The defciency of the Kaiso transcription factor in Mucin-2 knockout mice causes a decrease in the expression level of only theCox2andTff3genes. Perhaps a decline in the expression of the gene encoding cyclooxygenase-2 can lead to a decrease in the expression of the antibacterial factor Trefoil factor 3. However, in the experimental model of IBD, Kaiso protein did not play a signifcant role in the regulation of pro-inflammatory cytokines of tumor necrosis factor and interleukins 1 and 17.
Collapse
Affiliation(s)
| | | | | | - S. V. Zhenilo
- Federal Research Centre “Fundamentals of Biotechnology”, RAS, Institute of Bioengineering
| | - E. B. Prokhortchouk
- Federal Research Centre “Fundamentals of Biotechnology”, RAS, Institute of Bioengineering
| | | |
Collapse
|
79
|
Lee S, Kwon HK, Park H, Park Y. Solid–state fermentation of germinated black bean (Rhynchosia nulubilis) using Lactobacillus pentosus SC65 and its immunostimulatory effect. FOOD BIOSCI 2018. [DOI: 10.1016/j.fbio.2018.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
80
|
Nunes C, Almeida L, Barbosa RM, Laranjinha J. Luteolin suppresses the JAK/STAT pathway in a cellular model of intestinal inflammation. Food Funct 2018; 8:387-396. [PMID: 28067377 DOI: 10.1039/c6fo01529h] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Current treatment strategies for inflammatory bowel diseases (IBDs) are associated with a lower efficacy and with several side effects that strongly affect the quality of life of IBD patients. Consequently, the development of new therapies, combining efficacy and safety is an important goal in the field of intestinal inflammation. In this context, evidence supports that polyphenols can be promising candidates due to their ability to modulate intracellular inflammatory signalling cascades. Luteolin, a naturally occurring flavonoid, exhibits anti-inflammatory properties in several models of inflammation. However, its action against intestinal inflammation has been poorly explored. Therefore, there is a lack of scientific knowledge about the potential impact of luteolin in the intestinal inflammation, particularly regarding the underlying molecular mechanisms by which luteolin can exert its anti-inflammatory action. We assessed the potential anti-inflammatory effect of luteolin in a cellular model of intestinal inflammation using cytokine-stimulated HT-29 colon epithelial cells, and the underlying key molecular mechanisms were identified. Luteolin significantly inhibited interleukine-8 (IL-8) production, cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expression and nitric oxide (˙NO) overproduction induced by cytokines, indicating that luteolin negatively modulates key inflammatory signalling cascades underlying intestinal inflammation. Mechanistically, the inhibition of the JAK/STAT pathway was identified as a critical mechanism by which luteolin exerts its intestinal anti-inflammatory action. This study uncovers novel molecular mechanisms by which luteolin may act against intestinal inflammation, which might support the use of luteolin as a future therapeutic strategy in IBD.
Collapse
Affiliation(s)
- Carla Nunes
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Leonor Almeida
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Rui M Barbosa
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - João Laranjinha
- Center for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
81
|
McKay R, Ghodasra M, Schardt J, Quan D, Pottash AE, Shang W, Jay SM, Payne GF, Chang MW, March JC, Bentley WE. A platform of genetically engineered bacteria as vehicles for localized delivery of therapeutics: Toward applications for Crohn's disease. Bioeng Transl Med 2018; 3:209-221. [PMID: 30377661 PMCID: PMC6195910 DOI: 10.1002/btm2.10113] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/14/2022] Open
Abstract
For therapies targeting diseases of the gastrointestinal tract, we and others envision probiotic bacteria that synthesize and excrete biotherapeutics at disease sites. Toward this goal, we have engineered commensal E. coli that selectively synthesize and secrete a model biotherapeutic in the presence of nitric oxide (NO), an intestinal biomarker for Crohn's disease (CD). This is accomplished by co‐expressing the pore forming protein TolAIII with the biologic, granulocyte macrophage‐colony stimulating factor (GM‐CSF). We have additionally engineered these bacteria to accumulate at sites of elevated NO by engineering their motility circuits and controlling pseudotaxis. Importantly, because we have focused on in vitro test beds, motility and biotherapeutics production are spatiotemporally characterized. Together, the targeted recognition, synthesis, and biomolecule delivery comprises a “smart” probiotics platform that may have utility in the treatment of CD. Further, this platform could be modified to accommodate other pursuits by swapping the promoter and therapeutic gene to reflect other disease biomarkers and treatments, respectively.
