201
|
Xu B, Zheng J, Tian X, Yuan F, Liu Z, Zhou Y, Yang Z, Ding X. Protective mechanism of traditional Chinese medicine guizhi fuling pills against carbon tetrachloride-induced kidney damage is through inhibiting oxidative stress, inflammation and regulating the intestinal flora. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154129. [PMID: 35490491 DOI: 10.1016/j.phymed.2022.154129] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/09/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Chemical or drug-induced kidney damage has been recognized as a critical cause of kidney failure. The oxidative stress, inflammation, and imbalance of intestinal flora caused by carbon tetrachloride (CCl4) play a fundamental role in chronic kidney damage. Guizhi Fuling pills (GZFL) is a traditional formula consisting of five traditional Chinese medicinal herbs, which can promote blood circulation and improve kidney function. The underlying mechanisms of GZFL improving kidney damage are not fully understood yet. AIM The current study aimed to explore the effects of GZFL on CCl4-induced kidney damage and intestinal microbiota in mice. METHODS Male ICR mice were intraperitoneally administered with 20% CCl4 (mixed in a ratio of 1:4 in soybean oil) twice a week, for 4 weeks to induce kidney damage. Creatinine (CRE), urea nitrogen, antioxidant enzymes, and inflammatory cytokines were measured and the histology of the kidney, jejunum, and colon examination to assess kidney and intestinal damage. The expressions of nuclear factor-erythroid 2-related factor 2 (Nrf2) family members, nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome in kidney tissues, and the tight junction proteins in colonic tissues were detected by Western blot. The gut microbiota was analyzed through 16S rRNA gene sequencing. RESULTS GZFL treatment decreased the serum CRE and urea nitrogen levels. Moreover, GZFL reduced the levels of pro-inflammatory cytokines and increased antioxidant enzyme activities in kidney and colonic tissues. GZFL improved the kidney, jejunum, and colon histology. Furthermore, GZFL inhibited the expressions of NLRP3, ASC, and cleaved-Caspase-1, while Nrf2, HO-1, NQO1, GCLM, and tight junction proteins were increased. The dysbiosis of intestinal microbiota improved after GZFL treatment. CONCLUSIONS This study showed that GZFL could improve kidney damage, which might be mainly via the integrated regulations of the Nrf2 pathway, NLRP3 inflammasome, and composition of intestinal microbiota.
Collapse
Affiliation(s)
- Baogui Xu
- School of Food and Pharmacy, Zhejiang Ocean University; Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan 316022, China
| | - Jiawen Zheng
- School of Food and Pharmacy, Zhejiang Ocean University; Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan 316022, China
| | - Xiaoxiao Tian
- School of Food and Pharmacy, Zhejiang Ocean University; Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan 316022, China
| | - Falei Yuan
- School of Food and Pharmacy, Zhejiang Ocean University; Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan 316022, China
| | - Zhongliang Liu
- Zhoushan Hospital of Traditional Chinese Medicine, Zhoushan 316000, China
| | - Yafeng Zhou
- School of Food and Pharmacy, Zhejiang Ocean University; Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan 316022, China
| | - Zuisu Yang
- School of Food and Pharmacy, Zhejiang Ocean University; Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhoushan 316022, China.
| | - Xianjun Ding
- Zhoushan Hospital of Traditional Chinese Medicine, Zhoushan 316000, China.
| |
Collapse
|
202
|
Zhang T, Wang J, Yao Z, Ni L, Zhao Y, Wei S, Chen Z. Effect and Mechanism of Bifidobacterium animalis B94 in the Prevention and Treatment of Liver Injury in Rats. Front Cell Infect Microbiol 2022; 12:914684. [PMID: 35846768 PMCID: PMC9277360 DOI: 10.3389/fcimb.2022.914684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To investigate the effect of Bifidobacterium animalis B94 on the prevention and treatment of liver injury in rats and to elucidate the underlying mechanism of this relationship. Methods Specific pathogen-free (SPF) rats were selected as the healthy control group, liver injury group and B94 treatment group, with 6 rats in each group. After the model was established, the experimental animals were tested for serum liver function indicators, gut microbiota composition, metabolite composition, and histopathology. Results The albumin/globulin ratio and serum TBA, alanine aminotransferase, aspartate aminotransferase, and indirect bilirubin levels in the B94 treatment group were significantly lower than those in the liver injury group. 16S rRNA analysis showed that the gut microbiota of the three groups of rats were significantly different. Metabolic profile analysis showed that there were significant differences in the gut metabolomes of the three groups. Haematoxylin–eosin staining of the intestinal mucosa and liver tissues showed that the degree of liver and intestinal tissue damage in the B94 treatment group was significantly lower than that in the liver injury group. Conclusion Bifidobacterium animalis B94 can affect the process of liver injury in rats by improving liver function, reducing intestinal damage, and regulating gut microbiota and metabolite production.
Collapse
Affiliation(s)
- Tianfang Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Wang
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhao Yao
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lingmei Ni
- Infection Prevention and Control Department, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yifan Zhao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuang Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zuobing Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zuobing Chen,
| |
Collapse
|
203
|
Kuang ZS, Leng YX, Yang N, Li ZQ, Zong YN, Han DY, Li Y, He JD, Mi XN, Cong ZK, Zhu X, Wu CY, Guo XY. Inhibition of visfatin alleviates sepsis-induced intestinal damage by inhibiting Hippo signaling pathway. Inflamm Res 2022; 71:911-922. [PMID: 35731253 PMCID: PMC9307557 DOI: 10.1007/s00011-022-01593-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 01/11/2023] Open
Abstract
Background The aim of this study is to investigate role of Visfatin, one of the pro-inflammatory adipokines, in sepsis-induced intestinal injury and to clarify the potential mechanism. Methods C57BL/6 mice underwent cecal ligation and puncture (CLP) surgery to establish sepsis model in vivo. Intestinal epithelial cells were stimulated with LPS to mimic sepsis-induced intestinal injury in vitro. FK866 (the inhibitor of Visfatin) with or without XMU-MP-1 (the inhibitor of Hippo signaling) was applied for treatment. The expression levels of Visfatin, NF-κB and Hippo signaling pathways-related proteins were detected by western blot or immunohistochemistry. The intestinal cell apoptosis and intestinal injury were investigated by TUNEL staining and H&E staining, respectively. ELISA was used to determine the production of inflammatory cytokines. Results The expression of Visfatin increased in CLP mice. FK866 reduced intestinal pathological injury, inflammatory cytokines production, and intestinal cell apoptosis in sepsis mice. Meanwhile, FK866 affected NF-κB and Hippo signaling pathways. Additionally, the effects of FK866 on inflammatory response, apoptosis, Hippo signaling and NF-κB signaling were partly abolished by XMU-MP-1, the inhibitor of Hippo signaling. In vitro experiments also revealed that FK866 exhibited a protective role against LPS-induced inflammatory response and apoptosis in intestinal cells, as well as regulating NF-κB and Hippo signaling, whereas addition of XMU-MP-1 weakened the protective effects of FK866. Conclusion In short, this study demonstrated that inhibition of Visfatin might alleviate sepsis-induced intestinal injury through Hippo signaling pathway, supporting a further research on Visfatin as a therapeutic target.
Collapse
Affiliation(s)
- Zhong-Shen Kuang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | - Yu-Xin Leng
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | - Zheng-Qian Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | - Ya-Nan Zong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | | | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | - Jin-Dan He
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | - Xing-Ning Mi
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China
| | - Zhu-Kai Cong
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Xi Zhu
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Chang-Yi Wu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China.
| | - Xiang-Yang Guo
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing, 100191, China.
| |
Collapse
|
204
|
Putranto AS, Suyatna FD, Soetikno V, Moenadjat Y. Novel and simple method using cable ties to induce intestinal strangulation in a rat model. MEDICAL JOURNAL OF INDONESIA 2022. [DOI: 10.13181/mji.oa.225799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The pathogenesis of intestinal fibrosis after strangulation in abdominal surgery is not fully understood. Developing an experimental and reliable method is needed to overcome this problem. This study aimed to develop an effective method for intestinal strangulation leading to fibrosis, which might induce intestinal obstruction.
METHODS This study was conducted from 2018 to 2019 at the Department of Pharmacology, Faculty of Medicine, Universitas Indonesia. A total of 24 Sprague Dawley rats were used in this study. Artificial intestinal strangulation using cable ties made of durable plastic, which is generally used to ligate objects, was applied, and rats were further classified into control, 6-hour, and 24-hour strangulation groups. At the end of the study, the rats were sacrificed, intestinal tissues were collected, and histomorphological changes were observed using hematoxylin and eosin stain. Moreover, Masson’s trichome staining was used to assess collagen density.
RESULTS Median collagen density score of the 24-hour strangulation group was higher than the control. A significant difference in collagen density was found between the submucosal layer of the 24-hour strangulation group and the control (45.4 [11.4] versus 32.4 [14.0], p<0.001) and between the mucosa of the 6- and 24-hour strangulation groups and the control (26.9 [17] versus 6.46 [4.3], p = 0.01 and 24.9 [8] versus 6.46 [4.3], p = 0.004, respectively).
CONCLUSIONS The simple use of cable ties adequately promotes intestinal strangulation.
Collapse
|
205
|
Effect of TRAF6 in acute pancreatitis-induced intestinal barrier injury via TLR4/NF-κB signal pathway. Tissue Cell 2022; 76:101792. [DOI: 10.1016/j.tice.2022.101792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/28/2022]
|
206
|
Smoday IM, Petrovic I, Kalogjera L, Vranes H, Zizek H, Krezic I, Gojkovic S, Skorak I, Hriberski K, Brizic I, Kubat M, Strbe S, Barisic I, Sola M, Lovric E, Lozic M, Boban Blagaic A, Skrtic A, Seiwerth S, Sikiric P. Therapy Effect of the Stable Gastric Pentadecapeptide BPC 157 on Acute Pancreatitis as Vascular Failure-Induced Severe Peripheral and Central Syndrome in Rats. Biomedicines 2022; 10:1299. [PMID: 35740321 PMCID: PMC9220115 DOI: 10.3390/biomedicines10061299] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 12/15/2022] Open
Abstract
We revealed the therapy effect of the stable gastric pentadecapeptide BPC 157 (10 μg/kg, 10 ng/kg ig or po) with specific activation of the collateral rescuing pathways, the azygos vein, on bile duct ligation in particular, and acute pancreatitis as local disturbances (i.e., improved gross and microscopy presentation, decreased amylase level). Additionally, we revealed the therapy's effect on the acute pancreatitis as vascular failure and multiorgan failure, both peripherally and centrally following "occlusion-like" syndrome, major intoxication (alcohol, lithium), maintained severe intra-abdominal hypertension, and myocardial infarction, or occlusion syndrome, and major vessel occlusion. The application-sacrifice periods were ligation times of 0-30 min, 0-5 h, 0-24 h (cured periods, early regimen) and 4.30 h-5 h, 5 h-24 h (cured periods, delayed regimen). Otherwise, bile duct-ligated rats commonly presented intracranial (superior sagittal sinus), portal and caval hypertension and aortal hypotension, gross brain swelling, hemorrhage and lesions, heart dysfunction, lung lesions, liver and kidney failure, gastrointestinal lesions, and severe arterial and venous thrombosis, peripherally and centrally. Unless antagonized with the key effect of BPC 157 regimens, reversal of the inferior caval and superior mesenteric vein congestion and reversal of the failed azygos vein activated azygos vein-recruited direct delivery to rescue the inferior-superior caval vein pathway; these were all antecedent to acute pancreatitis major lesions (i.e., acinar, fat necrosis, hemorrhage). These lesions appeared in the later period, but were markedly attenuated/eliminated (i.e., hemorrhage) in BPC 157-treated rats. To summarize, while the innate vicious cycle may be peripheral (bile duct ligation), or central (rapidly developed brain disturbances), or peripheral and central, BPC 157 resolved acute pancreatitis and its adjacent syndrome.
Collapse
Affiliation(s)
- Ivan Maria Smoday
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Igor Petrovic
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Luka Kalogjera
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Hrvoje Vranes
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Helena Zizek
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Slaven Gojkovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Ivan Skorak
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Klaudija Hriberski
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Ivan Brizic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Milovan Kubat
- Department of Forensic Medicine and Criminology, School of Medicne, 10000 Zagreb, Croatia;
| | - Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Ivan Barisic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Marija Sola
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Eva Lovric
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (S.S.)
| | - Marin Lozic
- Department of Pediatric and Preventive Dentistry, School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| | - Anita Skrtic
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (S.S.)
| | - Sven Seiwerth
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (S.S.)
| | - Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.M.S.); (L.K.); (H.V.); (H.Z.); (I.K.); (S.G.); (I.S.); (K.H.); (I.B.); (S.S.); (I.B.); (M.S.); (A.B.B.)
