1
|
Li Y, Li J, Liu X, Cheng Z, Gao N, Kang J, Wang X. Polygonatum sibiricum Polysaccharides Alleviate LPS-Induced Liver Injury in Chicks by Suppressing Inflammation and Oxidative Stress Through the PPAR Signaling Pathway. Antioxidants (Basel) 2025; 14:418. [PMID: 40298760 PMCID: PMC12024365 DOI: 10.3390/antiox14040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Polygonatum sibiricum polysaccharides (PSPs), plant-derived polysaccharides widely used in the pharmaceutical field, exhibit various biological activities, including anti-inflammatory and antioxidant effects. However, research on their application in chicks remains limited. Therefore, the aim of this study is to investigate the protective mechanism of PSP against liver injury in chicks using an LPS-induced inflammatory model. A total of 200 one-day-old Hy-Line Brown laying chicks were randomly assigned to five groups (40 chicks each): a control group (CON) fed a basal diet, an LPS group, and three PSP groups receiving low (250 mg/L), medium (500 mg/L), and high (1000 mg/L) doses of PSP (PSP250_LPS, PSP500_LPS, and PSP1000_LPS, respectively). The experiment lasted 21 days. During this period, the LPS and PSP groups were intraperitoneally injected with 1500 μg/kg LPS on days 14, 16, 18, and 20, while the CON group received normal saline. On day 21, organs were collected for analysis. The results indicated that PSP treatment significantly reduced the liver and kidney indices that were elevated by LPS (p < 0.05) without affecting the indices of the spleen, thymus, or bursa of Fabricius (p > 0.05). Histological analysis revealed that PSP alleviated LPS-induced ballooning degeneration and cell swelling in hepatocytes. Furthermore, PSP treatment decreased the levels of ALT and AST and significantly mitigated increases in the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α while enhancing the level of the anti-inflammatory cytokine IL-10 (p < 0.05). Transcriptome sequencing of liver samples revealed that LPS significantly altered the expression of 10 genes in the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which were regulated by PSP intervention. qPCR validation supported these findings. Furthermore, biochemical analyses of liver tissue showed that PSP alleviated oxidative stress by affecting levels of SOD, MDA, NADPH, ROS, and H2O2. In conclusion, PSP may alleviate LPS-induced liver injury in chicks by modulating the PPAR signaling pathway. These findings provide valuable insights for promoting healthy chick rearing and ensuring the safe supply of poultry products.
Collapse
Affiliation(s)
- Yang Li
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| | - Jian Li
- Hebei Provincial Animal Husbandry Station, Shijiazhuang 050011, China; (J.L.); (X.L.); (Z.C.)
| | - Xiaowang Liu
- Hebei Provincial Animal Husbandry Station, Shijiazhuang 050011, China; (J.L.); (X.L.); (Z.C.)
| | - Zhili Cheng
- Hebei Provincial Animal Husbandry Station, Shijiazhuang 050011, China; (J.L.); (X.L.); (Z.C.)
| | - Nana Gao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| | - Jungang Kang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| | - Xiaodan Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| |
Collapse
|
2
|
Yang N, Guo J, Zhang J, Gao S, Xiang Q, Wen J, Huang Y, Rao C, Chen Y. A toxicological review of alkaloids. Drug Chem Toxicol 2024; 47:1267-1281. [PMID: 38465444 DOI: 10.1080/01480545.2024.2326051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
Alkaloids are naturally occurring compounds with complex structures found in natural plants. To further improve the understanding of plant alkaloids, this review focuses on the classification, toxicity and mechanisms of action, providing insight into the occurrence of alkaloid-poisoning events and guiding the safe use of alkaloids in food, supplements and clinical applications. Based on their chemical structure, alkaloids can be divided into organic amines, diterpenoids, pyridines, isoquinolines, indoles, pyrrolidines, steroids, imidazoles and purines. The mechanisms of toxicity of alkaloids, including neurotoxicity, hepatoxicity, nephrotoxicity, cardiotoxicity and cytotoxicity, have also been reviewed. Some cases of alkaloid poisoning have been introduced when used as food or clinically, including accidental food poisoning, excessive consumption, and poisoning caused by the improper use of alkaloids in a clinical setting, and the importance of safety evaluation was illustrated. This review summarizes the toxicity and mechanism of action of alkaloids and provides evidence for the need for the safe use of alkaloids in food, supplements and clinical applications.