Collapse
Affiliation(s)
- Ryan McKay
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Institute for Bioscience and Biotechnology Research University of Maryland College Park MD
| | - Monil Ghodasra
- Fischell Dept. of Bioengineering University of Maryland College Park MD
| | - John Schardt
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute National Institutes of Health Bethesda MD
| | - David Quan
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Institute for Bioscience and Biotechnology Research University of Maryland College Park MD
| | - Alex Eli Pottash
- Fischell Dept. of Bioengineering University of Maryland College Park MD
| | - Wu Shang
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Institute for Bioscience and Biotechnology Research University of Maryland College Park MD
| | - Steven M Jay
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute National Institutes of Health Bethesda MD.,Marlene and Stewart Greenebaum Comprehensive Cancer Center University of Maryland School of Medicine Baltimore MD.,Program in Molecular and Cellular Biology University of Maryland College Park MD
| | - Gregory F Payne
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Institute for Bioscience and Biotechnology Research University of Maryland College Park MD
| | - Matthew Wook Chang
- Dept. of Biochemistry, Yong Loo Lin School of Medicine National University of Singapore Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation, Life Sciences Institute National University of Singapore Singapore
| | - John C March
- Dept. of Biological and Environmental Engineering Cornell University Ithaca NY
| | - William E Bentley
- Fischell Dept. of Bioengineering University of Maryland College Park MD.,Institute for Bioscience and Biotechnology Research University of Maryland College Park MD
| |
Collapse
|
82
|
Johnson K, Iyer V, Katzka D, Ravi K, Lennon R, Pendegraft R, Geno D, Alexander J. Poor Relationship Between Fractionated Exhaled Nitric Oxide and Disease Activity in Eosinophilic Esophagitis. Dysphagia 2018; 34:138-144. [PMID: 30043082 DOI: 10.1007/s00455-018-9925-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/11/2018] [Indexed: 12/16/2022]
Abstract
Current eosinophilic esophagitis care requires monitoring with repeat endoscopy and biopsy, which has significant cost, risk, and inconvenience for patients. Fractionated exhaled nitric oxide testing (FeNO) is a standardized non-invasive test with proven utility in evaluation of asthma. Elevated FeNO has reported use in other eosinophilic inflammatory conditions; however, its use in eosinophilic esophagitis has not been fully evaluated. To assess the utility of FeNO in predicting severity of eosinophilic esophagitis activity. Fifty patients received fractionated exhaled nitric oxide testing within 1 week of endoscopic evaluation with biopsy for determination of peak eosinophil counts. Presence of furrows was also evaluated with respect to FeNO levels. Spearman correlation was calculated between FeNO and peak eosinophil counts (PEC) with subgroup analysis performed based on PPI use. Spearman correlation was performed on the change in FeNO and PEC on the patients receiving repeat testing. FeNO was poorly correlated to PEC (Spearman correlation 0.22). With a cut-off FeNO value of > 40 ppb, specificity of FeNO for detecting presence of ≥ 15 eos/hpf was 0.94 and sensitivity was 0.16. FeNO showed weak relationship to presence of furrows. Within the subgroup of patients not taking PPI, the spearman correlation was 0.21. Delta- FeNO versus Delta-PEC had spearman correlation of 0.72 for patients receiving repeat testing. FeNO likely has limited clinical utility for predicting severity of esophageal eosinophilia. In patients with FeNO levels > 40 ppb, specificity of testing was high, but very few patients reached this FeNO level.
Collapse
Affiliation(s)
- Kimberly Johnson
- Internal Medicine, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Vivek Iyer
- Pulmonary and Critical Care Medicine, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - David Katzka
- Gastroenterology and Hepatology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Karthik Ravi
- Gastroenterology and Hepatology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ryan Lennon
- Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Debra Geno
- Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jeffrey Alexander
- Gastroenterology and Hepatology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
83
|
Sproston NR, El Mohtadi M, Slevin M, Gilmore W, Ashworth JJ. The Effect of C-Reactive Protein Isoforms on Nitric Oxide Production by U937 Monocytes/Macrophages. Front Immunol 2018; 9:1500. [PMID: 30013561 PMCID: PMC6036124 DOI: 10.3389/fimmu.2018.01500] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/15/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammation is regulated by many endogenous factors including estrogen, a steroid hormone that declines with increasing age, leading to excessive inflammation in the elderly. C-reactive protein (CRP) is an acute phase inflammatory protein that exists in two forms, native CRP (nCRP) and monomeric CRP (mCRP), which mediate distinct biological activities. It is unclear how each CRP isoform mediates nitric oxide (NO), a signaling molecule generated by NO synthase (NOS). This study investigated whether CRP isoforms have distinct effects on NO production by unstimulated and lipopolysaccharide (LPS)-activated monocytes/macrophages and whether estrogen mediates CRP-induced NO production in an in vitro model of aging. NO and inducible NOS (iNOS) were measured (n = 12) by the Griess assay and an enzyme-linked immunosorbent assay, respectively following incubation (24 h) of human-derived U937 monocytes/macrophages with CRP isoforms [(nCRP) = 500 and 1,000 µg/ml; (mCRP) = 100 and 250 µg/ml] in the absence or presence of 17 beta-estradiol (1 × 10-7, 1 × 10-8, and 1 × 10-9 M). The response to each CRP isoform and estrogen was dependent on the differentiation and activation status of cells. Monocytes with or without prior LPS-activation significantly increased (P < 0.01) NO/iNOS production when treated with mCRP. The mCRP isoform had no effect (P > 0.05) on NO/iNOS production by unactivated or LPS-activated macrophages, whereas nCRP significantly (P < 0.05) reduced NO/iNOS production by macrophages, with or without prior LPS-activation. The nCRP isoform had opposing actions on monocytes, significantly (P < 0.01) increasing and reducing NO/iNOS by unactivated and LPS-activated monocytes, respectively. Estrogen significantly (P < 0.01) reversed nCRP-mediated NO inhibition by unactivated macrophages but decreased CRP-induced NO by unactivated monocytes treated with nCRP or mCRP and LPS-activated monocytes treated with mCRP. NO was differentially mediated by CRP isoforms in a cell-type/state-specific manner, with production corresponding to concomitant changes in iNOS levels. Collectively, the findings indicate nCRP and estrogen predominantly reduce NO production, whereas mCRP increases NO production. This supports growing evidence that mCRP exacerbates inflammation while nCRP and estrogen dampen the overall inflammatory response. Therapeutic strategies that restore estrogen levels to those found in youth and promote the stability of nCRP or/and prevent the formation of mCRP may reduce NO production in age-related inflammatory conditions.