| |
Collapse
|
207
|
Zhou Y, Liu J, Li X, Wang L, Hu L, Li A, Zhou J. JAC4 Protects from X-Ray Radiation-Induced Intestinal Injury by JWA-Mediated Anti-Oxidation/Inflammation Signaling. Antioxidants (Basel) 2022; 11:antiox11061067. [PMID: 35739964 PMCID: PMC9220415 DOI: 10.3390/antiox11061067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation-induced enteritis in the clinic. We synthesized a compound, named JAC4, which is an agonist and can increase JWA protein expression. JWA has been shown to reduce oxidative stress, DNA damage, anti-apoptosis, and anti-inflammatory; in addition, the small intestine epithelium showed dysplasia in JWA knockout mice. We hypothesized that JAC4 might exert a protective effect against radiation-induced intestinal damage. Herein, X-ray radiation models were built both in mice and in intestinal crypt epithelial cells (IEC-6). C57BL/6J mice were treated with JAC4 by gavage before abdominal irradiation (ABI); the data showed that JAC4 significantly reduced radiation-induced intestinal mucosal damage and increased the survival rate. In addition, radiation-induced oxidative stress damage and systemic inflammatory response were also mitigated by JAC4 treatment. Moreover, JAC4 treatment alleviated DNA damage, decreased cell apoptosis, and maintained intestinal epithelial cell proliferation in mice. In vitro data showed that JAC4 treatment significantly inhibited ROS formation and cell apoptosis. Importantly, all the above protective effects of JAC4 on X-ray radiation-triggered intestinal injury were no longer determined in the intestinal epithelium of JWA knockout mice. Therefore, our results provide the first evidence that JAC4 protects the intestine from radiation-induced enteritis through JWA-mediated anti-oxidation/inflammation signaling.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jingwen Liu
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Xiong Li
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Luman Wang
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Lirong Hu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China;
| | - Aiping Li
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jianwei Zhou
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
- Correspondence:
| |
Collapse
|
208
|
Yang B, Huang S, Li S, Feng Z, Zhao G, Ma Q. Safety Evaluation of Porcine Bile Acids in Laying Hens: Effects on Laying Performance, Egg Quality, Blood Parameters, Organ Indexes, and Intestinal Development. Front Vet Sci 2022; 9:895831. [PMID: 35685343 PMCID: PMC9171047 DOI: 10.3389/fvets.2022.895831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 01/14/2023] Open
Abstract
Bile acids (BAs) have long been known to facilitate digestion, transport, and absorption of lipids in the small intestine as well as regulate host lipid metabolic homeostasis. However, excessive BAs may lead to long-term damage to tissue. Also, it is unknown whether different levels of porcine BAs supplementation could improve performance, host metabolism, intestinal functions in laying hens. Hence, this study was aimed to investigate the potential effects of BAs addition on laying performance, egg quality, blood parameters, organ indexes, and intestinal histopathology of hens in the late phase. A total of 300 58-week-old Hy-line Gray hens were randomly divided into five groups which fed a basal diet (control) or basal diets supplemented with 60, 300, 600, and 3,000 mg/kg BAs for 56 days. Compared with the control group, no significant differences (P > 0.05) were observed in egg production, egg weight, ADFI, and FCR of hens in 60, 300, 600, and 3,000 mg/kg BAs groups. Dietary 60 mg/kg BAs supplementation resulted in a significant increase (P < 0.05) in egg mass. Meanwhile, no significant differences were observed in egg quality, including eggshell strength, eggshell thickness, albumen height, and Haugh unit among any treatment groups (P > 0.05). Dramatically, dietary 3,000 mg/kg BAs supplement decreased yolk color (P < 0.05). There was no significant difference in the blood parameters such as WBC, RBC, HGB, HCT, and PLT among any treatments. However, in 3,000 mg/kg BAs group, ovary coefficient was lower than the control (P < 0.05), and serum urea and creatinine were higher than the control (P < 0.05). Also, kidney and oviduct injury appeared in 3,000 mg/kg BAs group. These results indicated that a porcine BAs concentration of 3,000 mg/kg may cause harmful effects while 600 mg/kg was non-deleterious to laying hens after a daily administration for 56 days, namely that dietary supplement of up to 10 times the recommended dose of BAs was safely tolerated by laying hens.
Collapse
Affiliation(s)
- Bowen Yang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shupeng Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Zhihua Feng
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Guoxian Zhao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
- *Correspondence: Guoxian Zhao
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Qiugang Ma
| |
Collapse
|
209
|
Schaaf CR, Gonzalez LM. Use of Translational, Genetically Modified Porcine Models to Ultimately Improve Intestinal Disease Treatment. Front Vet Sci 2022; 9:878952. [PMID: 35669174 PMCID: PMC9164269 DOI: 10.3389/fvets.2022.878952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022] Open
Abstract
For both human and veterinary patients, non-infectious intestinal disease is a major cause of morbidity and mortality. To improve treatment of intestinal disease, large animal models are increasingly recognized as critical tools to translate the basic science discoveries made in rodent models into clinical application. Large animal intestinal models, particularly porcine, more closely resemble human anatomy, physiology, and disease pathogenesis; these features make them critical to the pre-clinical study of intestinal disease treatments. Previously, large animal model use has been somewhat precluded by the lack of genetically altered large animals to mechanistically investigate non-infectious intestinal diseases such as colorectal cancer, cystic fibrosis, and ischemia-reperfusion injury. However, recent advances and increased availability of gene editing technologies has led to both novel use of large animal models in clinically relevant intestinal disease research and improved testing of potential therapeutics for these diseases.
Collapse
|
210
|
Phage Cocktail Targeting STEC O157:H7 Has Comparable Efficacy and Superior Recovery Compared with Enrofloxacin in an Enteric Murine Model. Microbiol Spectr 2022; 10:e0023222. [PMID: 35536028 PMCID: PMC9241756 DOI: 10.1128/spectrum.00232-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
O157:H7 is the most important Shiga toxin-producing Escherichia coli (STEC) serotype in relation to public health. Given that antibiotics may contribute to the exacerbation of STEC-related disease and an increased frequency of antibiotic-resistant strains, bacteriophage (phage) therapy is considered a promising alternative. However, phage therapy targeting enteric pathogens is still underdeveloped with many confounding effects from the microbiota. Here we comprehensively compared the therapeutic efficacy of a phage cocktail with the antibiotic enrofloxacin in a mouse model of STEC O157:H7 EDL933 infection. Enrofloxacin treatment provided 100% survival and the phage cocktail treatment provided 90% survival. However, in terms of mouse recovery, the phage cocktail outperformed enrofloxacin in all measured outcomes. Compared with enrofloxacin treatment, phage treatment led to a faster elimination of enteric pathogens, decreased expression levels of inflammatory markers, increased weight gain, maintenance of a stable relative organ weight, and improved homeostasis of the gut microbiota. These results provide support for the potential of phage therapy to combat enteric pathogens and suggest that phage treatment leads to enhanced recovery of infected mice compared with antibiotics. IMPORTANCE With the increasing severity of antibiotic resistance and other adverse consequences, animal experiments and clinical trials investigating the use of phages for the control and prevention of enteric bacterial infections are growing. However, the effects of phages and antibiotics on organisms when treating intestinal infections have not been precisely studied. Here, we comprehensively compared the therapeutic efficacy of a phage cocktail to the antibiotic enrofloxacin in a mouse model of STEC O157:H7 EDL933 infection. We found that, despite a slightly lower protection rate, phage treatment contributed to a faster recovery of infected mice compared with enrofloxacin. These results highlight the potential benefits of phage therapy to combat enteric infections.
Collapse
|
211
|
Dong J, Ping L, Zhang K, Tang H, Liu J, Liu D, Zhao L, Evivie SE, Li B, Huo G. Immunomodulatory effects of mixed Lactobacillus plantarum on lipopolysaccharide-induced intestinal injury in mice. Food Funct 2022; 13:4914-4929. [PMID: 35395665 DOI: 10.1039/d1fo04204a] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The intestine is the largest digestive and immune organ in the human body, with an intact intestinal mucosal barrier. Lactobacillus plantarum is an important strain of probiotics in the intestine for boosting intestinal immunity to defend against intestinal injury. In the lipopolysaccharide-induced intestinal injury model, mixed L. plantarum (L. plantarum KLDS 1.0318, L. plantarum KLDS 1.0344, and L. plantarum KLDS 1.0386) was suggested to boost intestinal immunity. In detail, compared with LPS-induced mice, mice in the mixed L. plantarum group showed significantly reduced intestine (jejunum, ileum, and colon) tissue injury, pro-inflammatory cytokine (TNF-α, IL-6 and IL-12) levels, myeloperoxidase activities, and serum D-lactate (P < 0.05) content. Moreover, the mixed L. plantarum significantly increased the number of immunocytes (CD4+ T cells, IgA plasma cells) and the expression of tight junction proteins (Claudin1 and Occludin). The results also showed that the mixed L. plantarum significantly down-regulated (P < 0.05) the intestinal protein expression of TLR4, p-IκB, and NF-κB p65. The mixed L. plantarum group increased the relative abundance of the genera, including Lactobacillus, Lachnoclostridium, and Desulfovibrio, which are related to improving the levels of SCFAs (acetic acid, butyric acid) and total bile acid (P < 0.05). Overall, these results indicated that the mixed L. plantarum had great functionality in reducing LPS-induced intestinal injury.
Collapse
Affiliation(s)
- Jiahuan Dong
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Lijun Ping
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Kangyong Zhang
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Hongwei Tang
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Jie Liu
- Beijing Technology and Business University, Beijing 100048, China
| | - Deyu Liu
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Li Zhao
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Smith Etareri Evivie
- Department of Food Science and Human Nutrition, Faculty of Agriculture, University of Benin, Benin City 300001, Nigeria.,Department of Animal Science, Faculty of Agriculture, University of Benin, Benin City 300001, Nigeria
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Guicheng Huo
- Food College, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
212
|
Abstract
We demonstrate that AP patients and experimental AP mice exhibited a dysfunction of Paneth cells. Our
in vivo
research showed that the severity of AP was exacerbated by the long-term dysfunction of Paneth cells, which was associated with gut microbiota disorder.
Collapse
|
213
|
Lei W, Zhao C, Sun J, Jin Y, Duan Z. Activation of α7nAChR preserves intestinal barrier integrity by enhancing the HO-1 / STAT3 signaling to inhibit NF-κB activation in mice. Biomed Pharmacother 2022; 149:112733. [DOI: 10.1016/j.biopha.2022.112733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 11/02/2022] Open
|
214
|
Deng L, Zhou X, Lan Z, Tang K, Zhu X, Mo X, Zhao Z, Zhao Z, Wu M. Simotang Alleviates the Gastrointestinal Side Effects of Chemotherapy by Altering Gut Microbiota. J Microbiol Biotechnol 2022; 32:405-418. [PMID: 35283422 PMCID: PMC9628794 DOI: 10.4014/jmb.2110.10018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Simotang oral liquid (SMT) is a traditional Chinese medicine (TCM) consisting of four natural plants and is used to alleviate gastrointestinal side effects after chemotherapy and functional dyspepsia (FD). However, the mechanism by which SMT helps cure these gastrointestinal diseases is still unknown. Here, we discovered that SMT could alleviate gastrointestinal side effects after chemotherapy by altering gut microbiota. C57BL/6J mice were treated with cisplatin (DDP) and SMT, and biological samples were collected. Pathological changes in the small intestine were observed, and the intestinal injury score was assessed. The expression levels of the inflammatory factors IL-1β and IL-6 and the adhesive factors Occludin and ZO-1 in mouse blood or small intestine tissue were also detected. Moreover, the gut microbiota was analyzed by high-throughput sequencing of 16S rRNA amplicons. SMT was found to effectively reduce gastrointestinal mucositis after DDP injection, which lowered inflammation and tightened the intestinal epithelial cells. Gut microbiota analysis showed that the abundance of the anti-inflammatory microbiota was downregulated and that the inflammatory microbiota was upregulated in DDP-treated mice. SMT upregulated anti-inflammatory and anticancer microbiota abundance, while the inflammatory microbiota was downregulated. An antibiotic cocktail (ABX) was also used to delete mice gut microbiota to test the importance of gut microbiota, and we found that SMT could not alleviate gastrointestinal mucositis after DDP injection, showing that gut microbiota might be an important mediator of SMT treatment. Our study provides evidence that SMT might moderate gastrointestinal mucositis after chemotherapy by altering gut microbiota.
Collapse
Affiliation(s)
- Lijing Deng
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Xingyi Zhou
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Zhifang Lan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Kairui Tang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Xiaoxu Zhu
- Hubei University of Chinese Medicine, Wuhan 430065, P.R. China
| | - Xiaowei Mo
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Zongyao Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Zhiqiang Zhao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, P.R. China,Corresponding authors Zhiqiang Zhao Phone: +86-20-8775-5766 E-mail:
| | - Mansi Wu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, P.R. China,
Mansi Wu Phone: +86-20-8522-1543 E-mail:
| |
Collapse
|
215
|
Wang J, Xu H, Chen T, Xu C, Zhang X, Zhao S. Effect of Monoacylglycerol Lipase Inhibition on Intestinal Permeability of Rats With Severe Acute Pancreatitis. Front Pharmacol 2022; 13:869482. [PMID: 35496266 PMCID: PMC9039313 DOI: 10.3389/fphar.2022.869482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Endocannabinoid 2-arachidonoylglycerol (2-AG) is an anti-nociceptive lipid that is inactivated through cellular uptake and subsequent catabolism by monoacylglycerol lipase (MAGL). In this study, we investigated the effects of MAGL inhibition on intestinal permeability and explored the possible mechanism. Methods: A rat model of severe acute pancreatitis (SAP) was established. Rats were divided into three groups according to treatment. We analyzed intestinal permeability to fluorescein isothiocyanate-dextran and the levels of inflammatory factors interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) and 2-AG. Hematoxylin and eosin staining was used to assess histological tissue changes. In vivo intestinal permeability was evaluated by transmission electron microscopy. We obtained ileum tissues, extracted total RNA, and applied RNA-sequencing. Sequencing data were analyzed by bioinformatics. Results: Inflammatory factor levels were higher, while 2-AG levels were lower in the SAP group compared with the control group. Administration of JZL184 to rats with SAP increased the levels of 2-AG and lowered the levels of IL-6 and TNF-α. Notably, intestinal permeability was improved by JZL184 as demonstrated by fluorescein isothiocyanate-dextran measurement, hematoxylin and eosin staining, and transmission electron microscopy. RNA-sequencing showed significant transcriptional differences in SAP and JZL184 groups compared with the control group. KEGG analysis showed that the up- or downregulated genes in multiple comparison groups were enriched in two pathways, focal adhesion and PI3K-Akt signaling pathways. Differential alternative splicing (AS) genes, such as Myo9b, Lsp1, and Git2, have major functions in intestinal diseases. A total of 132 RNA-binding proteins (RBPs) were screened by crossing the identified abnormally expressed genes with the reported RBP genes. Among them, HNRNPDL coexpressed the most AS events as the main RBP. Conclusion: MAGL inhibition improved intestinal mucosal barrier injury in SAP rats and induced a large number of differentially expressed genes and alternative splicing events. HNRNPDL might play an important role in improving intestinal mucosal barrier injury by affecting alternative splicing events.