Collapse
Affiliation(s)
- Nannan Yang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jiafu Guo
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jian Zhang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Song Gao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiwen Xiang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jiayu Wen
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yan Huang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Chaolong Rao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yan Chen
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Andreis A, Dossi FC, De Ferrari GM, Alunni G, Imazio M. Anakinra-Dependent Recurrent Pericarditis: The Role of the R202Q Variant of the MEFV Gene. J Clin Med 2024; 13:6051. [PMID: 39458001 PMCID: PMC11508427 DOI: 10.3390/jcm13206051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Background: the role of the R202Q (c.605G>A, p.Arg202Gln) missense variant of the MEFV gene has been debated as either a benign polymorphism or a potentially pathogenic mutation. We report and discuss here the case of a young female with corticosteroid-dependent recurrent pericarditis carrying the homozygous R202Q variant, exhibiting distinctive clinical features possibly influenced by this genetic variant. Methods: a 30-year-old woman with a previous diagnosis of cancer and recent respiratory infection presented with severe pleuritic chest pain, hypotension, tachycardia, and fever. Initial diagnostic evaluation indicated cardiac tamponade, and emergent pericardiocentesis was performed. Despite initial treatment with NSAIDs, colchicine, and corticosteroids, the patient experienced multiple recurrences. Genetic testing identified homozygous R202Q variant in the MEFV gene. Given the corticosteroid dependency and recurrent nature of her condition, IL-1 inhibitor anakinra was introduced, leading to significant improvement, although tapering below 150 mg per week failed to prevent recurrences. Results: the introduction of anakinra resulted in rapid symptom relief and resolution of pericardial effusion. However, attempts to taper or discontinue anakinra led to pericarditis recurrences. Ultimately, a maintenance dose of 50 mg every three days was established, which maintained remission for 18 months without recurrence. Despite multiple tapering attempts, further reduction in anakinra dosage was unsuccessful without triggering relapses. Conclusions: the R202Q variant, although typically considered benign, may contribute to an autoinflammatory phenotype resembling familial Mediterranean fever. This case underscores the potential pathogenicity of the homozygous R202Q variant in recurrent pericarditis and its responsiveness to IL-1 inhibition. In patients with corticosteroid-dependent recurrent pericarditis, genetic testing for the R202Q variant should be considered when anti-IL-1 drugs cannot be withdrawn. Further studies are warranted to elucidate the variant's role in pericardial inflammation and guide personalized treatment strategies.
Collapse
Affiliation(s)
- Alessandro Andreis
- Advanced Cardiovascular Echocardiography Unit, Cardiovascular and Thoracic Department, Città della Salute e della Scienza di Torino University Hospital, 10126 Turin, Italy
- Division of Cardiology, Città della Salute e della Scienza di Torino University Hospital, 10126 Turin, Italy
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Federica Currò Dossi
- Division of Cardiology, Città della Salute e della Scienza di Torino University Hospital, 10126 Turin, Italy
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Gaetano Maria De Ferrari
- Division of Cardiology, Città della Salute e della Scienza di Torino University Hospital, 10126 Turin, Italy
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Gianluca Alunni
- Advanced Cardiovascular Echocardiography Unit, Cardiovascular and Thoracic Department, Città della Salute e della Scienza di Torino University Hospital, 10126 Turin, Italy
| | - Massimo Imazio
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy
- Cardiothoracic Department, University Hospital Santa Maria della Misericordia, 33100 Udine, Italy
| |
Collapse
|
4
|
Madanchi M, Young M, Tersalvi G, Maria Cioffi G, Attinger-Toller A, Cuculi F, Kurmann R, Bossard M. The impact of colchicine on patients with acute and chronic coronary artery disease. Eur J Intern Med 2024; 125:1-9. [PMID: 38238134 DOI: 10.1016/j.ejim.2024.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 07/04/2024]
Abstract
Inflammation plays a central role in coronary artery disease (CAD), and recent data have shown that anti-inflammatory drugs have the potential to reduce ischemic events in CAD patients. Colchicine is an ancient anti-inflammatory drug that targets neutrophil and inflammasome activities. It has been prescribed for decades for different rheumatological conditions. Given the important role of inflammation in the development of cardiovascular disease, there has been considerable interest in studying colchicine's potential to limit the progression of atherosclerosis among afflicted patients. In fact, there is a growing body of randomized data suggesting that use of low-dose colchicine reduces the risk of ischemic events in patients with CAD, particularly repeated revascularizations, new myocardial infarctions and strokes. This review article summarizes background information-including possible side effects and contraindications-as well as the current evidence backing up the use of colchicine in patients with established CAD.