Collapse
Affiliation(s)
| | | | | | | | - Jason J. Ashworth
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
84
|
Rahimi N, Hassanipour M, Allahabadi NS, Sabbaghziarani F, Yazdanparast M, Dehpour A. Cirrhosis induced by bile duct ligation alleviates acetic acid intestinal damages in rats: Involvements of nitrergic and opioidergic systems. Pharmacol Rep 2018; 70:426-433. [DOI: 10.1016/j.pharep.2017.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/24/2017] [Accepted: 11/21/2017] [Indexed: 02/07/2023]
|
85
|
Gupta RA, Motiwala MN, Mahajan UN, Sabre SG. Protective effect of Sesbania grandiflora on acetic acid induced ulcerative colitis in mice by inhibition of TNF-α and IL-6. JOURNAL OF ETHNOPHARMACOLOGY 2018; 219:222-232. [DOI: 10.1016/j.jep.2018.02.043] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
86
|
Han KH, Park JM, Jeong M, Han YM, Go EJ, Park J, Kim H, Han JG, Kwon O, Hahm KB. Heme Oxygenase-1 Induction and Anti-inflammatory Actions of Atractylodes macrocephala and Taraxacum herba Extracts Prevented Colitis and Was More Effective than Sulfasalazine in Preventing Relapse. Gut Liver 2018. [PMID: 28651306 PMCID: PMC5593328 DOI: 10.5009/gnl16496] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background/Aims In inflammatory bowel disease (IBD), repeated bouts of remission and relapse occur in patients and can impose a risk of colitis-associated cancer. We hypothesized that plant extracts of Atractylodes macrocephala (AM) or Taraxacum herba (TH) may be better than sulfasalazine for treating this disease because these extracts can promote additional regeneration. Methods Murine intestinal epithelial IEC-6 cells were pretreated with AM or TH before a lipopolysaccharide (LPS)-induced challenge. Acute colitis was induced with 7 days of dextran sulfate sodium (DSS) in male C57BL/6 mice, and extracts of AM and TH were administered for 2 weeks before DSS administration. Results In vitro studies demonstrated that AM or TH treatment reduced LPS-induced COX-2 and tumor necrosis factor-α mRNA levels but increased heme oxygenase-1 (HO-1). Oral preadministration of AM and TH rescued mice from DSS-induced colitis by inhibiting inflammatory mediators via inactivated extracellular signal regulated kinase and repressed nuclear factor κB and signal transducer and activator of transcription 3, but the effect was weaker for sulfasalazine than that for the extracts. Anti-inflammatory activities occurred via the inhibition of macrophage and T lymphocyte infiltrations. Unlike sulfasalazine, which did not induce HO-1, TH extracts afforded significant HO-1 induction. Conclusions Because the AM or TH extracts were far superior in preventing DSS-induced colitis than sulfasalazine, AM or TH extracts can be considered natural agents that can prevent IBD relapse.
Collapse
Affiliation(s)
- Kyu-Hyun Han
- Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Korea
| | - Jong-Min Park
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Migyeong Jeong
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Young-Min Han
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Eun-Jin Go
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Juyeon Park
- Korea Institute of Science and Technology for Eastern Medicine (KISTEM), NeuMed Inc., Seoul, Korea
| | - Hocheol Kim
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Jae Gab Han
- Department of Health Food Research & Development, Daesang Corp., Icheon, Korea
| | - Oran Kwon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Ki Baik Hahm
- Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Korea.,CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| |
Collapse
|
87
|
Abstract
Inflammatory bowel diseases broadly categorized into Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory disorders of the gastrointestinal tract with increasing prevalence worldwide. The etiology of the disease is complex and involves a combination of genetic, environmental, immunological and gut microbial factors. Recurring and bloody diarrhea is the most prevalent and debilitating symptom in IBD. The pathogenesis of IBD-associated diarrhea is multifactorial and is essentially an outcome of mucosal damage caused by persistent inflammation resulting in dysregulated intestinal ion transport, impaired epithelial barrier function and increased accessibility of the pathogens to the intestinal mucosa. Altered expression and/or function of epithelial ion transporters and channels is the principle cause of electrolyte retention and water accumulation in the intestinal lumen leading to diarrhea in IBD. Aberrant barrier function further contributes to diarrhea via leak-flux mechanism. Mucosal penetration of enteric pathogens promotes dysbiosis and exacerbates the underlying immune system further perpetuating IBD associated-tissue damage and diarrhea. Here, we review the mechanisms of impaired ion transport and loss of epithelial barrier function contributing to diarrhea associated with IBD.