Collapse
|
216
|
The healing effects of Ganoderma lucidum on intestinal ischemia-reperfusion damage in rats. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.1029203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
217
|
Lyu H, Ni H, Huang J, Yu G, Zhang Z, Zhang Q. VX-765 prevents intestinal ischemia-reperfusion injury by inhibiting NLRP3 inflammasome. Tissue Cell 2022; 75:101718. [PMID: 35131633 DOI: 10.1016/j.tice.2021.101718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intestinal ischemia-reperfusion injury (IIRI) is a common clinical event that can cause serious consequences. The study aimed to investigated the effect of VX-765 in IIRI and its mechanism. METHODS The hypoxia-reoxygenation (H/R) cell model and IIRI mouse model were generated to examine the in vitro and in vivo effects of VX-765 on IIRI. IIRI was evaluated by histological assessment. ELISA was performed to determine the levels of IL-6, TNF-α, IL-1β, caspase-1, and GSDMD in intestinal tissues as well as the levels of MDA, SOD, CAT, caspase-1, and GSDMD in Caco-2 cells. Relative protein levels of NLRP3, ASC, IL-18, IL-1β, cleaved Caspase1, and GSDMD-N were analyzed by Western blotting. CCK-8 Assay was conducted to determine the optimal concentration of VX-765 for the in vitro studies. Flow cytometry, fluorescence microscopy and real-time PCR (RT-PCR) were used to assess ROS levels and the mRNA levels of IL-18 and IL-1β, respectively. Immunofluorescence staining was performed to examine the subcellular localization of P65 and NLRP3. RESULTS VX-765 reduced IIRI-induced oxidative stress and inflammatory response both in vivo and in vitro, while it decreased the levels of TNF-α, IL-6, IL-1β as well as the modified Park/Chiu scores. The optimal concentration of VX-765 for the in vitro studies was 10 μM. Moreover, VX-765 inhibited the nuclear translocation of P65, reduced oxidative stress and down-regulated the activation of NLRP3 inflammasome. CONCLUSION VX-765 prevents IIRI presumably by inhibiting the activation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Heping Lyu
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Haizhen Ni
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Jingyong Huang
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Guanfeng Yu
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Zhongjing Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Qiyu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China.
| |
Collapse
|
218
|
Han X, Feng Z, Chen Y, Zhu L, Li X, Wang X, Sun H, Li J. Effects of High-Fructose Corn Syrup on Bone Health and Gastrointestinal Microbiota in Growing Male Mice. Front Nutr 2022; 9:829396. [PMID: 35433775 PMCID: PMC9005738 DOI: 10.3389/fnut.2022.829396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/07/2022] [Indexed: 01/21/2023] Open
Abstract
Here, we explored the correlation between gut microbiota and bone health and the effects of high-fructose corn syrup (HFCS) on both. Sixteen 3-week-old male C57BL/6J mice were randomly divided into two groups and given purified water (control group) or 30% HFCS in water (HFCS group) for 16 weeks. The effects of HFCS were assessed via enzyme-linked immunosorbent assays, histopathological assays of colon and bone, and 16S rDNA sequence analysis of gut microbiota. The serum of HFCS group mice had lower levels of bone alkaline phosphatase (BALP), bone Gla protein (BGP), insulin-like growth factor 1 (IGF-1), and testosterone, and higher levels of type I collagen carboxyl-terminal telopeptide (ICTP) and tartrate-resistant acid phosphatase (TRAP) than that of the control group. HFCS caused trabecular bone damage by decreasing trabecular number and thickness and increasing trabecular separation. The HFCS group colons were shorter than the control group colons. The HFCS-fed mice showed mild, localized shedding of epithelial cells in the mucosal layer, focal lymphocytic infiltration of the lamina propria, mild submucosal edema, and loosely arranged connective tissue. The HFCS group displayed lower abundance and altered composition of gut microbiota. The abundance of Defluviitaleaceae UCG-011, Erysipelatoclostridium, Ruminococcaceae UCG-009, Lactobacillus, Blautia, and Parasutterella increased, positively correlating with BALP, BGP, IGF-1, and testosterone levels, and negatively correlating with ICTP and TRAP levels. Our study revealed a potential diet-gut microbiota-bone health axis.
Collapse
Affiliation(s)
- Xiaoqiang Han
- Department of Orthopedics, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhiguo Feng
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yizhang Chen
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Liying Zhu
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xiaoqiong Li
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xin Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Haibiao Sun
- Department of Orthopedics, The First Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Haibiao Sun,
| | - Jinjun Li
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- Jinjun Li,
| |
Collapse
|
219
|
Oral H, Türkyılmaz Z, Karabulut R, Kaya C, Dayanır D, Karakaya C, Sonmez K. Protective Effects of Hydrogen-Rich Saline on Experimental Intestinal Volvulus in Rats. J INVEST SURG 2022; 35:1427-1433. [PMID: 35331073 DOI: 10.1080/08941939.2022.2056273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Intestinal volvulus can cause morbidity and mortality. Surgical reduction, on the other hand, could result in ischemia-reperfusion (I/R) injury. Hydrogen rich saline solution (HRSS neutralizes free radicals in the body. This study aimed to investigate the effects of HRSS in I/R injury in experimental intestinal volvulus in rats. METHODS Thirty rats were randomly allocated into 5 groups. All procedures were done under general anesthesia and sterile conditions in each animal. Five ml/kg of saline and HRSS were administered intraperitoneally (ip) in Sham (Group 1) and HRSS (Group 2) groups, respectively. Groups 3, 4, and 5 constituted the study groups in which volvulus was created in a 5-cm- long ileal segment 2 cm proximal to the ileocecal valve. After 2 hours the volvuli were reduced and following 2 hours of reperfusion, these segments were removed. In volvulus-I/R group (Group 3) no additional procedure was done. HRSS was administered shortly before reperfusion (reduction of the volvulus) in Treatment I (Group 4) and 1 h before experimental volvulus in Treatment II (Group 5) groups. Blood and intestinal tissue samples were obtained from all rats at the 4th hour. Both tissue and blood total oxidant (TOS) and antioxidant status (TAS) levels were determined and tissue histomorphologies were studied. Oxidative stress indices (TOS ÷ TAS) (OSI) were calculated. RESULTS Tissue TOS and OSI levels and histomorphological injury scores were statistically lower in treatment groups than I/R group, whereas blood TOS and OSI levels were similar between the groups. CONCLUSIONS This study provides biochemical and histomorphological evidence that HRSS prevents intestinal damage in I/R injury caused by volvulus.
Collapse
Affiliation(s)
- Hayrunnisa Oral
- Departments of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Zafer Türkyılmaz
- Departments of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ramazan Karabulut
- Departments of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Cem Kaya
- Departments of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Duygu Dayanır
- Histology and Embryology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Cengiz Karakaya
- Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Kaan Sonmez
- Departments of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
220
|
Guo S, Huang Z, Zhu J, Yue T, Wang X, Pan Y, Bu D, Liu Y, Wang P, Chen S. CBS-H 2S axis preserves the intestinal barrier function by inhibiting COX-2 through sulfhydrating human antigen R in colitis. J Adv Res 2022; 44:201-212. [PMID: 36725190 PMCID: PMC9936422 DOI: 10.1016/j.jare.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/15/2021] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Lipopolysaccharide (LPS) causes lesions of the epithelial barrier, which allows translocation of pathogens from the intestinal lumen to the host's circulation. Hydrogen sulfide (H2S) regulates multiple physiological and pathological processes in colonic epithelial tissue, and CBS-H2S axis involved in multiple gastrointestinal disorder. However, the mechanism underlying the effect of the CBS-H2S axis on the intestinal and systemic inflammation in colitis remains to be illustrated. OBJECTIVES To investigate the effect of CBS-H2S axis on the intestinal and systematic inflammation related injuries in LPS induced colitis and the underlying mechanisms. METHODS Wild type and CBS-/+ mice were used to evaluate the effect of endogenous and exogenous H2S on LPS-induced colitis in vivo. Cytokine quantitative antibody array, western blot and real-time PCR were applied to detect the key cytokines in the mechanism of action. Biotin switch of S-sulfhydration, CRISPR/Cas9 mediated knockout, immunofluorescence and ActD chase assay were used in the in vitro experiment to further clarify the molecular mechanisms. RESULTS H2S significantly alleviated the symptoms of LPS-induced colitis in vivo and attenuated the increase of COX-2 expression. The sulfhydrated HuR increased when CBS express normally or GYY4137 was administered. While after knocking kown CBS, the expression of COX-2 in mice colon increased significantly, and the sulfhydration level of HuR decreased. The results in vitro illustrated that HuR can increase the stability of COX-2 mRNA, and the decrease of COX-2 were due to increased sulfhydration of HuR rather than the reduction of total HuR levels. CONCLUSION These results indicated that CBS-H2S axis played an important role in protecting intestinal barrier function in colitis. CBS-H2S axis increases the sulfhydration level of HuR, by which reduces the binding of HuR with COX-2 mRNA and inhibited the expression of COX-2.
Collapse
Affiliation(s)
- Shihao Guo
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China,Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Zhihao Huang
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Jing Zhu
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Taohua Yue
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Xin Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Dingfang Bu
- Central Laboratory, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Yucun Liu
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People’s Republic of China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People's Republic of China.
| | - Shanwen Chen
- Division of General Surgery, Peking University First Hospital, Peking University, 8, Beijing 100034, People's Republic of China.
| |
Collapse
|
221
|
Lei W, Zhao C, Sun J, Jin Y, Duan Z. Electroacupuncture Ameliorates Intestinal Barrier Destruction in Mice With Bile Duct Ligation-Induced Liver Injury by Activating the Cholinergic Anti-Inflammatory Pathway. Neuromodulation 2022; 25:1122-1133. [PMID: 35300921 DOI: 10.1016/j.neurom.2022.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Electroacupuncture (EA) at Zusanli (ST36) can attenuate inflammation in different rodent models. However, the therapeutic mechanisms underlying its action in inhibiting intestinal barrier destruction and liver injury in cholestasis mice have not been clarified. This study aimed at investigating whether EA at ST36 could activate the cholinergic anti-inflammatory pathway to inhibit intestinal barrier destruction and liver injury in cholestasis mice. MATERIALS AND METHODS Male Hmox1floxp/floxp C57BL/6 mice were randomized and subjected to a sham or bile duct ligation (BDL) surgery. The BDL mice were randomized and treated with, or without (BDL group), sham EA at ST36 (BDL+sham-ST36) or EA at ST36 (BDL+ST36), or received α-bungarotoxin (α-BGT), a specific inhibitor of nicotinic acetylcholine receptor α7 subunit (α7nAChR), before stimulation (BDL+ST36+α-BGT). These mice, together with a group of intestine-specific heme oxygenase-1 (HO-1) knockout (KO) Villin-Cre-HO-1-/- mice, were monitored for their body weights before and 14 days after BDL. The levels of plasma cytokines and liver injury-related alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured by enzyme-linked immunoassay, and pathological changes in the intestinal mucosa and liver fibrosis as well as intestinal barrier permeability in individual mice were examined by histology and immunohistochemistry. The levels of α7nAChR, HO-1, ZO-1, Occludin, Claudin-1, and NF-κBp65 expression and NF-κBp65 phosphorylation in intestinal tissues were quantified. RESULTS Compared with the sham group, BDL significantly increased the levels of plasma interleukin (IL)-1β, IL-6, IL-10, tumor necrosis factor α, ALT, and AST and caused intestinal mucosal damages, high permeability, and liver fibrosis in mice, which were remarkably mitigated, except for further increased levels of plasma IL-10 in the BDL+ST36 group of mice. Similarly, EA at ST36 significantly up-regulated α7nAChR and HO-1 expression; mitigated the BDL-decreased ZO-1, Occludin, and Claudin-1 expression; and attenuated the BDL-increased NF-κBp65 phosphorylation in intestinal tissues of mice. The therapeutic effects of EA at ST36 were significantly abrogated by pretreatment with α-BGT or HO-1 KO. CONCLUSION EA at ST36 inhibits the BDL-induced intestinal mucosal damage and liver fibrosis by activating the HO-1 cholinergic anti-inflammatory pathway in intestinal tissues of mice.
Collapse
Affiliation(s)
- Wei Lei
- Second Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, China; Laboratory of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Changcheng Zhao
- Second Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, China; Laboratory of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiasen Sun
- Second Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, China; Laboratory of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanling Jin
- Pathology Department, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhijun Duan
- Second Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, China; Laboratory of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
222
|
Barry M, Trivedi A, Pathipati P, Miyazawa BY, Vivona LR, Togarrati PP, Khakoo M, Tanner H, Norris P, Pati S. Mesenchymal stem cell extracellular vesicles mitigate vascular permeability and injury in the small intestine and lung in a mouse model of hemorrhagic shock and trauma. J Trauma Acute Care Surg 2022; 92:489-498. [PMID: 34882596 PMCID: PMC8866219 DOI: 10.1097/ta.0000000000003487] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hemorrhagic shock and trauma (HS/T)-induced gut injury may play a critical role in the development of multi-organ failure. Novel therapies that target gut injury and vascular permeability early after HS/T could have substantial impacts on trauma patients. In this study, we investigate the therapeutic potential of human mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC EVs) in vivo in HS/T in mice and in vitro in Caco-2 human intestinal epithelial cells. METHODS In vivo, using a mouse model of HS/T, vascular permeability to a 10-kDa dextran dye and histopathologic injury in the small intestine and lungs were measured among mice. Groups were (1) sham, (2) HS/T + lactated Ringer's (LR), (3) HS/T + MSCs, and (4) HS/T + MSC EVs. In vitro, Caco-2 cell monolayer integrity was evaluated by an epithelial cell impedance assay. Caco-2 cells were pretreated with control media, MSC conditioned media (CM), or MSC EVs, then challenged with hydrogen peroxide (H2O2). RESULTS In vivo, both MSCs and MSC EVs significantly reduced vascular permeability in the small intestine (fluorescence units: sham, 456 ± 88; LR, 1067 ± 295; MSC, 765 ± 258; MSC EV, 715 ± 200) and lung (sham, 297 ± 155; LR, 791 ± 331; MSC, 331 ± 172; MSC EV, 303 ± 88). Histopathologic injury in the small intestine and lung was also attenuated by MSCs and MSC EVs. In vitro, MSC CM but not MSC EVs attenuated the increased permeability among Caco-2 cell monolayers challenged with H2O2. CONCLUSION Mesenchymal stem cell EVs recapitulate the effects of MSCs in reducing vascular permeability and injury in the small intestine and lungs in vivo, suggesting MSC EVs may be a potential cell-free therapy targeting multi-organ dysfunction in HS/T. This is the first study to demonstrate that MSC EVs improve both gut and lung injury in an animal model of HS/T.