Collapse
Affiliation(s)
- Mehdi Madanchi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland
| | - Mabelle Young
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland
| | - Gregorio Tersalvi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland
| | - Giacomo Maria Cioffi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland
| | - Adrian Attinger-Toller
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland
| | - Florim Cuculi
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland
| | - Reto Kurmann
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland; Department of Cardiovascular Diseases, Mayo Clinic Rochester, MN, USA
| | - Matthias Bossard
- Cardiology Division, Heart Center, Luzerner Kantonsspital, Spitalstrasse 16, 6000 Lucerne, Switzerland.
| |
Collapse
|
5
|
Saha B, A T R, Adhikary S, Banerjee A, Radhakrishnan AK, Duttaroy AK, Pathak S. Exploring the Relationship Between Diet, Lifestyle and Gut Microbiome in Colorectal Cancer Development: A Recent Update. Nutr Cancer 2024; 76:789-814. [PMID: 39207359 DOI: 10.1080/01635581.2024.2367266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/18/2024] [Accepted: 06/05/2024] [Indexed: 09/04/2024]
Abstract
Colorectal cancer (CRC) is one of the major causes of cancer-related mortality worldwide. Despite advances in treatment modalities, its prevalence continues to rise, notably among younger populations. Unhealthy dietary habits, sedentary routines, and obesity have been identified as one of the key contributors to the development of colorectal cancer, apart from genetic and epigenetic modifications. Recognizing the profound impact of diet and lifestyle on the intricate gut microbiota ecosystem offers a promising avenue for understanding CRC development and its treatment. Gut dysbiosis, characterized by imbalances favoring harmful microbes over beneficial ones, has emerged as a defining feature of CRC. Changes in diet and lifestyle can profoundly alter the composition of gut microbes and the metabolites they produce, potentially contributing to CRC onset. Focusing on recent evidence, this review discussed various dietary factors, such as high consumption of red and processed meats and low fiber intake, and lifestyle factors, including obesity, lack of physical activity, smoking, and excessive alcohol consumption, that influence the gut microbiome composition and elevate CRC risk.
Collapse
Affiliation(s)
- Biki Saha
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Rithi A T
- Department of Pharmacology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Subhamay Adhikary
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Arun Kumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Surajit Pathak
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| |
Collapse
|
6
|
Rusnak T, Azarcoya-Barrera J, Makarowski A, Jacobs RL, Richard C. Plant- and Animal-Derived Dietary Sources of Phosphatidylcholine Have Differential Effects on Immune Function in The Context of A High-Fat Diet in Male Wistar Rats. J Nutr 2024; 154:1936-1944. [PMID: 38582387 PMCID: PMC11217025 DOI: 10.1016/j.tjnut.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/20/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Phosphatidylcholine (PC) derived from eggs has been shown to beneficially modulate T cell response and intestinal permeability under the context of a high-fat diet. OBJECTIVES The objective of this study was to determine whether there is a differential effect of plant and animal-derived sources of PC on immune function. METHODS Four-week-old male Wistar rats were randomly assigned to consume 1 of 4 diets (n = 10/group) for 12 wk, all containing 1.5 g of total choline/kg of diet but differing in choline forms: 1-Control Low-Fat [CLF, 20% fat, 100% free choline (FC)]; 2-Control High-Fat (CHF, 50% fat, 100% FC); 3-High-Fat Egg-derived PC (EPC, 50% fat, 100% Egg-PC); 4-High-Fat Soy-derived PC (SPC, 50% fat, 100% Soy-PC). Immune cell functions and phenotypes were measured in splenocytes by ex vivo cytokine production after mitogen stimulation and flow cytometry, respectively. RESULTS The SPC diet increased splenocyte IL-2 production after PMA+I stimulation compared with the CHF diet. However, the SPC group had a lower proportion of splenocytes expressing the IL-2 receptor (CD25+, P < 0.05). After PMA+I stimulation, feeding EPC normalized splenocyte production of IL-10 relative to the CLF diet, whereas SPC did not (P < 0.05). In mesenteric lymph node lymphocytes, the SPC diet group produced more IL-2 and TNF-α after PMA+I stimulation than the CHF diet, whereas the EPC diet group did not. CONCLUSIONS Our results suggest that both egg- and soy-derived PC may attenuate high-fat diet-induced T cell dysfunction. However, egg-PC enhances, to a greater extent, IL-10, a cytokine involved in promoting the resolution phase of inflammation, whereas soy-PC appears to elicit a greater effect on gut-associated immune responses.