Collapse
Affiliation(s)
- Arivarasu N Anbazhagan
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA
| | - Shubha Priyamvada
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA
| | - Waddah A Alrefai
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA.,b Jesse Brown VA Medical Center , Chicago , IL , USA
| | - Pradeep K Dudeja
- a Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago , IL , USA.,b Jesse Brown VA Medical Center , Chicago , IL , USA
| |
Collapse
|
88
|
Alabi QK, Akomolafe RO, Omole JG, Adefisayo MA, Ogundipe OL, Aturamu A, Sanya JO. Polyphenol-rich extract of Ocimum gratissimum leaves ameliorates colitis via attenuating colonic mucosa injury and regulating pro-inflammatory cytokines production and oxidative stress. Biomed Pharmacother 2018; 103:812-822. [PMID: 29684860 DOI: 10.1016/j.biopha.2018.04.071] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/13/2022] Open
Abstract
Colitis is a chronic inflammation and ulcer on the inner lining of the large intestine. For many centuries Ocimum gratissimum (OG) leaves have been used in folk medicine in Nigeria to treat inflammatory bowel diseases, however, to date, the anti-colitis effects of OG have not been scientifically proven. In this study we investigated the effects of polyphenol rich extract of Ocimum gratissimum (PREOG) leaf on colonic mucosa injury in colitis, its mechanisms, initial administration time and dosage. Dextran sodium sulfate (DSS)-induced rat colitis models was used. PREOG administration was initiated at 3 and 7 d after the model was established at doses of 200, 400 and 800 mg/kg for 7 d. 5-aminosalicylic acid (5-ASA) was used as a reference drug. The disease activity index (DAI), vascular permeability, markers of oxidative stress, granulocyte infiltration, inflammation and histopathological alteration were evaluated. Obvious colonic inflammation and mucosa injuries were observed in DSS-induced colitis groups. PREOG administration promoted repair of colonic mucosa injuries, attenuated inflammation, and decreased DAI scores in rats with colitis. PREOG also decreased the plasma concentrations of Interleukin-(IL)-6 and tumor necrosis factor (TNF)-α, and concentrations of myeloperoxidase, nitric oxide, cyclooxygenase-2 and malondialdehyde in the colon, and increased the plasma concentrations of IL-4 and IL-10 as well as the concentration of superoxide dismutase, catalase and reduced glutathione in the colon. The efficacy of PREOG was dosage dependent. In conclusion, OG repairs colonic mucosa injury in experimental colitis through its ant-inflammatory and ant-oxidant. Its efficacy related to initial administration time and dose.
Collapse
Affiliation(s)
- Quadri K Alabi
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria; Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria.
| | - Rufus O Akomolafe
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Joseph G Omole
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Modinat A Adefisayo
- Department of Physiology, Faculty of Basic Medical Sciences, University of Medical Sciences,Ondo State, Nigeria
| | - Olaofe L Ogundipe
- Department of Public Health and Community Medicine, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria
| | - Ayodeji Aturamu
- Health Center College of Education, Ikere Ekiti, Ekiti State, Nigeria
| | - Joseph O Sanya
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria
| |
Collapse
|
89
|
Rahman SU, Li Y, Huang Y, Zhu L, Feng S, Wu J, Wang X. Treatment of inflammatory bowel disease via green tea polyphenols: possible application and protective approaches. Inflammopharmacology 2018. [DOI: 10.1007/s10787-018-0462-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
90
|
Rossi G, Cerquetella M, Scarpona S, Pengo G, Fettucciari K, Bassotti G, Jergens AE, Suchodolski JS. Effects of probiotic bacteria on mucosal polyamines levels in dogs with IBD and colonic polyps: a preliminary study. Benef Microbes 2018; 9:247-255. [PMID: 29022381 DOI: 10.3920/bm2017.0024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Spermine (SPM) and its precursor putrescine (PUT), regulated by ornithine decarboxylase (ODC) and diamino-oxidase (DAO), are polyamines required for cell growth and proliferation. Only a few studies have investigated the anti-inflammatory and tumour inhibitory properties of probiotics on mucosal polyamine levels. We investigated the effects of a high concentration multistrain probiotic for human use on colonic polyamine biosynthesis in dogs. Histological sections (inflammatory bowel disease, n=10; polyposis, n=5) were assessed after receiving 112 to 225×109 lyophilised bacteria daily for 60 days at baseline (T0) and 30 days after treatment end (T90). Histology scores, expression of PUT, SPM, ODC and DAO, and a clinical activity index (CIBDAI) were compared at T0 and T90. In polyps, cellular proliferation (Ki-67 expression), and apoptosis (caspase-3 protein expression) were also evaluated. After treatment, in inflammatory bowel disease significant decreases were observed for CIBDAI (P=0.006) and histology scores (P<0.001); PUT, SPM and ODC expression increased (P<0.01). In polyps, a significant decrease in polyamine levels, ODC activity, and Ki-67, and a significant increase in caspase-3 positivity and DAO expression (P=0.005) was noted. Our results suggest potential anti-proliferative and anti-inflammatory effects of the probiotic mixture in polyps and inflammation, associated with reduced mucosal infiltration and up-regulation of PUT, SPM, and ODC levels.