Collapse
Affiliation(s)
- Mark Barry
- University of California, San Francisco. Department of Surgery. 513 Parnassus Ave. San Francisco, CA 94143
| | - Alpa Trivedi
- University of California, San Francisco. Department of Laboratory Medicine. 513 Parnassus Ave. San Francisco, CA 94143
| | - Praneeti Pathipati
- University of California, San Francisco. Department of Laboratory Medicine. 513 Parnassus Ave. San Francisco, CA 94143
| | - Byron Y. Miyazawa
- University of California, San Francisco. Department of Laboratory Medicine. 513 Parnassus Ave. San Francisco, CA 94143
| | - Lindsay R. Vivona
- University of California, San Francisco. Department of Laboratory Medicine. 513 Parnassus Ave. San Francisco, CA 94143
| | | | - Manisha Khakoo
- University of California, San Francisco. Department of Laboratory Medicine. 513 Parnassus Ave. San Francisco, CA 94143
| | - Heather Tanner
- Vitalant Research Institute. 270 Masonic Ave. San Francisco, CA 94118
| | - Philip Norris
- Vitalant Research Institute. 270 Masonic Ave. San Francisco, CA 94118
| | - Shibani Pati
- University of California, San Francisco. Department of Laboratory Medicine. 513 Parnassus Ave. San Francisco, CA 94143
| |
Collapse
|
223
|
Niu Q, Liu F, Zhang J, Yang X, Wang X. Carbon monoxide-releasing molecule-2 protects intestinal mucosal barrier function by reducing epithelial tight-junction damage in rats undergoing cardiopulmonary resuscitation. JOURNAL OF INTENSIVE MEDICINE 2022; 2:118-126. [PMID: 36789186 PMCID: PMC9923997 DOI: 10.1016/j.jointm.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/15/2021] [Accepted: 01/12/2022] [Indexed: 11/29/2022]
Abstract
Background Ischemia-reperfusion injury (IRI) to the small intestine is associated with the development of systemic inflammation and multiple organ failure after cardiopulmonary resuscitation (CPR). It has been reported that exogenous carbon monoxide (CO) reduces IRI. This study aimed to assess the effects of carbon monoxide-releasing molecule-2 (CORM-2) on intestinal mucosal barrier function in rats undergoing CPR. Methods We established a rat model of asphyxiation-induced cardiac arrest (CA) and resuscitation to study intestinal IRI, and measured the serum levels of intestinal fatty acid-binding protein. Morphological changes were investigated using light and electron microscopes. The expression levels of claudin 3 (CLDN3), occludin (OCLN), zonula occludens 1 (ZO-1), tumor necrosis factor-alpha (TNF-α), interleukin-10 (IL-10), and nuclear factor kappa B (NF-κB) p65 were detected by western blotting. Results Compared with the sham-operated group, histological changes and transmission electron microscopy revealed severe intestinal mucosal injury in the CPR and inactive CORM-2 (iCORM-2) groups. In contrast, CORM-2 alleviated intestinal IRI. CORM-2, unlike iCORM-2, markedly decreased the Chiu's scores (2.38 ± 0.38 vs. 4.59 ± 0.34; P < 0.05) and serum intestinal fatty acid-binding protein level (306.10 ± 19.22 vs. 585.64 ± 119.84 pg/mL; P < 0.05) compared with the CPR group. In addition, CORM-2 upregulated the expression levels of tight junction proteins (CLDN3, OCLN, and ZO-1) (P < 0.05) and downregulated those of IL-10, TNF-α, and NF-кB p65 (P < 0.05) in the ileum tissue of rats that received CPR. Conclusions CORM-2 prevented intestinal mucosal damage as a result of IRI during CPR. The underlying protective mechanism was associated with inhibition of ischemia-reperfusion-induced changes in intestinal epithelial permeability and inflammation in intestinal tissue.
Collapse
Affiliation(s)
- Qingsheng Niu
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China,Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Fang Liu
- Department of Critical Care Medicine, Heze Second People's Hospital, Heze, Shandong 274000, China
| | - Jun Zhang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Xiaojun Yang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China
| | - Xiaohong Wang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China,Corresponding author: Xiaohong Wang, Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, China.
| |
Collapse
|
224
|
Chen C, Lin Z, Zhang X, Zhang X, Cheng Z, Jin T, Liu T, Deng L, Guo J, Wang G, Xia Q. Extracellular histones cause intestinal epithelium injury and disrupt its barrier function in vitro and in vivo. Toxicology 2022; 469:153117. [PMID: 35122914 DOI: 10.1016/j.tox.2022.153117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023]
Abstract
Extracellular histones are cytotoxic to various cells and have been extensively proven a vital mediator of multiple organ injuries. However, the effect of extracellular histones on the intestine remains largely unknown. This study aimed to clarify the effect of extracellular histones on the intestine. IEC-6, a cell line of rat small intestinal epithelial crypt, and C57BL/6 or ICR mice were treated with histones. The IEC-6 cells treated with histones from 20 μg/mL to 200 μg/mL for 0-24 h displayed a decline of cell viability and an increase of cell death in a concentration- and time-dependent manner. Moreover, histones (100 μg/mL) induced IEC-6 apoptosis through activating caspase 3 and necroptosis through up-regulation of receptor-interacting serine/threonine protein kinase 1 and 3 (RIPK1 and RIPK3), phosphorylated mixed-lineage kinase domain-like protein (p-MLKL) along with the decrease of caspase-8. Histones treatment disturbed zonular occludens 1 (ZO-1) expression and increased permeability of IEC-6 cell monolayer. In vivo, histones 50 mg/kg injection caused mice intestinal edema, loss apex of villus, epithelial lifting down the sides of the villi, and increased neutrophil infiltration. Elevation of serum intestinal fatty acid binding protein (I-FABP), D-lactate, or Diamine oxidase (DAO) and loss of tight junction protein, ZO-1, at 3 h and 6 h after histones injection strongly indicated severe intestinal epithelium injury, which led to increased permeability of the intestine. In conclusion, extracellular histones cause intestinal epithelial damage via direct cytotoxicity. Consequently, intestinal epithelial tight junction and barrier integrity are disrupted, which may play pivotal roles in diverse diseases.
Collapse
Affiliation(s)
- Chanjuan Chen
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziqi Lin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiaoxin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoying Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenxing Cheng
- The Medical School, Southeast University, Nanjing, 210009, China; Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Tao Jin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tingting Liu
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lihui Deng
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia Guo
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guozheng Wang
- The Medical School, Southeast University, Nanjing, 210009, China
| | - Qing Xia
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
225
|
Vecchio Dezillio LE, Romanin DE, Ivanoff Marinoff IM, Vernengo J, Abate Zárate JC, Machuca MA, Gondolesi GE, Lausada NR, Stringa PL, Rumbo M. Experimental study to assess the impact of vasopressors administered during maintenance of the brain-dead donation in the quality of the intestinal graft. J Trauma Acute Care Surg 2022; 92:380-387. [PMID: 35081098 DOI: 10.1097/ta.0000000000003473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The hemodynamic maintenance of brain-dead donors will influence the quality of the organs procured for transplantation, including the intestine. Although norepinephrine (NE) and dopamine (DA) are commonly used to sustain mean arterial pressure in humans, there are no standardized protocols for their use during maintenance of brain-dead donors. Our aim was to compare the effects of each drug, in the intestinal graft quality using a rat brain-dead donation model. METHODS Wistar rats (N = 17) underwent brain death (BD) for 2 hours with NE (NE group) or with DA (DA group) administration; the control group was mechanically ventilated for 2 hours without BD. Jejunum biopsies were obtained at the end of the maintenance period. Histological damage was evaluated using Park-Chiu scale. Villi/crypt ratio, mucosal thickness, Goblet cell count, and villi density were evaluated using ImageJ software (US National Institutes of Health, Bethesda, MD). Barrier damage was assessed by bacterial translocation culture counting on liver samples. The inflammatory status of the intestine was evaluated by CD3+ counting by immunohistochemistry and gene expression analysis of interleukin (IL)-6, IL-22, and CXCL10. RESULTS Norepinephrine-treated donors had higher focal ischemic injury in the intestinal mucosa without a substantial modification of morphometrical parameters compared with DA-treated donors. CD3+ mucosal infiltration was greater in intestines procured from brain-dead donors, being highest in NE (p ˂ 0.001). Local inflammatory mediators were affected in BD: DA and NE groups showed a trend to lower expression of IL-22, whereas CXCL10 expression was higher in NE versus control group. Brain death promoted intestinal bacterial translocation, but the use of NE resulted in the highest bacterial counting in the liver (p ˂ 0.01). CONCLUSION Our results favor the use of DA instead of NE as main vasoactive drug to manage BD-associated hemodynamic instability. Dopamine may contribute to improve the quality of the intestinal graft, by better preserving barrier function and lowering immune cell infiltration.
Collapse
Affiliation(s)
- Leandro Emmanuel Vecchio Dezillio
- From the Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP-CONICET-UNLP) (L.E.V.D., D.E.R., I.M.I.M., J.V., P.L.S., M.R.), Universidad Nacional de La Plata, La Plata, Argentina; Cátedra de Trasplante de Órganos (L.E.V.D., J.C.A.Z., N.R.L., P.L.S.), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina; Laboratorio de Patología Especial (M.A.M.), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata, Argentina; Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB-CONICET) (G.E.G.), Buenos Aires, Argentina; and Instituto de Trasplante Multiorgánico (G.E.G.), Hospital Universitario Fundación Favaloro, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Barisic I, Balenovic D, Udovicic M, Bardak D, Strinic D, Vlainić J, Vranes H, Smoday IM, Krezic I, Milavic M, Sikiric S, Uzun S, Zivanovic Posilovic G, Strbe S, Vukoja I, Lovric E, Lozic M, Sever M, Lovric Bencic M, Boban Blagaic A, Skrtic A, Seiwerth S, Sikiric P. Stable Gastric Pentadecapeptide BPC 157 May Counteract Myocardial Infarction Induced by Isoprenaline in Rats. Biomedicines 2022; 10:265. [PMID: 35203478 PMCID: PMC8869603 DOI: 10.3390/biomedicines10020265] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/17/2022] Open
Abstract
We revealed that the stable gastric pentadecapeptide BPC 157, a useful peptide therapy against isoprenaline myocardial infarction, as well as against isoprenaline myocardial reinfarction, may follow the counteraction of the recently described occlusion-like syndrome, induced peripherally and centrally, which was described for the first time in isoprenaline-treated rats. BPC 157 (10 ng/kg, 10 µg/kg i.p.), L-NAME (5 mg/kg i.p.), and L-arginine (200 mg/kg i.p.) were given alone or together at (i) 30 min before or, alternatively, (ii) at 5 min after isoprenaline (75 or 150 mg/kg s.c.). At 30 min after isoprenaline 75 mg/kg s.c., we noted an early multiorgan failure (brain, heart, lung, liver, kidney and gastrointestinal lesions), thrombosis, intracranial (superior sagittal sinus) hypertension, portal and caval hypertension, and aortal hypotension, in its full presentation (or attenuated by BPC 157 therapy (given at 5 min after isoprenaline) via activation of the azygos vein). Further, we studied isoprenaline (75 or 150 mg/kg s.c.) myocardial infarction (1 challenge) and reinfarction (isoprenaline at 0 h and 24 h, 2 challenges) in rats (assessed at the end of the subsequent 24 h period). BPC 157 reduced levels of all necrosis markers, CK, CK-MB, LDH, and cTnT, and attenuated gross (no visible infarcted area) and histological damage, ECG (no ST-T ischemic changes), and echocardiography (preservation of systolic left ventricular function) damage induced by isoprenaline. Its effect was associated with a significant decrease in oxidative stress parameters and likely maintained NO system function, providing that BPC 157 interacted with eNOS and COX2 gene expression in a particular way and counteracted the noxious effect of the NOS-blocker, L-NAME.
Collapse
Affiliation(s)
- Ivan Barisic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Diana Balenovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Mario Udovicic
- Department of Internal Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.U.); (M.L.B.)
| | - Darija Bardak
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Dean Strinic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Josipa Vlainić
- Laboratory for Advanced Genomics, Division of Molecular Medicine, lnstitute Ruder Boskovic, 10000 Zagreb, Croatia;
| | - Hrvoje Vranes
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Ivan Maria Smoday
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Marija Milavic
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.M.); (S.S.); (E.L.); (S.S.)
| | - Suncana Sikiric
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.M.); (S.S.); (E.L.); (S.S.)
| | - Sandra Uzun
- Clinic of Anaesthesiology, Reanimatology and Intensive Care Zagreb, University Hospital Centre Zagreb, 10000 Zagreb, Croatia;
| | - Gordana Zivanovic Posilovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Ivan Vukoja
- School of Medicine, University of Osijek, 31000 Osijek, Croatia;
| | - Eva Lovric
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.M.); (S.S.); (E.L.); (S.S.)
| | - Marin Lozic
- Department of Pediatric and Preventive Dentistry, School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Marko Sever
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Martina Lovric Bencic
- Department of Internal Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.U.); (M.L.B.)