Collapse
Affiliation(s)
- Tianna Rusnak
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
Zeng J, Li Y, Zou Y, Yang Y, Yang T, Zhou Y. Intestinal toxicity alleviation and efficacy potentiation through therapeutic administration of Lactobacillus paracasei GY-1 in the treatment of gout flares with colchicine. Food Funct 2024; 15:1671-1688. [PMID: 38251779 DOI: 10.1039/d3fo04858f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Gout flares have emerged as a significant public health concern. Colchicine (COL) is a first-line and standard drug for treating gout flares. However, its clinical use is limited due to various adverse effects. Besides, COL fails to adequately meet the needs of patients, particularly young patients. In this study, we investigate the therapeutic administration of Lactobacillus paracasei GY-1 (GY-1) to overcome the limitations of COL. Our results demonstrate that GY-1 attenuates COL toxicity in terms of body weight loss, decreased feed intake, mortality, reduced locomotor activity, colon shortening, increased oxidative stress, histological damage, and impaired gut permeability. Meanwhile, we demonstrate that GY-1 enhances the therapeutic effect for gout flares when combined with COL, as evidenced by the reduction in paw swelling, decreased levels of proinflammatory cytokines including IL-1β and TNF-α, and an increase in the anti-inflammatory cytokine IL-10. Additionally, the absolute quantification of the gut microbiota shows that GY-1 restores the gut microbiota imbalance caused by COL. Furthermore, GY-1 reduces the abundance of 4 Alistipes species and 6 Porphyromonadaceae species, which may be responsible for toxicity alleviation. At the same time, GY-1 increases the abundance of Bacteroides sartorii and Enterococcus sp., which may contribute to its therapeutic efficacy. This study demonstrates the feasibility of developing probiotic-based adjuvant therapy or bacteriotherapy for treating gout flares. To our knowledge, GY-1 is the first probiotic that could be used as an alternative synergetic agent with COL for the therapeutic treatment of gout flares.
Collapse
Affiliation(s)
- Jiaqi Zeng
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Yan Li
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Yizhi Zou
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Ying Yang
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, China
| | - Tingting Yang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Yizhuang Zhou
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
8
|
Zhou QX, Zhou Q, Zhang P, Xie YQ, Yang ZY, Tan WH, Khan A, Duan WG, Zhou ZH, Liu L. Integrating multi-level interactive network and in vivo/vitro studies to explore the protective mechanism of Ampelopsis grossedentata in hyperuricemia. Fitoterapia 2024; 172:105718. [PMID: 37931719 DOI: 10.1016/j.fitote.2023.105718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
The strategies or drugs for preventing and treating Hyperuricemia (HUA) are still lacking. As a traditional Chinese medicine (TCM) with a profound history, Ampelopsis grossedentata has been shown to play diverse biological roles. The purpose of the present study was to evaluate hypouricemic effect of A. grossedentata, and investigate its involved material basis and mechanism. A HUA mice model was established to evaluate the therapeutic effects of A. grossedentata. And then some extracts from A. grossedentata were prepared, isolated and analyzed. Furthermore, network pharmacology, based on the above results, was used to discover potential active ingredients and therapeutic targets, and they were further verified and explored by molecular docking and in vitro experiments. In vivo experiments showed that A. grossedentata exerted hypouricemic effect on mice of HUA. The core active ingredients (quercetin, myricetin and dihydromyricetin etc.) and core targets (PTGS2, XOD and ABCG2 etc.) for A. grossedentata to treat HUA were predicted by network pharmacology. And molecular docking showed that the spontaneous binding activities of above components and targets were marvelous. In vitro experiments further demonstrated that A. grossedentata exerted hypouricemic effect by decreasing the levels of UA, XOD, antioxidant factors, inflammatory factors, GLUT9 and URAT1 in HK-2 cells of HUA. Taken together, this study integrates multi-level interaction network with in vivo/vitro experiments to systematically reveal the material basis and mechanism of A. grossedentata in treating HUA, which provides a scientific basis for further study of A. grossedentata and HUA.