Collapse
Affiliation(s)
- G Rossi
- 1 School of Biosciences and Veterinary Medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Macerata (MC), Italy
| | - M Cerquetella
- 1 School of Biosciences and Veterinary Medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Macerata (MC), Italy
| | - S Scarpona
- 1 School of Biosciences and Veterinary Medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Macerata (MC), Italy
| | - G Pengo
- 2 Clinic 'St. Antonio', Strada Statale 415, km 38,50, 26020 Madignano (CR), Italy
| | - K Fettucciari
- 3 Department of Experimental Medicine, University of Perugia School of Medicine, Piazzale Lucio Severi 1-8, 06123 Perugia, Italy
| | - G Bassotti
- 4 Gastroenterology and Hepatology Section, Department of Medicine, University of Perugia School of Medicine, Santa Maria della Misericordia Hospital, Piazzale Menghini 1, 06156 San Sisto, Italy
| | - A E Jergens
- 5 College of Veterinary Medicine, Iowa State University, 1800 Christensen Dr., Ames, IA 50010, USA
| | - J S Suchodolski
- 6 Gastrointestinal Laboratory, Texas A&M University, 4474 TAMU, College Station, TX 77843, USA
| |
Collapse
|
91
|
Mandal P. Molecular signature of nitric oxide on major cancer hallmarks of colorectal carcinoma. Inflammopharmacology 2017; 26:331-336. [PMID: 29289998 DOI: 10.1007/s10787-017-0435-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/11/2017] [Indexed: 11/28/2022]
Abstract
Colorectal cancer (CRC) is the one of the most important diseases throughout the world. Several aetiological risk factors, viz. sedentary life style, smoking, alcohol intake, less physical activity, red meat, and microbiota, are associated with the development of CRC. Molecular pathophysiology of CRC implies inflammation, metastasis, apotosis and angiogenesis. Inflammation involves interaction between various immune cells, inflammatory cells, chemokines, cytokines, and pro-inflammatory mediators, such as cyclooxygenase (COX) and lipoxygenase (LOX) pathways, which may lead to signalling towards, tumour cell proliferation, growth, and invasion whereas nitric oxide (NO) has been associated with metastasis, apoptosis, and angiogenesis. Therefore, this review emphasises on the potential molecular mechanisms associated with NO with alteration of cancer biomarkers during development of colorectal carcinogenesis.
Collapse
Affiliation(s)
- Paramita Mandal
- Department of Zoology, The University of Burdwan, Burdwan, India.
| |
Collapse
|
92
|
Betulinic acid alleviates dextran sulfate sodium-induced colitis and visceral pain in mice. Naunyn Schmiedebergs Arch Pharmacol 2017; 391:285-297. [PMID: 29279966 DOI: 10.1007/s00210-017-1455-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022]
|
93
|
Kim Y, Lim HJ, Jang HJ, Lee S, Jung K, Lee SW, Lee SJ, Rho MC. Portulaca oleracea extracts and their active compounds ameliorate inflammatory bowel diseases in vitro and in vivo by modulating TNF-α, IL-6 and IL-1β signalling. Food Res Int 2017; 106:335-343. [PMID: 29579933 DOI: 10.1016/j.foodres.2017.12.058] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 11/27/2022]
Abstract
Portulaca oleracea L. (P. oleracea) is an herb that is widely used in traditional medicine to treat various diseases. However, its effects on inflammatory diseases, such as inflammatory bowel disease (IBD), are not yet well characterized. Here, we investigated the impact of the ethyl acetate (EtOAc) and ethanol (EtOH) extracts of P. oleracea on lipopolysaccharide (LPS)-induced inflammatory responses and phosphorylation of ERK, JNK, and p38 expression in RAW264.7 macrophages. In addition, the inhibitory effects of these extracts and fractions on 3% dextran sulphate sodium (DSS)-induced ulcerative colitis were examined using an ICR mouse model. DSS-induced colitis, including body weight loss, reduced colon length, and histological colon injury, was significantly ameliorated in mice fed the P. oleracea extracts (200 and 500mg/kg). In particular, P. oleracea extracts also inhibited pro-inflammatory cytokine (TNF-α, IL-6, and 1L-1β) production in mice with DSS-induced colitis; the P. oleracea extracts displayed higher and/or similar inhibitory activity to sulfasalazine at high concentrations. Furthermore, the chemical structures of active compounds separated from the EtOAc extract of P. oleracea were elucidated using nuclear magnetic resonance (NMR) spectroscopy (see Figure in supplementary materials), resulting in the identification of three known compounds. Among these active compounds, cis-N-feruloyl-3'-methoxytyramine (2) exhibited the strongest effects on preventing DSS-induced IBD in animal models. Thus, extract of P. oleracea and their active compounds represents a new therapeutic approach for patients with inflammatory bowel diseases.