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| | - Anita Skrtic
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.M.); (S.S.); (E.L.); (S.S.)
| | - Sven Seiwerth
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.M.); (S.S.); (E.L.); (S.S.)
| | - Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (I.B.); (D.B.); (D.B.); (D.S.); (H.V.); (I.M.S.); (I.K.); (G.Z.P.); (S.S.); (A.B.B.)
| |
Collapse
|
227
|
JAK2/STAT3 inhibition attenuates intestinal ischemia-reperfusion injury via promoting autophagy: in vitro and in vivo study. Mol Biol Rep 2022; 49:2857-2867. [PMID: 35067814 DOI: 10.1007/s11033-021-07099-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/16/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Intestinal ischemia-reperfusion (I/R) causes severe injury to the intestine, leading to systemic inflammation and multiple organ failure. Autophagy is a stress-response mechanism that can protect against I/R injury by removing damaged organelles and toxic protein aggregates. Recent evidence has identified JAK-STAT signaling pathway as a new regulator of autophagy process, however, their regulatory relationship in intestinal I/R remains unknown. METHODS AND RESULTS We systematically analyzed intestinal transcriptome data and found that JAK-STAT pathway was largely activated in response to I/R with most significant upregulation observed for JAK2 and STAT3. ChIP-Seq and luciferase assays in an in vitro oxygen-glucose deprivation and reoxygenation model revealed that activated JAK2/STAT3 signaling directly inhibited the transcription of autophagy regulator Beclin-1, leading to the suppression of autophagy and the activation of intestinal cell death. These findings were further confirmed in an in vivo mouse model, in which, intestinal I/R injury was associated with the activation of JAK2/STAT3 pathway and the deactivation of Beclin-1-mediated autophagy, while inhibiting JAK2/STAT3 with AG490 reactivated autophagy and improved survival after intestinal I/R injury. CONCLUSIONS JAK2/STAT3 signaling suppresses autophagy process during intestinal I/R, while inhibiting JAK-STAT can be protective against intestinal I/R injury by activating autophagy. These findings expand our knowledge on intestinal I/R injury and provide therapeutic targets for clinical treatment.
Collapse
|
228
|
Malo ME, Frank C, Khokhoev E, Gorbunov A, Dontsov A, Garg R, Dadachova E. Mitigating effects of sublethal and lethal whole-body gamma irradiation in a mouse model with soluble melanin. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2022; 42:011508. [PMID: 35037901 DOI: 10.1088/1361-6498/ac3dcf] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/25/2021] [Indexed: 05/27/2023]
Abstract
The field of radiation countermeasures is growing, however, currently there are no effective and non-toxic compounds which could be administered orally to the individuals post exposure to high doses of ionising radiation. The pigment melanin is ubiquitous through all kingdoms of life and provides selective advantage under radiation stress through its role as a chemical and physical shield, and its capacity to respond and react to exposures. Soluble allomelanin was administered to mice following whole-body exposure to lethal or sublethal doses of gamma radiation to determine its capacity to mitigate the effects of acute radiation syndrome, and its utility as a radiation countermeasure. Allomelanin has shown a trend to improve survival post an 8 Gy sublethal radiation exposure when administered up to 48 h post-irradiation. Furthermore, it improved median and overall survival to a 10 Gy lethal radiation exposure, specifically when administered at 24 h post-irradiation. Histological analysis on the jejunum region of the small intestine of this treatment group indicated that alterations of the mucosal and submucosal architecture, and disruption of the lymphatic system associated with lethal radiation exposure were mitigated when allomelanin was administered at 24 h post-irradiation. Based on this work soluble allomelanin derived from a fungal source could serve as an easily sourced, cost-effective, and viable countermeasure to accidental radiation exposure and merits further investigation.
Collapse
Affiliation(s)
- Mackenzie E Malo
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Connor Frank
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | | | | | - Alexander Dontsov
- Emanuel Institute of Biochemical Physics, Russian Academy of Science, Moscow, Russia
| | - Ravendra Garg
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
229
|
Xu H, Hou Q, Zhu J, Feng M, Wang P, Pan Y. The protective effect of Escherichia coli Nissle 1917 on the intestinal barrier is mediated by inhibition of RhoA/ROCK2/MLC signaling via TLR-4. Life Sci 2022; 292:120330. [PMID: 35051420 DOI: 10.1016/j.lfs.2022.120330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 02/07/2023]
Abstract
AIMS This study investigated the protective effect of Escherichia coli Nissle 1917 (EcN) on intestinal barrier and the mechanism in the context of acute severe inflammation. MATERIALS AND METHODS In this study, mice received lipopolysaccharide (LPS) intraperitoneal injection with or without EcN administration to construct a mouse model of endotoxemia. Clinical scores, intestinal permeability, inflammatory cytokines and histopathological analysis of four main organs from different groups were assessed. The expression of tight junction proteins and activation of RhoA/ROCK2/MLC signaling were examined using western blotting. The localization of tight junction proteins was examined by immunofluorescence. Caco-2 monolayers with or without TLR-4 knockdown were incubated with EcN or TNF-α/IFN-γ and the monolayer barrier function was assessed by transepithelial electrical resistance (TER) and FITC-dextran 4000 Da (FD-4) flux. The expression of tight junction proteins and activation of RhoA/ROCK2/MLC signaling were examined by western blotting. The localization of tight junction proteins was examined by immunofluorescence. KEY FINDINGS We found that EcN downregulated the RhoA/ROCK2/MLC signaling pathway to preserve barrier function and alleviated systemic inflammation in mouse model. And EcN also protected barrier function of Caco-2 monolayers by inhibiting the activation of RhoA/ROCK2/MLC signaling via TLR-4. SIGNIFICANCE The results indicated that EcN protected the intestinal barrier function in endotoxemia through inhibiting the activation of RhoA/ROCK2/MLC signaling via TLR-4.
Collapse
Affiliation(s)
- Hao Xu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing 100034, People's Republic of China; Translational Cancer Research Center, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing, 100034, People's Republic of China
| | - Qisheng Hou
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing 100034, People's Republic of China
| | - Jing Zhu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing 100034, People's Republic of China
| | - Mei Feng
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing 100034, People's Republic of China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing 100034, People's Republic of China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi Shiku Street, Beijing 100034, People's Republic of China.
| |
Collapse
|
230
|
Yang J, Tang X, Wu Q, Ren P, Yan Y. A Severe Acute Pancreatitis Mouse Model Transited from Mild Symptoms Induced by a “Two-Hit” Strategy with L-Arginine. Life (Basel) 2022; 12:life12010126. [PMID: 35054519 PMCID: PMC8779052 DOI: 10.3390/life12010126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 01/17/2023] Open
Abstract
To develop a severe acute pancreatitis (SAP) model transited from mild symptoms, we investigated a “two-hit” strategy with L-arginine in mice. The mice were intraperitoneally injected with ice-cold L-arginine (4 g/kg) twice at an interval of 1 h on the first day and subjected to the repeated operation 72 h afterwards. The results showed the “two-hit” strategy resulted in the destructive damage and extensive necrosis of acinar cells in the pancreas compared with the “one-hit” model. Meanwhile, excessive levels of pro-inflammatory mediators, namely IL-6 and TNF-α, were released in the serum. Remarkably, additional deleterious effects on multiple organs were observed, including high intestinal permeability, kidney injury, and severe acute lung injury. Therefore, we confirmed that the SAP animal model triggered by a “two-hit” strategy with L-arginine was successfully established, providing a solid foundation for a deeper understanding of SAP initiation and therapy research to prevent worsening of the disease.
Collapse
|
231
|
Liu Z, Qi M, Tian S, Yang Q, Liu J, Wang S, Ji M, Yu R, Zeng S, Li J, Wei Y, Dong W. Ubiquitin-Specific Protease 25 Aggravates Acute Pancreatitis and Acute Pancreatitis-Related Multiple Organ Injury by Destroying Tight Junctions Through Activation of The STAT3 Pathway. Front Cell Dev Biol 2022; 9:806850. [PMID: 35096833 PMCID: PMC8793747 DOI: 10.3389/fcell.2021.806850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Ubiquitin-specific protease 25 (USP25) plays an important role in inflammation and immunity. However, the role of USP25 in acute pancreatitis (AP) is still unclear. To evaluate the role of USP25 in AP, we conducted research on clinical AP patients, USP25wild-type(WT)/USP25 knockout (USP25-/-) mice, and pancreatic acinar cells. Our results showed that serum USP25 concentration was higher in AP patients than in healthy controls and was positively correlated with disease severity. AP patients' serum USP25 levels after treatment were significantly lower than that at the onset of AP. Moreover, USP25 expression was upregulated in cerulein-induced AP in mice, while USP25 deficiency attenuates AP and AP-related multiple organ injury. In vivo and in vitro studies showed that USP25 exacerbates AP by promoting the release of pro-inflammatory factors and destroying tight junctions of the pancreas. We showed that USP25 aggravates AP and AP-related multiple organ injury by activating the signal transducer and activator of transcription 3 (STAT3) pathway. Targeting the action of USP25 may present a potential therapeutic option for treating AP.
Collapse
|
232
|
Kaisinova AS, Uzdenov MB, Fedorov AA, Badakhova DK, Gusova BA, Khodova TV. [Evaluation of resistance to transient occlusion of anterior mesenteric artery under the influence of the course of mineral water enriched with selenium (experimental research)]. VOPROSY KURORTOLOGII, FIZIOTERAPII, I LECHEBNOI FIZICHESKOI KULTURY 2022; 99:50-55. [PMID: 36511467 DOI: 10.17116/kurort20229906250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
OBJECTIVE To evaluate the effect of the preventive course of drinking mineral water enriched with selenium on the processes of resistance to the damaging action of reversible occlusion of the anterior mesenteric artery based on the comparison of intestinal morphological changes in the experiment. MATERIAL AND METHODS There has been modeled ischemic reperfusion injury of the intestinal wall according to H. Ikeda and co-authors using reversible occlusion of the anterior mesenteric artery with 33 outbred male rats. The rats were divided into four groups by block randomization: the 1st group - intact animals (n=7) - without an exposure; the control group - sham operated animals (n=6); the group of comparison (n=7) - with a model-operation; the experimental group (n=11) - animals with a model operation that had courses of intragastric watering of bottled sulfate-chloride-hydrocarbonate-sodium low-mineralized (2.2 g/l) drinking mineral water «Psyzh» enriched with selenium. Biopsies of the small intestine were taken for histological examination. RESULTS Histological examination of the small intestine of experimental animals determines various degrees of severity of damage: on average, the animals of the experimental group on the scale of C.J. Chiu (1970) had the lowest degree of severity of pathological changes, the animals of the group of comparison - 1.4 times higher (p=0.02). That is, the effect of a preventive course of mineral water «Psyzh» enriched with selenium was manifested in the formation of resistance to the damaging effect of reversible occlusion of the anterior mesenteric artery; in the presence of ischemic reperfusion damage to the intestinal wall, comparable in severity to changes with the animals without prevention, the most significant positive effect was realized in the containment of reactive changes. CONCLUSION The effect of the preventive course of drinking mineral water «Psyzh» enriched with selenium manifested itself in the formation of resistance to the damaging effect of reversible occlusion of the anterior mesenteric artery, which is the basis for introducing this technique into clinical practice in order to prevent the development of reperfusion injuries of the intestine.
Collapse
Affiliation(s)
- A S Kaisinova
- North Caucasian Federal Scientific and Clinical Center of Federal Medical and Biological Agency, Essentuki, Russia
- Pyatigorsk Medical and Pharmaceutical Institute - Branch of the Volgograd State Medical University, Pyatigorsk, Russia
| | - M B Uzdenov
- North Caucasian State Academy, Cherkessk, Russia
| | - A A Fedorov
- Ural State Medical University, Yekaterinburg, Russia
- Yekaterinburg Medical and Scientific Center for the Prevention and Protection of Health of Workers' Enterprises, Yekaterinburg, Russia
| | | | - B A Gusova
- Pyatigorsk Medical and Pharmaceutical Institute - Branch of the Volgograd State Medical University, Pyatigorsk, Russia
| | - T V Khodova
- North Caucasian Federal Scientific and Clinical Center of Federal Medical and Biological Agency, Essentuki, Russia
- Pyatigorsk Medical and Pharmaceutical Institute - Branch of the Volgograd State Medical University, Pyatigorsk, Russia
| |
Collapse
|
233
|
Protective Effect of Luminal Uric Acid Against Indomethacin-Induced Enteropathy: Role of Antioxidant Effect and Gut Microbiota. Dig Dis Sci 2022; 67:121-133. [PMID: 33569665 DOI: 10.1007/s10620-021-06848-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Uric acid (UA) has anti- and pro-inflammatory properties. We previously revealed that elevated serum UA levels provide protection against murine small intestinal injury probably via luminal UA secreted in the small intestine. Luminal UA may act as an antioxidant, preventing microbiota vulnerability to oxidative stress. However, whether luminal UA is increased under hyperuricemia and plays a protective role in a dose-dependent manner as well as the mechanism by which luminal UA exerts its protective effects on enteropathy remains unknown. METHODS Inosinic acid (IMP) (1000 mg/kg, i.p.) was administered to obtain high serum UA (HUA) and moderate serum UA (500 mg/kg IMP, i.p.) mice. UA concentrations and levels of oxidative stress markers in the serum and intestine were measured. Mice received indomethacin (20 mg/kg, i.p.) to evaluate the effects of UA on indomethacin-induced enteropathy. Reactive oxygen species (ROS) on the ileal mucosa were analyzed. The fecal microbiota of HUA mice was transplanted to investigate its effect on indomethacin-induced enteropathy. RESULTS IMP increased luminal UA dose-dependently, with higher levels of luminal antioxidant markers. Indomethacin-induced enteropathy was significantly ameliorated in both UA-elevated groups, with decreased indomethacin-induced luminal ROS. The microbiota of HUA mice showed a significant increase in α-diversity and a significant difference in β-diversity from the control. Fecal microbiota transplantation from HUA mice ameliorated indomethacin-induced enteropathy. CONCLUSIONS The protective role of luminal UA in intestinal injury is likely exerted via oxidative stress elimination and microbiota composition modulation, preferably for gut immunity. Therefore, enhancing anaerobic conditions using antioxidants is a potential therapeutic target.