Collapse
Affiliation(s)
- Qi-Xiu Zhou
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Qian Zhou
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Peng Zhang
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yan-Qing Xie
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Zhu-Ya Yang
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Wen-Hong Tan
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Wei-Gang Duan
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Zhi-Hong Zhou
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lu Liu
- Yunnan Yunzhong Institute of Nutrition and Health, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
9
|
He F, Xie T, Ni D, Tang T, Cheng X. Efficacy and safety of inhibiting the NLRP3/IL-1β/IL-6 pathway in patients with ST-elevation myocardial infarction: A meta-analysis. Eur J Clin Invest 2023; 53:e14062. [PMID: 37427709 DOI: 10.1111/eci.14062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND The NLRP3/IL-1β/IL-6 pathway plays a key role in mediating inflammatory responses after ST-elevation myocardial infarction (STEMI). However, the clinical benefits of inhibiting this pathway in STEMI are uncertain. We aimed to evaluate the efficacy and safety of inhibiting the NLRP3/IL-1β/IL-6 pathway in STEMI patients. METHODS This study followed PRISMA guidelines. PubMed, Embase, CENTRAL and ClinicalTrials.gov databases were searched for randomized controlled trials (RCTs) of inhibiting the NLRP3/IL-1β/IL-6 pathway in STEMI patients within 7 days of symptom onset. The efficacy outcomes included all-cause death, cardiovascular death, recurrent MI, new-onset or worsening heart failure (HF) and stroke. The safety outcomes were serious infection, gastrointestinal adverse events and injection site reactions. RESULTS Of 316 screened records, nine trials with 1211 patients were included in the meta-analysis. Colchicine reduced the risk of recurrent MI (RR 0.28, 95% CI 0.10-0.74; I2 = 0.0%). Anakinra was associated with reduced risk of new-onset or worsening HF (RR 0.32, 95% CI 0.13-0.77; I2 = 0.0%) and decreased C-reactive protein levels (SMD -1.34, 95% CI -2.04 to -0.65; I2 = 0.0%). Colchicine and anakinra increased the risk of gastrointestinal adverse events (RR 4.43, 95% CI 2.75-7.13; I2 = 38.1%) and injection site reactions (RR 4.52, 95% CI 1.32-15.49; I2 = 0.8%), respectively. None of the three medications affected the risks of all-cause death, cardiovascular death, stroke and serious infection. CONCLUSIONS There is still no large-scale RCT evidence on the efficacy and safety of inhibiting the NLRP3/IL-1β/IL-6 pathway for the treatment of STEMI. Preliminary results from the available RCTs suggest colchicine and anakinra may respectively reduce the risks of recurrent MI and new-onset or worsening HF. The available RCTs in this meta-analysis lack power to determine any differences on mortality.
Collapse
Affiliation(s)
- Fang He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Ni
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Ying H, Guo W, Tang X, Pan J, Yu P, Fan H, Wang X, Jiang R, Jiang C, Liang P. Colchicine attenuates the electrical remodeling of post-operative atrial fibrillation through inhibited expression of immune-related hub genes and stabilization of microtubules. Int J Biol Sci 2023; 19:2934-2956. [PMID: 37324937 PMCID: PMC10266076 DOI: 10.7150/ijbs.81961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Rationale: Acute inflammation is a major risk factor for post-operative atrial fibrillation (POAF), and epicardial adipose tissue (EAT) is considered as a source of inflammatory mediators. However, underlying mechanisms and pharmacological targets of POAF are poorly understood. Methods: Integrative analysis of array data from EAT and right atrial appendage (RAA) samples was conducted to identify potential hub genes. Lipopolysaccharide (LPS)-stimulated inflammatory models in mice and in induced pluripotent stem cell-derived atrial cardiomyocytes (iPSC-aCMs) were used to examine the exact mechanism underlying POAF. Electrophysiological analysis, multi-electrode array, and Ca2+ imaging was employed to explore the alterations of electrophysiology and Ca2+ homeostasis under inflammation. Flow cytometry analysis, histology and immunochemistry were performed to investigate immunological alterations. Results: We observed electrical remodeling, enhanced atrial fibrillation (AF) susceptibility, immune cell activation, inflammatory infiltration, and fibrosis in LPS-stimulated mice. LPS-stimulated iPSC-aCMs showed arrhythmias, abnormal Ca2+ signaling, reduced cell viability, disrupted microtubule network and increased α-tubulin degradation. VEGFA, EGFR, MMP9 and CCL2 were identified as hub genes simultaneously targeted in the EAT and RAA of POAF patients. Notably, treatment of colchicine in LPS-stimulated mice resulted in a U-shape dose-response curve, where greatly improved survival rates were observed only at doses between 0.10-0.40 mg/kg. At this therapeutic dose level, colchicine inhibited the expression of all the identified hub genes and effectively rescued the pathogenic phenotypes observed in LPS-stimulated mice and iPSC-aCM models. Conclusions: Acute inflammation promotes α-tubulin degradation, induces electrical remodeling, and both recruits and facilitates the infiltration of circulating myeloid cells. A certain dose of colchicine attenuates electrical remodeling and decreases the recurrence of AF.