Collapse
Affiliation(s)
- Yesol Kim
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea
| | - Hyung Jin Lim
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea; Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Hyun-Jae Jang
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea
| | - Kyungsook Jung
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea
| | - Seung Woong Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea
| | - Seung-Jae Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea.
| | - Mun-Chual Rho
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeonbuk 56212, Republic of Korea.
| |
Collapse
|
94
|
Sikder MOF, Yang S, Ganapathy V, Bhutia YD. The Na+/Cl−-Coupled, Broad-Specific, Amino Acid Transporter SLC6A14 (ATB0,+): Emerging Roles in Multiple Diseases and Therapeutic Potential for Treatment and Diagnosis. AAPS JOURNAL 2017; 20:12. [DOI: 10.1208/s12248-017-0164-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022]
|
95
|
Yang N, Xia Z, Shao N, Li B, Xue L, Peng Y, Zhi F, Yang Y. Carnosic acid prevents dextran sulfate sodium-induced acute colitis associated with the regulation of the Keap1/Nrf2 pathway. Sci Rep 2017; 7:11036. [PMID: 28887507 PMCID: PMC5591263 DOI: 10.1038/s41598-017-11408-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/22/2017] [Indexed: 12/20/2022] Open
Abstract
Crohn's disease and ulcerative colitis are inflammatory bowel diseases (IBDs) with high prevalence in humans. Carnosic acid (CA) has been reported to possess antioxidative properties; however, its role in IBDs has not been determined. In the present study, we found that CA significantly prevented the loss of body weight and shortening of colon length in acute colitis induced by dextran sodium sulfate (DSS). Pronounced infiltration of immune cells and a loss of crypt architecture and goblet cells were ameliorated by CA. CA significantly decreased the activity of MPO and infiltration of F4/80+ macrophages in the colon. DSS-induced pro-inflammatory cytokine mRNA and protein levels in the colon were also attenuated by CA. CA decreased the activation of p65 and c-Jun signalling. CA inhibited DSS-induced NLRP3 inflammasome activation by reducing caspase 1 activity. In addition, CA increased the level of Nrf2 and prevented the degradation of Nrf2 via ubiquitination by blocking the interaction between Cullin3 and Keap1, which resulted in the decrease of Nrf2 target genes. Finally, GSH levels and SOD activity were increased after CA treatment, while MDA and iNOS levels were significantly reduced. Taken together, our data showed that CA may be useful as a potential therapeutic candidate for IBDs.
Collapse
Affiliation(s)
- Neng Yang
- Office of Drug Clinical Trial Institution, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China.,Department of Pharmacy, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Zongling Xia
- Department of Pharmacy, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Naiyuan Shao
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Bowen Li
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Lian Xue
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ya Peng
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China.
| | - Feng Zhi
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China. .,Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| | - Yilin Yang
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China. .,Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
96
|
Odukanmi OA, Salami AT, Ashaolu OP, Adegoke AG, Olaleye SB. Kolaviron attenuates ischemia/reperfusion injury in the stomach of rats. Appl Physiol Nutr Metab 2017; 43:30-37. [PMID: 28841395 DOI: 10.1139/apnm-2017-0138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Kolaviron (KV), an active complex of at least 3 compounds in Garcinia kola seed, which is known for its antioxidant and anti-inflammatory activity, was investigated for its gastro-protective effect in the stomach of rats subjected to ischemia/reperfusion-induced gastric ulceration. Male adult Wistar rats (180-210 g) were randomized into 6 groups (n = 15) as follows: (i) control, (ii) ulcerated untreated (UU), (iii) KV alone (KVA), (iv) KV + ulcer (KVU), (v) ulcer + KV (UKV), and (vi) ulcer + omeprazole (20 mg/kg). Ulcer was induced through ischemia/reperfusion method after 2 weeks of daily oral KV (100 mg/kg). Rats were weighed daily, and gastric acid secretion, ulcer scores, hematological, biochemical, and histological variables were assessed 1 h after induction at 3 and 7 days post-ulceration. Body weight decreased in KVA (179.1 ± 1.6 g), and KVU (170.1 ± 2.2 g) compared with UU (199.0 ± 1.4 g). Gastric acid secretion decreased significantly in KVU after 1 h and 3 days post-ulceration (0.27 ± 0.03 mEq/L; 0.49 ± 0.02 mEq/L) compared with UU (0.60 ± 0.06 mEq/L; 0.85 ± 0.29 mEq/L), respectively. There was significant reduction in neutrophil/lymphocyte ratio of KVA (0.29 ± 0.06) and KVU (0.35 ± 0.02) compared with UU (0.54 ± 0.04). Malondialdehyde level decreased significantly with concomitant increase in anti-oxidative activities and nitric oxide level in the KV treated groups (KVA, KVU, UKV) compared with UU. In conclusion, treatment with KV protects the stomach by reducing gastric acid secretion, promoting antioxidant activity and suppressing action of reactive oxygen species.