Collapse
|
234
|
Xiao J, Yang Q, Zhang Y, Xu H, Ye Y, Li L, Yang Y, Jin S. Maresin conjugates in tissue regeneration-1 suppresses ferroptosis in septic acute kidney injury. Cell Biosci 2021; 11:221. [PMID: 34961563 PMCID: PMC8711186 DOI: 10.1186/s13578-021-00734-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Background Ferroptosis is unique among different types of regulated cell death and closely related to organ injury. Whether ferroptosis occurs in sepsis-associated acute kidney injury (SA-AKI) is not clear. Nuclear factor-erythroid-2-related factor 2 (Nrf2) is crucial to the regulation of ferroptosis. We and others have shown that Maresin conjugates in tissue regeneration 1 (MCTR1) or other members of specialized pro-resolving mediators (SPMs) can actively regulate inflammation resolution and protect organs against injury in inflammatory diseases by activating the Nrf2 signaling. The aim of this study was to determine whether ferroptosis occurs in SA-AKI. Furthermore, we investigated the potential role and mechanism of MCTR1 in the regulation of ferroptosis in SA-AKI, which mainly focus on the Nrf2 signaling. Results We demonstrated for the first time that ferroptosis is present in SA-AKI. Moreover, MCTR1 effectively suppressed ferroptosis in SA-AKI. Meanwhile, MCTR1 upregulated the expression of Nrf2 in the kidney of septic mice. Nrf2 inhibitor ML-385 reversed MCTR1-regulated ferroptosis and AKI, implying that Nrf2 is involved in the inhibitory effects of MCTR1 on ferroptosis in SA-AKI. Further, MCTR1 inhibited ferroptosis and elevated the expression of Nrf2 in LPS-induced HK-2 cells. However, Nrf2 siRNA offset the effect of MCTR1 on ferroptosis. Finally, we observed that MCTR1 ameliorates multi-organ injury and improves survival in animal models of sepsis. Conclusions These data demonstrate that MCTR1 suppresses ferroptosis in SA-AKI through the Nrf2 signaling. Our study enriches the pathophysiological mechanism of SA-AKI and provides new therapeutic ideas and potential intervention targets for SA-AKI. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00734-x.
Collapse
Affiliation(s)
- Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qian Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ye'an Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Haoran Xu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yang Ye
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Linchao Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
235
|
Zhang YN, Chang ZN, Liu ZM, Wen SH, Zhan YQ, Lai HJ, Zhang HF, Guo Y, Zhang XY. Dexmedetomidine Alleviates Gut-Vascular Barrier Damage and Distant Hepatic Injury Following Intestinal Ischemia/Reperfusion Injury in Mice. Anesth Analg 2021; 134:419-431. [PMID: 34889823 DOI: 10.1213/ane.0000000000005810] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) challenge often results in gut barrier dysfunction and induces distant organ injury. Dexmedetomidine has been shown to protect intestinal epithelial barrier against I/R attack. The present study aims to investigate the degree to which intestinal I/R attack will contribute to gut-vascular barrier (GVB) damage, and to examine the ability of dexmedetomidine to minimize GVB and liver injuries in mice. METHODS In vivo, intestinal ischemic challenge was induced in mice by clamping the superior mesenteric artery for 45 minutes. After clamping, the mice were subjected to reperfusion for either 2, 4, 6, or 12 hours. Intraperitoneal injection of dexmedetomidine 15, 20, or 25 μg·kg-1 was performed intermittently at the phase of reperfusion. For the in vitro experiments, the challenge of oxygen-glucose deprivation/reoxygenation (OGD/R) was established in cultured vascular endothelial cells, and dexmedetomidine (1 nM) was used to treat the cells for 24 hours. Moreover, in vivo and in vitro, SKL2001 (a specific agonist of β-catenin) or XAV939 (a specific inhibitor of β-catenin) was applied to determine the role of β-catenin in the impacts provided by dexmedetomidine. RESULTS The attack of intestinal I/R induced GVB damage. The greatest level of damage was observed at 4 hours after intestinal reperfusion. There was a significant increase in plasmalemma vesicle-associated protein-1 (PV1, a specific biomarker for endothelial permeability) expression (5.477 ± 0.718 vs 1.000 ± 0.149; P < .001), and increased translocation of intestinal macromolecules and bacteria to blood and liver tissues was detected (all P < .001). Liver damages were observed. There were significant increases in histopathological scores, serum parameters, and inflammatory factors (all P < .001). Dexmedetomidine 20 μg·kg-1 reduced PV1 expression (0.466 ± 0.072 vs 1.000 ± 0.098; P < .001) and subsequent liver damages (all P < .01). In vitro, dexmedetomidine significantly improved vascular endothelial cell survival (79.387 ± 6.447% vs 50.535 ± 1.766%; P < .001) and increased the productions of tight junction protein and adherent junction protein (all P < .01) following OGD/R. Importantly, in cultured cells and in mice, β-catenin expression significantly decreased (both P < .001) following challenge. Dexmedetomidine or SKL2001 upregulated β-catenin expression and produced protective effects (all P < .01). However, XAV939 completely eliminated the protective effects of dexmedetomidine on GVB (all P < .001). CONCLUSIONS The disruption of GVB occurred following intestinal I/R. Dexmedetomidine alleviated I/R-induced GVB impairment and subsequent liver damage.
Collapse
Affiliation(s)
| | - Ze-Nan Chang
- Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zi-Meng Liu
- Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | - Han-Jin Lai
- Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Yi Guo
- From the Departments of Anesthesiology
| | | |
Collapse
|
236
|
Jian YP, Yang G, Zhang LH, Liang JY, Zhou HL, Wang YS, Xu ZX. Lactobacillus plantarum alleviates irradiation-induced intestinal injury by activation of FXR-FGF15 signaling in intestinal epithelia. J Cell Physiol 2021; 237:1845-1856. [PMID: 34881818 DOI: 10.1002/jcp.30651] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/23/2022]
Abstract
Abdominal irradiation (IR) may destroy the intestinal mucosal barrier, leading to severe intestinal infection and multiple organ dysfunction syndromes. The role of intestinal microbiota in the development of IR-induced intestinal injury remains largely unknown. Herein, we reported that abdominal IR altered the composition of the microbiota and reduced the abundance and diversity of the gut microbiome. Alterations of bacteria, in particular reduction of Lactobacillus, played a critical role in IR-induced intestinal injury. Fecal microbiota transplant (FMT) from normal mice or administration of Lactobacillus plantarum to intestinal microbiota-eliminated mice substantially reduced IR-induced intestinal damage and prevented mice from IR-induced death. We further characterized that L. plantarum activated the farnesoid X receptor (FXR) - fibroblast growth factor 15 (FGF15) signaling in intestinal epithelial cells and hence promoted DNA-damage repair. Application of GW4064, an activator of FXR, to microbiota eliminated mice markedly mitigated IR-induced intestinal damage, reduced intestinal epithelial cell death and promoted the survival of IR mice. In contrast, suppression of FXR with Gly-β-MCA, a bile acid and an intestine-selective and high-affinity FXR inhibitor, abrogated L. Plantarum-mediated protection on the ileum of IR mice. Taken together, our findings not only provide new insights into the role of intestinal flora in radiation-induced intestinal injury but also shed new light on the application of probiotics for the protection of radiation-damaged individuals.
Collapse
Affiliation(s)
- Yong-Ping Jian
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China
| | - Ge Yang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China
| | - Li-Hong Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China
| | - Ji-Yong Liang
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Hong-Lan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yi-Shu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin, China.,School of Life Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
237
|
Rodriguez-Leon G, Estremiana F, Miro M, Bettonica C, Aranda H, Farran L, de Oca J, Sabench F, Jorba R. Ischemic Conditioning in the Right Colon and Terminal Ileum: An Experimental Rat Model. Eur Surg Res 2021; 63:278-284. [PMID: 34875649 DOI: 10.1159/000520676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/28/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Preoperative gastric ischemic conditioning (IC) improves the outcome of esophageal replacement gastroplasty and is associated with low morbidity. However, when the stomach cannot be used for esophageal replacement, a colonic replacement is required. The study aim was to assess the viability of right colon and terminal ileum IC in a rat model and the histological damage/recovery sequence and determine if neovascularization is a potential adaptive mechanism. METHODS The study was conducted in Rattus norvegicus with ileocolic vascular ligation. Seven groups of animals were established (6 rats per group) with groups defined by the date of their post-IC euthanasia (+1, +3, +6, +10, +15, and +21 days). Comparisons were made with a sham group. Viability of the model was defined as <10% of transmural necrosis. The evaluation of histological damage used the Chiu score in hematoxylin and eosin sections of paraffin-embedded specimens with CD31 immunohistochemical assessment of neovascularization by the median of submucosal vessel counts in 5 high-magnification fields. RESULTS Transmural colon necrosis occurred in 1/36 animals (2.78%) with no animal demonstrating transmural ileal necrosis. The maximum damage was observed in the colon on +1 day post-IC (average Chiu score 1.67, p = 0.015), whereas in the ileum, it was on days +1, +3, and +6 (average Chiu score 1.5, 1.3, and 1.17; p = 0.015, 0.002, and 0.015, respectively). In the +21-day group, histological recovery was complete in the colon in 4 (66.7%) of the 6 animals and in the ileum in 5 (83.3%) of 6 animals. There were no significant differences in quantitative neovascularization in any of the groups when compared with the sham group or when comparisons were made between groups. CONCLUSIONS The tested animal model for IC of the colon and terminal ileum appeared to be feasible. Histological damage was maximal between the 1st and 3rd day following IC, but by day 21, recovery was complete in two-thirds of the rats. There was no evidence in this preliminary IC model that would suggest neovascularization as an adaptive mechanism.
Collapse
Affiliation(s)
| | - Fernando Estremiana
- Esophagogastric Surgery Department, Hospital Universitari Bellvitge, L'Hospitalet de Llobregat, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Monica Miro
- Esophagogastric Surgery Department, Hospital Universitari Bellvitge, L'Hospitalet de Llobregat, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Carla Bettonica
- Esophagogastric Surgery Department, Hospital Universitari Bellvitge, L'Hospitalet de Llobregat, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Humberto Aranda
- Esophagogastric Surgery Department, Hospital Universitari Bellvitge, L'Hospitalet de Llobregat, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Leandre Farran
- Esophagogastric Surgery Department, Hospital Universitari Bellvitge, L'Hospitalet de Llobregat, Spain.,Universitat de Barcelona, Barcelona, Spain
| | | | | | - Rosa Jorba
- Universitat Rovira Virgili, Tarragona, Spain.,General Surgery Department, Hospital Universitary Joan XXIII, Tarragona, Spain
| |
Collapse
|
238
|
Li H, Wang Q, Chen P, Zhou C, Zhang X, Chen L. Ursodeoxycholic Acid Treatment Restores Gut Microbiota and Alleviates Liver Inflammation in Non-Alcoholic Steatohepatitic Mouse Model. Front Pharmacol 2021; 12:788558. [PMID: 34938193 PMCID: PMC8685972 DOI: 10.3389/fphar.2021.788558] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota dysbiosis plays an important role in the progression of non-alcoholic fatty liver disease (NAFLD), and no approved drugs are available for NAFLD treatment. In this study, we aimed to explore the dynamic changes of gut microbiota at the different stages of NAFLD and determine whether ursodeoxycholic acid (UDCA) could improve liver histopathological features of non-alcoholic steatohepatitis (NASH) mice induced by a high-fat high-cholesterol (HFHC) diet and its impact on gut microbiota. 6-week-old male C57BL/6 mice were fed with a HFHC or normal diet for 12, 18, and 24 weeks, respectively, to simulate the different stages of NAFLD. 16s ribosomal RNA genes from mice fecal samples at the different time points were sequenced to evaluate the dynamic changes of the gut microbiota. Then, C57BL/6 mice were fed with a HFHC diet for 24 weeks to establish the NASH model. Different doses of UDCA were administered intragastrically for additional 4 weeks. Normal diet-fed mice were taken as control. Serum samples, liver, and intestine tissues were harvested for biochemical tests and histopathological examinations. 16s ribosomal RNA genes from mice fecal samples were sequenced to assess the structural changes of gut microbiota. HFHC diet-fed mice developed simple steatosis, steatohepatitis, and fibrosis at 12, 18, and 24 weeks, respectively. The profile of gut microbiota dynamically changed with the different stages of NAFLD. NASH mice had significantly higher abundance of Fecalibaculum, Coriobacteriaceae_UCG-002, and Enterorhabdus, and lower abundance of norank_f_Muribaculaceae, Bacteroides, and Alistipes, which were partially restored by UDCA treatment. UDCA treatment significantly attenuated hepatic inflammation of NASH mice as indicated by the sum of ballooning and lobular inflammation of the NALFD activity score (3.2 ± 0.8 vs 1.8 ± 0.8, p = 0.029), and partially restored gut microbiota dysbiosis, and increased the expression of Claudin-1 and ZO-1 in the intestine, but did not activate the suppressed Farnesoid X receptor signal pathway. Conclusions: The gut microbiota dynamically changes with the different stages of NAFLD. UDCA treatment (120 mg/kg) could partially restore gut microbiota, repair gut barrier integrity, and attenuate hepatic inflammation in the NASH mouse model.