Collapse
Affiliation(s)
- Hangying Ying
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou 310016, China
| | - Wenpu Guo
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou 310016, China
| | - Xiaomei Tang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou 310016, China
| | - Jun Pan
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310009, China
| | - Pengcheng Yu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou 310016, China
| | - Hangping Fan
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Xiaochen Wang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Ruhong Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou 310016, China
| | - Chenyang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou 310016, China
| | - Ping Liang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
11
|
Shi Y, Li J, Wang J, Cao H, Tian H, Yu F, Gao L. Colchicine increases intestinal toxic load by disturbing fecal metabolome homeostasis in mice. Chem Biol Interact 2022; 368:110193. [PMID: 36179773 DOI: 10.1016/j.cbi.2022.110193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/07/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022]
Abstract
Colchicine (COL) has been used to treat gout for over a millennium, but its medicinal use has been controversial due to its potent toxicity in the gastrointestinal tract. Nausea, vomiting, and diarrhea are the most prominent external manifestations of COL gastrointestinal toxicity, but the cause of these adverse events remains obscure. In this study, the mice were exposed to COL (2.5 mg/kg b.w./day) for one week to study the mechanism of COL-induced diarrhea from the perspective of intestinal metabolism. The results showed that COL exposure disturbed intestinal metabolic homeostasis, resulting in a significant accumulation of 116 metabolites and, conversely, significant depletion of 64 metabolites, with the number of differential metabolites being one-eighth of the total metabolites (180/1445). Also, it was found that cAMP, Adenosine 5'-monophosphate, GDP, Inositol, and Cortisol are core metabolites that play crucial roles in COL-induced metabolic disorders. These metabolites could be used as biomarkers to differentiate control and COL-treated groups, implying that these metabolites may be closely related to COL-induced diarrhea. Furthermore, changes in the metabolic pathways (Purine metabolism, biosynthesis and metabolism of aromatic amino acids, and Bile secretion) involved in these five core metabolites increased the toxic load in the gut, which was the culprit leading to intestinal metabolic disorders. In addition, the abnormal bile secretion caused by COL exposure may play an important role in COL-induced diarrhea. In conclusion, our study opens new avenues for understanding the mechanisms of COL-induced gastrointestinal adverse reactions and broadens the scientific horizon on the interactions between COL and host metabolism.
Collapse
Affiliation(s)
- Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiande Li
- College of Life Science, Northwest Normal University, Lanzhou, 730000, China
| | - Ji Wang
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Huanbing Tian
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - FeiFei Yu
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
12
|
Multidirectional analysis for a colchicine poisoning case revealed detail cause of death and its mechanism. Leg Med (Tokyo) 2022; 58:102092. [DOI: 10.1016/j.legalmed.2022.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/22/2022]
|
13
|
Ivnitsky JJ, Schäfer TV, Rejniuk VL, Vakunenkova OA. Secondary Dysfunction of the Intestinal Barrier in the Pathogenesis of Complications of Acute Poisoning. J EVOL BIOCHEM PHYS+ 2022; 58:1075-1098. [PMID: 36061072 PMCID: PMC9420239 DOI: 10.1134/s0022093022040123] [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: 04/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
Abstract
The last decade has been marked by an exponential increase
in the number of publications on the physiological role of the normal
human gut microbiota. The idea of a symbiotic relationship between
the human organism and normal microbiota of its gastrointestinal
tract has been firmly established as an integral part of the current
biomedical paradigm. However, the type of this symbiosis varies
from mutualism to parasitism and depends on the functional state
of the host organism. Damage caused to the organism by external
agents can lead to the emergence of conditionally pathogenic properties
in the normal gut microbiota, mediated by humoral factors and affecting
the outcome of exogenous exposure. Among the substances produced
by symbiotic microbiota, there are an indefinite number of compounds
with systemic toxicity. Some occur in the intestinal chyme in potentially
lethal amounts in the case they enter the bloodstream quickly. The quick
entry of potential toxicants is prevented by the intestinal barrier
(IB), a set of structural elements separating the intestinal chyme
from the blood. Hypothetically, severe damage to the IB caused by
exogenous toxicants can trigger a leakage and subsequent systemic
redistribution of toxic substances of bacterial origin. Until recently,
the impact of such a redistribution on the outcome of acute exogenous
poisoning remained outside the view of toxicology. The present review
addresses causal relationships between the secondary dysfunction
of the IB and complications of acute poisoning. We characterize
acute systemic toxicity of such waste products of the normal gut microflora
as ammonia and endotoxins, and demonstrate their involvement in
the formation of such complications of acute poisoning as shock,
sepsis, cerebral insufficiency and secondary lung injuries. The
principles of assessing the functional state of the IB and the approaches
to its protection in acute poisoning are briefly considered.