Collapse
Affiliation(s)
- Olugbenga Adeola Odukanmi
- Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeola Temitope Salami
- Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Onaara Peter Ashaolu
- Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeoti Gbemisola Adegoke
- Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Samuel Babafemi Olaleye
- Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Laboratory for Gastrointestinal Secretions and Inflammation Research, Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
97
|
Schreiber D, Marx L, Felix S, Clasohm J, Weyland M, Schäfer M, Klotz M, Lilischkis R, Erkel G, Schäfer KH. Anti-inflammatory Effects of Fungal Metabolites in Mouse Intestine as Revealed by In vitro Models. Front Physiol 2017; 8:566. [PMID: 28824460 PMCID: PMC5545603 DOI: 10.3389/fphys.2017.00566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023] Open
Abstract
Inflammatory bowel diseases (IBD), which include Crohn's disease and ulcerative colitis, are chronic inflammatory disorders that can affect the whole gastrointestinal tract or the colonic mucosal layer. Current therapies aiming to suppress the exaggerated immune response in IBD largely rely on compounds with non-satisfying effects or side-effects. Therefore, new therapeutical options are needed. In the present study, we investigated the anti-inflammatory effects of the fungal metabolites, galiellalactone, and dehydrocurvularin in both an in vitro intestinal inflammation model, as well as in isolated myenteric plexus and enterocyte cells. Administration of a pro-inflammatory cytokine mix through the mesenteric artery of intestinal segments caused an up-regulation of inflammatory marker genes. Treatment of the murine intestinal segments with galiellalactone or dehydrocurvularin by application through the mesenteric artery significantly prevented the expression of pro-inflammatory marker genes on the mRNA and the protein level. Comparable to the results in the perfused intestine model, treatment of primary enteric nervous system (ENS) cells from the murine intestine with the fungal compounds reduced expression of cytokines such as IL-6, TNF-α, IL-1β, and inflammatory enzymes such as COX-2 and iNOS on mRNA and protein levels. Similar anti-inflammatory effects of the fungal metabolites were observed in the human colorectal adenocarcinoma cell line DLD-1 after stimulation with IFN-γ (10 ng/ml), TNF-α (10 ng/ml), and IL-1β (5 ng/ml). Our results show that the mesenterially perfused intestine model provides a reliable tool for the screening of new therapeutics with limited amounts of test compounds. Furthermore, we could characterize the anti-inflammatory effects of two novel active compounds, galiellalactone, and dehydrocurvularin which are interesting candidates for studies with chronic animal models of IBD.
Collapse
Affiliation(s)
- Dominik Schreiber
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany.,Department of Biotechnology, Technical University of KaiserslauternKaiserslautern, Germany
| | - Lisa Marx
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Silke Felix
- Department of Biotechnology, Technical University of KaiserslauternKaiserslautern, Germany
| | - Jasmin Clasohm
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Maximilian Weyland
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Maximilian Schäfer
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Markus Klotz
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Rainer Lilischkis
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany
| | - Gerhard Erkel
- Department of Biotechnology, Technical University of KaiserslauternKaiserslautern, Germany
| | - Karl-Herbert Schäfer
- Department of Biotechnology, University of Applied Sciences KaiserslauternKaiserslautern, Germany.,Pediatric Surgery, University Hospital MannheimMannheim, Germany
| |
Collapse
|
98
|
El-Ashmawy NE, Khedr NF, El-Bahrawy HA, El-Adawy SA. Downregulation of iNOS and elevation of cAMP mediate the anti-inflammatory effect of glabridin in rats with ulcerative colitis. Inflammopharmacology 2017; 26:551-559. [PMID: 28707183 DOI: 10.1007/s10787-017-0373-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/01/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alternative medicine is widely accepted by public and becoming an attractive approach for treatment of various diseases. Glabridin (Gla), a major flavonoid present in licorice root, was reported to have antioxidant and anti-inflammatory properties. OBJECTIVE The study aimed to investigate the possible protective role of Gla against dextran sulphate sodium (DSS)-induced ulcerative colitis (UC) in rats and to clarify the molecular mechanisms underlying Gla function. METHODS Forty male Wistar rats were divided into control, colitis group (rats received 5% DSS in drinking water for 7 days), Gla group (50 mg/kg, orally, once daily), and sulfasalazine (SLZ) group (500 mg/kg, orally, once daily). Each of Gla and SLZ was administered 1 week ahead of DSS and parallel with its administration. RESULTS Gla ameliorated the inflammatory alterations induced by DSS. Gla group showed a reduction in colon concentration of tumor necrosis factor-alpha (TNF-α) and a decreased colon myeloperoxidase activity (MPO). Gla treatment downregulated inducible nitric oxide synthase (iNOS) gene expression in rat colon with a decreased content of nitric oxide (NO). Gla also increased cyclic AMP (cAMP) concentration in rat colon compared to colitis group. Such findings were comparable to or even better than those obtained by SLZ treatment. The histological features of UC such as ulceration and inflammatory cell infiltrations were improved in rat group treated by Gla. CONCLUSION Gla proved a potent anti-inflammatory role in UC through different mechanisms and, being a natural product, it could be safely used as a protective measure in inflammatory bowel diseases.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Biochemistry Department, Faculty of Pharmacy, Tanta University, El-Bahr Street, Tanta, El-Gharbia, 31527, Egypt
| | - Naglaa F Khedr
- Biochemistry Department, Faculty of Pharmacy, Tanta University, El-Bahr Street, Tanta, El-Gharbia, 31527, Egypt
| | - Hoda A El-Bahrawy
- Biochemistry Department, Faculty of Pharmacy, Tanta University, El-Bahr Street, Tanta, El-Gharbia, 31527, Egypt
| | - Samar A El-Adawy
- Biochemistry Department, Faculty of Pharmacy, Tanta University, El-Bahr Street, Tanta, El-Gharbia, 31527, Egypt.