Collapse
Affiliation(s)
- Hu Li
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People`s Hospital, Shanghai, China
| | - Qingling Wang
- Department of Infectious Disease, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Department of Infectious Disease, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Peizhan Chen
- Central Laboratory, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenghua Zhou
- Central Laboratory, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinxin Zhang
- Department of Infectious Disease, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Li Chen
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
239
|
Dyer WB, Tung JP, Li Bassi G, Wildi K, Jung JS, Colombo SM, Rozencwajg S, Simonova G, Chiaretti S, Temple FT, Ainola C, Shuker T, Palmieri C, Shander A, Suen JY, Irving DO, Fraser JF. An Ovine Model of Hemorrhagic Shock and Resuscitation, to Assess Recovery of Tissue Oxygen Delivery and Oxygen Debt, and Inform Patient Blood Management. Shock 2021; 56:1080-1091. [PMID: 34014886 DOI: 10.1097/shk.0000000000001805] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Aggressive fluid or blood component transfusion for severe hemorrhagic shock may restore macrocirculatory parameters, but not always improve microcirculatory perfusion and tissue oxygen delivery. We established an ovine model of hemorrhagic shock to systematically assess tissue oxygen delivery and repayment of oxygen debt; appropriate outcomes to guide Patient Blood Management. METHODS Female Dorset-cross sheep were anesthetized, intubated, and subjected to comprehensive macrohemodynamic, regional tissue oxygen saturation (StO2), sublingual capillary imaging, and arterial lactate monitoring confirmed by invasive organ-specific microvascular perfusion, oxygen pressure, and lactate/pyruvate levels in brain, kidney, liver, and skeletal muscle. Shock was induced by stepwise withdrawal of venous blood until MAP was 30 mm Hg, mixed venous oxygen saturation (SvO2) < 60%, and arterial lactate >4 mM. Resuscitation with PlasmaLyte® was dosed to achieve MAP > 65 mm Hg. RESULTS Hemorrhage impacted primary outcomes between baseline and development of shock: MAP 89 ± 5 to 31 ± 5 mm Hg (P < 0.01), SvO2 70 ± 7 to 23 ± 8% (P < 0.05), cerebral regional tissue StO2 77 ± 11 to 65 ± 9% (P < 0.01), peripheral muscle StO2 66 ± 8 to 16 ± 9% (P < 0.01), arterial lactate 1.5 ± 1.0 to 5.1 ± 0.8 mM (P < 0.01), and base excess 1.1 ± 2.2 to -3.6 ± 1.7 mM (P < 0.05). Invasive organ-specific monitoring confirmed reduced tissue oxygen delivery; oxygen tension decreased and lactate increased in all tissues, but moderately in brain. Blood volume replacement with PlasmaLyte® improved primary outcome measures toward baseline, confirmed by organ-specific measures, despite hemoglobin reduced from baseline 10.8 ± 1.2 to 5.9 ± 1.1 g/dL post-resuscitation (P < 0.01). CONCLUSION Non-invasive measures of tissue oxygen delivery and oxygen debt repayment are suitable outcomes to inform Patient Blood Management of hemorrhagic shock, translatable for pre-clinical assessment of novel resuscitation strategies.
Collapse
Affiliation(s)
- Wayne B Dyer
- Australian Red Cross Lifeblood, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - John-Paul Tung
- Australian Red Cross Lifeblood, Brisbane, Australia
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Australia
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Karin Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Cardiovascular Research Institute, Basel, Switzerland
| | - Jae-Seung Jung
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Sebastiano Maria Colombo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milano, Italy
| | - Sacha Rozencwajg
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Sorbonne Université, INSERM, UMRS-1166, ICAN Institute of Cardiometabolism and Nutrition, Medical ICU, Pitié-Salpêtrière University Hospital, Paris, France
| | - Gabriela Simonova
- Australian Red Cross Lifeblood, Brisbane, Australia
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | | | - Fergal T Temple
- Australian Red Cross Lifeblood, Brisbane, Australia
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
| | - Tristan Shuker
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
| | - Chiara Palmieri
- School of Veterinary Science, The University of Queensland, Brisbane, Australia
| | - Aryeh Shander
- Department of Anesthesiology, Critical Care and Hyperbaric Medicine, Englewood Health, Englewood
- TeamHealth, Englewood Health, Englewood
- UF College of Medicine, University of Florida, Gainesville
- Department of Anesthesiology, Medicine and Surgery, Icahn School of Medicine, Mount Sinai Hospital, New York
- Department of Anesthesiology and Critical Care, Rutgers University, Newark
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - David O Irving
- Australian Red Cross Lifeblood, Sydney, Australia
- Faculty of Health, University of Technology, Sydney, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
240
|
Chen Y, Zhang H, Ji S, Jia P, Chen Y, Li Y, Wang T. Resveratrol and its derivative pterostilbene attenuate oxidative stress-induced intestinal injury by improving mitochondrial redox homeostasis and function via SIRT1 signaling. Free Radic Biol Med 2021; 177:1-14. [PMID: 34648904 DOI: 10.1016/j.freeradbiomed.2021.10.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/05/2021] [Accepted: 10/10/2021] [Indexed: 12/12/2022]
Abstract
Oxidative stress inflicts mitochondrial dysfunction, which has been recognized as a key driver of intestinal diseases. Resveratrol (RSV) and its derivative pterostilbene (PTS) are natural antioxidants and exert a protective influence on intestinal health. However, the therapeutic effects and mechanisms of RSV and PTS on oxidative stress-induced mitochondrial dysfunction and intestinal injury remain unclear. The present study used porcine and cellular settings to compare the effects of RSV and PTS on mitochondrial redox homeostasis and function to alleviate oxidative stress-induced intestinal injury. Our results indicated that PTS was more potent than RSV in reducing oxidative stress, maintaining intestinal integrity, and preserving the mitochondrial function of diquat-challenged piglets. In the in vitro study, RSV and PTS protected against hydrogen peroxide (H2O2)-induced mitochondrial dysfunction in intestinal porcine enterocyte cell line (IPEC-J2) by facilitating mitochondrial biogenesis and increasing the activities of mitochondrial complexes. In addition, both RSV and PTS efficiently mitigated mitochondrial oxidative stress by increasing sirtuin 3 protein expression and the deacetylation of superoxide dismutase 2 and peroxiredoxin 3 in H2O2-exposed IPEC-J2 cells. Furthermore, RSV and PTS preserved mitochondrial membrane potential, which restrained the release of cytochrome C from mitochondria to the cytoplasm and caspase-3 activation and further reduced apoptotic rates in H2O2-exposed IPEC-J2 cells. Mechanistically, depletion of sirtuin 1 (SIRT1) abrogated RSV's and PTS's benefits against mitochondrial reactive oxygen species overproduction, mitochondrial dysfunction, and apoptosis in H2O2-exposed IPEC-J2 cells, suggesting that SIRT1 was required for RSV and PTS to protect against oxidative stress-induced intestinal injury. In conclusion, RSV and PTS improve oxidative stress-induced intestinal injury by regulating mitochondrial redox homeostasis and function via SIRT1 signaling pathway. In offering this protection, PTS is superior to RSV.
Collapse
Affiliation(s)
- Yanan Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Hao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Shuli Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Peilu Jia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Yueping Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Yue Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
241
|
Age-Dependent Intestinal Repair: Implications for Foals with Severe Colic. Animals (Basel) 2021; 11:ani11123337. [PMID: 34944114 PMCID: PMC8697879 DOI: 10.3390/ani11123337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022] Open
Abstract
Colic is a leading cause of death in horses, with the most fatal form being strangulating obstruction which directly damages the intestinal barrier. Following surgical intervention, it is imperative that the intestinal barrier rapidly repairs to prevent translocation of gut bacteria and their products and ensure survival of the patient. Age-related disparities in survival have been noted in many species, including horses, humans, and pigs, with younger patients suffering poorer clinical outcomes. Maintenance and repair of the intestinal barrier is regulated by a complex mucosal microenvironment, of which the ENS, and particularly a developing network of subepithelial enteric glial cells, may be of particular importance in neonates with colic. Postnatal development of an immature enteric glial cell network is thought to be driven by the microbial colonization of the gut and therefore modulated by diet-influenced changes in bacterial populations early in life. Here, we review the current understanding of the roles of the gut microbiome, nutrition, stress, and the ENS in maturation of intestinal repair mechanisms after foaling and how this may influence age-dependent outcomes in equine colic cases.
Collapse
|
242
|
Anderson KM, Gayer CP. The Pathophysiology of Farnesoid X Receptor (FXR) in the GI Tract: Inflammation, Barrier Function and Innate Immunity. Cells 2021; 10:cells10113206. [PMID: 34831429 PMCID: PMC8624027 DOI: 10.3390/cells10113206] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
The Farnesoid-X Receptor, FXR, is a nuclear bile acid receptor. Its originally described function is in bile acid synthesis and regulation within the liver. More recently, however, FXR has been increasingly appreciated for its breadth of function and expression across multiple organ systems, including the intestine. While FXR’s role within the liver continues to be investigated, increasing literature indicates that FXR has important roles in responding to inflammation, maintaining intestinal epithelial barrier function, and regulating immunity within the gastrointestinal (GI) tract. Given the complicated and multi-factorial nature of intestinal barrier dysfunction, it is not surprising that FXR’s role appears equally complicated and not without conflicting data in different model systems. Recent work has suggested translational applications of FXR modulation in GI pathology; however, a better understanding of FXR physiology is necessary for these treatments to gain widespread use in human disease. This review aims to discuss current scientific work on the role of FXR within the GI tract, specifically in its role in intestinal inflammation, barrier function, and immune response, while also exploring areas of controversy.
Collapse
Affiliation(s)
- Kemp M. Anderson
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Christopher P. Gayer
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Division of Pediatric Surgery, Childrens Hospital Los Angeles, Los Angeles, CA 90027, USA
- Correspondence: ; Tel.: +1-323-361-4974
| |
Collapse
|
243
|
Zhang Q, Liu XM, Hu Q, Liu ZR, Liu ZY, Zhang HG, Huang YL, Chen QH, Wang WX, Zhang XK. Dexmedetomidine inhibits mitochondria damage and apoptosis of enteric glial cells in experimental intestinal ischemia/reperfusion injury via SIRT3-dependent PINK1/HDAC3/p53 pathway. J Transl Med 2021; 19:463. [PMID: 34772407 PMCID: PMC8588684 DOI: 10.1186/s12967-021-03027-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) injury commonly occurs during perioperative periods, resulting in high morbidity and mortality on a global scale. Dexmedetomidine (Dex) is a selective α2-agonist that is frequently applied during perioperative periods for its analgesia effect; however, its ability to provide protection against intestinal I/R injury and underlying molecular mechanisms remain unclear. METHODS To fill this gap, the protection of Dex against I/R injury was examined in a rat model of intestinal I/R injury and in an inflammation cell model, which was induced by tumor necrosis factor-alpha (TNF-α) plus interferon-gamma (IFN-γ) stimulation. RESULTS Our data demonstrated that Dex had protective effects against intestinal I/R injury in rats. Dex was also found to promote mitophagy and inhibit apoptosis of enteric glial cells (EGCs) in the inflammation cell model. PINK1 downregulated p53 expression by promoting the phosphorylation of HDAC3. Further studies revealed that Dex provided protection against experimentally induced intestinal I/R injury in rats, while enhancing mitophagy, and suppressing apoptosis of EGCs through SIRT3-mediated PINK1/HDAC3/p53 pathway in the inflammation cell model. CONCLUSION Hence, these findings provide evidence supporting the protective effect of Dex against intestinal I/R injury and its underlying mechanism involving the SIRT3/PINK1/HDAC3/p53 axis.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xiao-Ming Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Qian Hu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zheng-Ren Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Zhi-Yi Liu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Huai-Gen Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yuan-Lu Huang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Qiu-Hong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Wen-Xiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xue-Kang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
244
|
Zhenzhen L, Wenting L, Jianmin Z, Guangru Z, Disheng L, Zhiyu Z, Feng C, Yajing S, Yingxiang H, Jipeng L, Zhanhai W, Yan Z, Xin L, Yongqiang L, Yufang L. miR-146a-5p/TXNIP axis attenuates intestinal ischemia-reperfusion injury by inhibiting autophagy via the PRKAA/mTOR signaling pathway. Biochem Pharmacol 2021; 197:114839. [PMID: 34774846 DOI: 10.1016/j.bcp.2021.114839] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 11/02/2022]
Abstract
Autophagy is being increasingly recognized as an important regulator of intestinal ischemia-reperfusion(I/R)injury, but its exact role is still debated. Emerging evidence suggests that miR-146a-5p is involved in the initiation and development of I/R injury, but its role in intestinal I/R injury remains unclear. The present study generated an intestinal I/R mouse model and an oxygen glucose deprivation/reoxygenation (OGD/R) Caco-2 cell model and found that autophagy was increased and contributed to the intestinal injury and cell death induced by I/R and OGD/R. In addition, in both I/R and OGD/R models, the miR-146a-5p expression level was decreased and accompanied by an increase in TXNIP expression. By transfecting cells with an miR-146a-5p inhibitor or mimic, we observed that miR-146a-5p inhibits autophagy during OGD/R by targeting TXNIP; this was confirmed by the dual luciferase reporter gene assay. Additionally, through overexpression and knockdown cell lines, we established that TXNIP regulates autophagy during intestinal I/R via the PRKAA/mTOR pathway. The interaction between TXNIP and p-PRKAA was verified by immunofluorescence co-localization and immunoprecipitation assays. Moreover, we confirmed that TXNIP is indispensable for miR-146a-5p-mediated cell protection. Finally, we observed that miR-146a-5p overexpression inhibits autophagy and attenuates intestinal I/R injury via the PRKAA/mTOR pathway by targeting TXNIP in vivo. In conclusion, this study highlights the role of miR-146a-5p in regulating autophagy by targeting TXNIP, suggesting that miR-146a-5p may be a novel drug target for intestinal I/R therapy.