Collapse
Affiliation(s)
- Ju. Ju. Ivnitsky
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| | - T. V. Schäfer
- State Scientific Research Test Institute of Military Medicine, Ministry of Defense of the Russian Federation, St. Petersburg, Russia
| | - V. L. Rejniuk
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| | - O. A. Vakunenkova
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| |
Collapse
|
14
|
A New Insight into Toxicity of Colchicine Analogues by Molecular Docking Analysis Based on Intestinal Tight Junction Protein ZO-1. Molecules 2022; 27:molecules27061797. [PMID: 35335160 PMCID: PMC8955668 DOI: 10.3390/molecules27061797] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Colchicine (COL) is a well-known plant alkaloid long used for medical purposes due to the selective anti-inflammatory effect on acute gouty arthritis. It is also a kind of mitosis toxin with strong inhibitory effects of cell division and is therefore being applied to the treatment of various cancers. However, this product shows a variety of adverse effects that are significantly correlated with the dosage and have attracted much attention. For the first time, the present work obtained a new insight into the gastrointestinal toxicity of colchicine analogues by molecular docking analysis, which was based on the 3D structure of intestinal tight junction protein ZO-1 and the ligand library containing dozens of small-molecule compounds with the basic skeleton of COL and its metabolites. The binding energy and mode of protein–ligand interaction were investigated to better understand the structure–toxicity relationships of COL analogues and the mechanism of action as well. Cluster analysis clearly demonstrated the strong correlation between the binding energy and toxicity of ligand molecules. The interaction mode further revealed that the hydrogen bonding (via the C-7 amide or C-9 carbonyl group) and hydrophobic effect (at ring A or C) were both responsible for ZO-1-related gastrointestinal toxicity of COL analogues, while metabolic transformation via phase I and/or phase II reaction would significantly attenuate the gastrointestinal toxicity of colchicine, indicating an effective detoxication pathway through metabolism.
Collapse
|
15
|
Liu YQ, Lu XX, Hu KX, Peng XB, Jiang Y, Han LM, Ma ZQ, Peng MF, Wan K, Zhang XG, Qiu ZW. Dynamic observation of bone marrow suppression and chromosomal aberrations in patients with acute colchicine poisoning. World J Emerg Med 2022; 13:130-134. [PMID: 35237367 DOI: 10.5847/wjem.j.1920-8642.2022.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/26/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Yan-Qing Liu
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xiao-Xia Lu
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China.,Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100850, China
| | - Kai-Xun Hu
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xiao-Bo Peng
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yun Jiang
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Li-Mei Han
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Zhi-Qiang Ma
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Ming-Fei Peng
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Kun Wan
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xi-Gang Zhang
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Ze-Wu Qiu
- Poisoning Treatment Department, Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| |
Collapse
|
16
|
Keiner M, Dreller V, Klemm I, Lehmann H. [Accidental colchicine intoxication in a cross-breed dog]. TIERARZTLICHE PRAXIS. AUSGABE K, KLEINTIERE/HEIMTIERE 2022; 50:46-56. [PMID: 35235962 DOI: 10.1055/a-1696-3847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A 2-year old cross-breed dog presented due to acute vomiting and progressive lethargy following ingestion of the owner's anti-gout medication (colchicine, 0.35 mg/kg) 1-3 hours prior to presentation.The dog developed signs of all 3 stages of colchicine poisoning (gastrointestinal phase, multi-organ phase, recovery phase) and the clinical course was complicated by the presence of multi-organ dysfunction syndrome (MODS) and numerous negative prognostic factors.This case report describes the clinical and laboratory effects of colchicine poisoning and represents the first successful treatment of an accidental colchicine ingestion in a dog in Europe.