| |
Collapse
|
99
|
Inducible Nitric Oxide Synthase Polymorphisms and Nitric Oxide Levels in Individuals with Chronic Periodontitis. Int J Mol Sci 2017; 18:ijms18061128. [PMID: 28617311 PMCID: PMC5485952 DOI: 10.3390/ijms18061128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/17/2017] [Accepted: 05/20/2017] [Indexed: 12/27/2022] Open
Abstract
This study aimed to investigate whether the -1026(A>C)(rs2779249) and +2087(A>G)(2297518) polymorphisms in the NOS2 gene were associated with chronic periodontitis (CP) and with salivary levels of nitrite (NO₂-) and/or nitrate + nitrite (NOx). A group of 113 mixed-race patients were subjected to periodontal, genetic, and biochemical evaluations (65 CP/48 periodontally healthy subjects). DNA was extracted from oral epithelial cells and used for genotyping by polymerase chain reaction (real-time). Salivary NOx concentrations were determined using an ozone-based chemiluminescence assay. Association of CP with alleles and genotypes of the -1026(A>C) polymorphism was found (X² test, p = 0.0075; 0.0308), but this was not maintained after multiple logistic regression, performed to estimate the effect of covariates and polymorphisms in CP. This analysis demonstrated, after correction for multiple comparisons, that only the female gender was significantly associated with CP. Polymorphisms analyzed as haplotypes were not associated with CP. NOx levels were significantly higher in the control group of heterozygous individuals for both polymorphisms. In conclusion, the female gender was significantly associated with CP, and higher levels of salivary NOx were found in control subjects and associated with the heterozygous state of the NOS2 polymorphisms, reinforcing the potential of NO metabolites as markers of periodontitis status.
Collapse
|
100
|
Mirmiran P, Ejtahed HS, Angoorani P, Eslami F, Azizi F. Camel Milk Has Beneficial Effects on Diabetes Mellitus: A Systematic Review. Int J Endocrinol Metab 2017; 15:e42150. [PMID: 29026408 PMCID: PMC5626114 DOI: 10.5812/ijem.42150] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/21/2017] [Accepted: 02/22/2017] [Indexed: 11/16/2022] Open
Abstract
CONTEXT Controlling diabetes, a worldwide metabolic disease, by effective alternative treatments is currently a topic of great interest. Camel milk is believed to be a suitable hypoglycemic agent in experimental animals and patients with diabetes. The current systematic review aimed at evaluating the effect of camel milk on diabetes. EVIDENCE ACQUISITION A comprehensive search was dine in PubMed and Scopus for all clinical trials and animal studies documented up to 2015, which focused on the effect of camel milk on diabetes markers. Studies which assessed the effects of camel milk, with no dose limit, on glucose parameters and lipid profiles in animals or humans with diabetes, were included. The quality of the included clinical trials was evaluated by the Delphi score checklist. RESULTS The initial search yielded 73 articles. After screening abstracts and full texts, 22 articles were included consisting of 11 animal studies and 11 clinical trials, 8 of which focused on type 1 diabetes and the other three on type 2diabetes. All animal studies except for 1 showed significant reductions in at least 1 of the diabetes parameters such as blood glucose, insulin resistance, glycated hemoglobin, and lipid profile. In most of the clinical trials, the recommended dose of camel milk was 500 mL/day, which led to improvement of diabetes markers even after 3 months in patients with diabetes. CONCLUSIONS Most of the studies in the current systematic review demonstrated the favorable effects of camel milk on diabetes mellitus by reducing blood sugar, decreasing insulin resistance and improving lipid profiles.
Collapse
Affiliation(s)
- Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, IR Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Pooneh Angoorani
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Fariba Eslami
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Fereidoun Azizi, Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box: 19395-4763, Tehran, IR Iran. Tel: +98-2122409309, Fax: +98-2122402463, E-mail:
| |
Collapse
|