Collapse
Affiliation(s)
- Liu Zhenzhen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Leng Wenting
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhang Jianmin
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhang Guangru
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Liu Disheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhao Zhiyu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Chen Feng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shi Yajing
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hao Yingxiang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Lv Jipeng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Wan Zhanhai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Zhang Yan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Liu Xin
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Liu Yongqiang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Leng Yufang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China; The Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
245
|
Zhu Y, Wang A, Li R, Zhu H, Hu L, Chen W. Total ginsenosides promote the IEC-6 cell proliferation via affecting the regulatory mechanism mediated by polyamines. Saudi Pharm J 2021; 29:1223-1232. [PMID: 34744477 PMCID: PMC8551508 DOI: 10.1016/j.jsps.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/11/2021] [Indexed: 01/17/2023] Open
Abstract
Epithelial cell proliferation has been demonstrated to be a critical modality for mucosal repair after gastrointestinal mucosal injury. This research aimed to investigate the effect of total ginsenosides upon the proliferation of intestinal epithelial cells (IEC-6), and elucidate its potential mechanisms through polyamine-regulated pathway including the expression of proliferation-related proteins. Total ginsenosides (PGE3) were extracted from Panax ginseng, a Chinese herbal medicine, whose chromatogram was obtained by high performance liquid chromatographic method with evaporative light scattering detection (HPLC-ELSD). The cell proliferation, cell cycle distribution and the level of c-Myc, RhoA, Cdk2 proteins were detected to determine the effects of PGE3 at 25, 50 and100 mg/l doses on IEC-6. Furthermore, rats model of intestinal mucosal injury were induced by the subcutaneous injection of indomethacin, and the effect of Panax ginseng aqueous extracts (PGE1) on intestinal mucosal injury was observed. PGE3 could promote IEC-6 cell proliferation, reduce the proportion of G0/G1 phase cells and elevate the proportion of G2/M + S phase cells, and revert the proliferation and cell cycle arrest induced by DFMO (DL-a-difluoromethylornithine, an inhibitor of polyamines synthesis). PGE3 exposure enhanced the level of c-Myc, RhoA and Cdk2 proteins, and reversed the inhibition of these proteins expression induced by DFMO. The results of gross and pathological scores showed administration of PGE1 significantly alleviated intestinal mucosal injury of rats. Our findings indicate that total ginsenosides promoted the IEC-6 proliferation presumably via its regulation on cell cycle and the expression of proliferation-related proteins regulated by polyamines, and provided a novel perspective for exploring the repair effect of Panax ginseng upon gastrointestinal mucosal injury.
Collapse
Affiliation(s)
| | | | - Ruliu Li
- Corresponding author at: Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang road, Guang zhou 510405, PR China.
| | | | | | | |
Collapse
|
246
|
Wu J, Yin Y, Qin M, Li K, Liu F, Zhou X, Song X, Li B. Vagus Nerve Stimulation Protects Enterocyte Glycocalyx After Hemorrhagic Shock Via the Cholinergic Anti-Inflammatory Pathway. Shock 2021; 56:832-839. [PMID: 33927140 PMCID: PMC8519159 DOI: 10.1097/shk.0000000000001791] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/08/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Electrical vagal nerve stimulation is known to decrease gut permeability and alleviate gut injury caused by traumatic hemorrhagic shock. However, the specific mechanism of action remains unclear. Glycocalyx, located on the surface of the intestinal epithelium, is associated with the buildup of the intestinal barrier. Therefore, the goal of our study was to explore whether vagal nerve stimulation affects enterocyte glycocalyx, gut permeability, gut injury, and remote lung injury. MATERIALS AND METHODS Male Sprague Dawley rats were anesthetized and their cervical nerves were exposed. The rats underwent traumatic hemorrhagic shock (with maintenance of mean arterial pressure of 30-35 mmHg for 60 min) with fluid resuscitation. Vagal nerve stimulation was added to two cohorts of animals before fluid resuscitation, and one of them was injected with methyllycaconitine to block the cholinergic anti-inflammatory pathway. Intestinal epithelial glycocalyx was detected using immunofluorescence. Intestinal permeability, the degree of gut and lung injury, and inflammation factors were also assessed. RESULTS Vagal nerve stimulation alleviated the damage to the intestinal epithelial glycocalyx and decreased intestinal permeability by 43% compared with the shock/resuscitation phase (P < 0.05). Methyllycaconitine partly eliminated the effects of vagal nerve stimulation on the intestinal epithelial glycocalyx (P < 0.05). Vagal nerve stimulation protected against traumatic hemorrhagic shock/fluid resuscitation-induced gut and lung injury, and some inflammatory factor levels in the gut and lung tissue were downregulated after vagal nerve stimulation (P < 0.05). CONCLUSIONS Vagal nerve stimulation could relieve traumatic hemorrhagic shock/fluid resuscitation-induced intestinal epithelial glycocalyx damage via the cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Juan Wu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yushuang Yin
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Mingzhe Qin
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Kun Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Fang Liu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Xiang Zhou
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Xiaoyang Song
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
| | - Bixi Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
247
|
Hanawa Y, Higashiyama M, Kurihara C, Tanemoto R, Ito S, Mizoguchi A, Nishii S, Wada A, Inaba K, Sugihara N, Horiuchi K, Okada Y, Narimatsu K, Komoto S, Tomita K, Hokari R. Acesulfame potassium induces dysbiosis and intestinal injury with enhanced lymphocyte migration to intestinal mucosa. J Gastroenterol Hepatol 2021; 36:3140-3148. [PMID: 34368996 DOI: 10.1111/jgh.15654] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/01/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM The artificial sweetener acesulfame potassium (ACK) is officially approved as safe for intake and has been used in processed foods. However, ACKs have been reported to induce metabolic syndrome, along with alteration of the gut microbiota in mice. In recent years, studies have suggested that this artificial sweetener promotes myeloperoxidase reactivity in Crohn's disease-like ileitis. We aimed to investigate the effect of ACK on the intestinal mucosa and gut microbiota of normal mice. METHODS Acesulfame potassium was administered to C57BL/6J mice (8 weeks old) via free drinking. Intestinal damage was evaluated histologically, and messenger RNA (mRNA) levels of TNF-α, IFN-γ, IL1-β, MAdCAM-1, GLP1R, and GLP2R were determined with quantitative reverse transcription polymerase chain reaction (qRT-PCR). Immunohistochemistry was performed to examine the expression of MAdCAM-1 in the small intestine. The composition of gut microbiota was assessed using high-throughput sequencing. We performed intravital microscopic observation to examine if ACK altered lymphocyte migration to the intestinal microvessels. RESULTS Acesulfame potassium increased the expression of proinflammatory cytokines, decreased the expression of GLP-1R and GLP-2R, and induced small intestinal injury with an increase in intestinal permeability, and ACK treatment induced microbial changes, but the transfer of feces alone from ACK mice did not reproduce intestinal damage in recipient mice. ACK treatment significantly increased the migration of lymphocytes to intestinal microvessels. CONCLUSION Acesulfame potassium induces dysbiosis and intestinal injury with enhanced lymphocyte migration to intestinal mucosa. Massive use of non-caloric artificial sweeteners may not be as safe as we think.
Collapse
Affiliation(s)
- Yoshinori Hanawa
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Masaaki Higashiyama
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Chie Kurihara
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Rina Tanemoto
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Suguru Ito
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Akinori Mizoguchi
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Shin Nishii
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Akinori Wada
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Kenichi Inaba
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Nao Sugihara
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Kazuki Horiuchi
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Yoshikiyo Okada
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Kazuyuki Narimatsu
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Shunsuke Komoto
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Kengo Tomita
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| |
Collapse
|
248
|
Chen G, Zeng H, Li X, Liu J, Li Z, Xu R, Ma Y, Liu C, Xue B. Activation of G protein coupled estrogen receptor prevents chemotherapy-induced intestinal mucositis by inhibiting the DNA damage in crypt cell in an extracellular signal-regulated kinase 1- and 2- dependent manner. Cell Death Dis 2021; 12:1034. [PMID: 34718327 PMCID: PMC8557214 DOI: 10.1038/s41419-021-04325-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a common adverse reaction to antineoplastic treatment with few appropriate, specific interventions. We aimed to identify the role of the G protein coupled estrogen receptor (GPER) in CIM and its mechanism. Adult male C57BL/6 mice were intraperitoneally injected with 5-fluorouracil to establish the CIM model. The selective GPER agonist G-1 significantly inhibited weight loss and histological damage in CIM mice and restored mucosal barrier dysfunction, including improving the expression of ZO-1, increasing the number of goblet cells, and decreasing mucosal permeability. Moreover, G-1 treatment did not alter the antitumor effect of 5-fluorouracil. In the CIM model, G-1 therapy reduced the expression of proapoptotic protein and cyclin D1 and cyclin B1, reversed the changes in the number of TUNEL+ cells, Ki67+ and bromodeoxyuridine+ cells in crypts. The selective GPER antagonist G15 eliminated all of the above effects caused by G-1 on CIM, and application of G15 alone increased the severity of CIM. GPER was predominantly expressed in ileal crypts, and G-1 inhibited the DNA damage induced by 5-fluorouracil in vivo and vitro, as confirmed by the decrease in the number of γH2AX+ cells in the crypts and the comet assay results. Referring to the data from GEO dataset we verified GPER activation restored ERK1/2 activity in CIM and 5-fluorouracil-treated IEC-6 cells. Once the effects of G-1 on ERK1/2 activity were abolished with the ERK1/2 inhibitor PD0325901, the effects of G-1 on DNA damage both in vivo and in vitro were eliminated. Correspondingly, all of the manifestations of G-1 protection against CIM were inhibited by PD0325901, such as body weight and histological changes, the mucosal barrier, the apoptosis and proliferation of crypt cells. In conclusion, GPER activation prevents CIM by inhibiting crypt cell DNA damage in an ERK1/2-dependent manner, suggesting GPER might be a target preventing CIM.
Collapse
Affiliation(s)
- Guanyu Chen
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Honghui Zeng
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinyun Li
- The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Jianbo Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhao Li
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Runze Xu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuntao Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chuanyong Liu
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bing Xue
- Department of Physiology and Pathophysiology, School of basic medical science, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
249
|
Huang J, Huang J, Li Y, Lv H, Yin T, Fan S, Zhang C, Li H. Fucoidan Protects Against High-Fat Diet-Induced Obesity and Modulates Gut Microbiota in Institute of Cancer Research Mice. J Med Food 2021; 24:1058-1067. [PMID: 34668763 DOI: 10.1089/jmf.2021.k.0030] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Fucoidan possesses various biological activities, such as anticoagulant, immunomodulatory, anti-inflammatory, potential antioxidant, and others. In this study, we investigated the effect of fucoidan on high-fat diet-induced obesity, inflammation, and gut microbiota in Institute of Cancer Research mice. Mice were gavaged with 50 mg/(kg·d) (Fuc0.5 group) or 250 mg/(kg·d) (Fuc2.5 group) of fucoidan for 5 weeks. Fucoidan alleviated obesity and tissue damage by decreasing body weight and body mass index, decreasing body weight gain, improved organ index, liver steatosis, and improved the structure of the small intestine. In addition, fucoidan decreased total cholesterol, triglyceride, and low-density lipoprotein cholesterol, and increased high-density lipoprotein cholesterol. Moreover, fucoidan reduced serum lipopolysaccharide concentrations, tumor necrosis factor-α, and total bile acid. Furthermore, fucoidan improved the structure of gut microbiota and significantly increased the abundance (Shannon diversity index, evenness, and Faecalibacterium prausnitzii) determined by denaturing gradient gel electrophoresis and quantitative PCR. In conclusion, our study provides a scientific basis for fucoidan as a functional food for modulating the gut microbiota and protecting against obesity.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Department of Pediatrics, Xijing Hospital, Air Force the Fourth Military Medical University, Xi'an, China
| | - Juan Huang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yao Li
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Huiyun Lv
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Tianyi Yin
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shujun Fan
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Caihua Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Huajun Li
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
250
|
Strbe S, Gojkovic S, Krezic I, Zizek H, Vranes H, Barisic I, Strinic D, Orct T, Vukojevic J, Ilic S, Lovric E, Muzinic D, Kolenc D, Filipčić I, Zoricic Z, Marcinko D, Boban Blagaic A, Skrtic A, Seiwerth S, Sikiric P. Over-Dose Lithium Toxicity as an Occlusive-like Syndrome in Rats and Gastric Pentadecapeptide BPC 157. Biomedicines 2021; 9:1506. [PMID: 34829735 PMCID: PMC8615292 DOI: 10.3390/biomedicines9111506] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 12/15/2022] Open
Abstract
Due to endothelial impairment, high-dose lithium may produce an occlusive-like syndrome, comparable to permanent occlusion of major vessel-induced syndromes in rats; intracranial, portal, and caval hypertension, and aortal hypotension; multi-organ dysfunction syndrome; brain, heart, lung, liver, kidney, and gastrointestinal lesions; arterial and venous thrombosis; and tissue oxidative stress. Stable gastric pentadecapeptide BPC 157 may be a means of therapy via activating loops (bypassing vessel occlusion) and counteracting major occlusion syndromes. Recently, BPC 157 counteracted the lithium sulfate regimen in rats (500 mg/kg/day, ip, for 3 days, with assessment at 210 min after each administration of lithium) and its severe syndrome (muscular weakness and prostration, reduced muscle fibers, myocardial infarction, and edema of various brain areas). Subsequently, BPC 157 also counteracted the lithium-induced occlusive-like syndrome; rapidly counteracted brain swelling and intracranial (superior sagittal sinus) hypertension, portal hypertension, and aortal hypotension, which otherwise would persist; counteracted vessel failure; abrogated congestion of the inferior caval and superior mesenteric veins; reversed azygos vein failure; and mitigated thrombosis (superior mesenteric vein and artery), congestion of the stomach, and major hemorrhagic lesions. Both regimens of BPC 157 administration also counteracted the previously described muscular weakness and prostration (as shown in microscopic and ECG recordings), myocardial congestion and infarction, in addition to edema and lesions in various brain areas; marked dilatation and central venous congestion in the liver; large areas of congestion and hemorrhage in the lung; and degeneration of proximal and distal tubules with cytoplasmic vacuolization in the kidney, attenuating oxidative stress. Thus, BPC 157 therapy overwhelmed high-dose lithium intoxication in rats.
Collapse
Affiliation(s)
- Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Slaven Gojkovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Helena Zizek
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Hrvoje Vranes
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Ivan Barisic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Dean Strinic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Tatjana Orct
- Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia; (T.O.); (D.M.)
| | - Jaksa Vukojevic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Spomenko Ilic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Eva Lovric
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (D.K.); (S.S.)
| | - Darija Muzinic
- Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia; (T.O.); (D.M.)
| | - Danijela Kolenc
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (D.K.); (S.S.)
| | - Igor Filipčić
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Zoran Zoricic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Darko Marcinko
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| | - Anita Skrtic
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (D.K.); (S.S.)
| | - Sven Seiwerth
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (E.L.); (D.K.); (S.S.)
| | - Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (S.S.); (S.G.); (I.K.); (H.Z.); (H.V.); (I.B.); (D.S.); (J.V.); (S.I.); (I.F.); (Z.Z.); (D.M.); (A.B.B.)
| |
Collapse
|