Collapse
Affiliation(s)
- Miriam Keiner
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, Justus-Liebig-Universität Gießen
| | - Vanessa Dreller
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, Justus-Liebig-Universität Gießen
| | - Isabelle Klemm
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, Justus-Liebig-Universität Gießen
| | - Hendrik Lehmann
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, Justus-Liebig-Universität Gießen
| |
Collapse
|
17
|
Hafiane A, Daskalopoulou SS. Targeting the Residual Cardiovascular Risk by Specific Anti-inflammatory Interventions as a Therapeutic Strategy in Atherosclerosis. Pharmacol Res 2022; 178:106157. [DOI: 10.1016/j.phrs.2022.106157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022]
|
18
|
Wang L, Zhang P, Li C, Xu F, Chen J. A polysaccharide from Rosa roxburghii Tratt fruit attenuates high-fat diet-induced intestinal barrier dysfunction and inflammation in mice by modulating the gut microbiota. Food Funct 2021; 13:530-547. [PMID: 34932054 DOI: 10.1039/d1fo03190b] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity-induced colonic inflammation-stimulated colitis is one of the main causes of colorectal cancer. Dietary polysaccharides are considered an effective agent for relieving obesity-induced inflammatory diseases such as diabetes and colitis. In this work, the protective effects of a polysaccharide (RTFP) extracted from Rosa roxburghii Tratt fruit on barrier dysfunction and inflammation were investigated using obesity-induced colitis model mice. RTFP treatment repaired intestinal barrier dysfunction by increasing the expression of tight junction proteins (ZO-1, claudin-1, and occludin) and reducing the levels of inflammatory cytokines, intestinal permeability, and colonic oxidative stress in mice fed a high-fat diet. Most significantly, RTFP decreased gut inflammation and ameliorated the metabolic dysbiosis of intestinal microflora by decreasing the Firmicutes/Bacteroidetes ratio, reducing the levels of serum D-lactic acid and lipopolysaccharides, and inhibiting the TLR4/NF-κB signaling pathway. Furthermore, RTFP significantly increased the abundance of beneficial bacteria (Ruminococcaceae, Muribaculaceae, Akkermansiaceae, etc.) but decreased the abundance of pathogenic bacteria. These findings indicate that RTFP can be used as a natural anti-inflammatory agent to reduce chronic obesity-induced colitis.
Collapse
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Pan Zhang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Chao Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Fei Xu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Jie Chen
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| |
Collapse
|
19
|
Wang K, Chen X, Zuo L, Pan C, Liu G, Zhang X, Du J, Zhang C, Zhang B, Wang Z, Li M, Zhang A, Jiang N. Dietary sodium butyrate protects lipopolysaccharide-induced inflammatory response in lambs through inhibiting TLR4/NF-κB signalling pathway. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1955627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kexin Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Xiang Chen
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Lijun Zuo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Chunyuan Pan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Gan Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Xinyu Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Jiahua Du
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Cunhao Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Bofu Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Zhao Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Muyang Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Aizhong Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| | - Ning Jiang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Feed Resource Efficient Utilization and Nutrition Manipulation in Cold Region of Heilongjiang Province, Daqing, China
| |
Collapse
|
20
|
Andreis A, Imazio M, Casula M, Avondo S, De Ferrari GM. Colchicine efficacy and safety for the treatment of cardiovascular diseases. Intern Emerg Med 2021; 16:1691-1700. [PMID: 33704674 PMCID: PMC7947153 DOI: 10.1007/s11739-021-02654-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/22/2021] [Indexed: 12/24/2022]
Abstract
The emerging role of colchicine in the treatment of cardiovascular diseases is a strong demand for a comprehensive understanding of its efficacy and safety. This meta-analysis and systematic review aimed to study the efficacy in the reduction of adverse cardiovascular outcomes (CO), and the risk of colchicine-related adverse events (CRAEs). Fourteen thousand and nine eighty three patients from 22 randomized controlled trials (RCTs) were included, 9 in patients with coronary artery disease-CAD, 9 in patients with pericarditis, 4 in patients with atrial fibrillation-AF or heart failure. Colchicine was efficacious in the reduction of adverse CO across different settings: pericardial diseases (reduced risk of recurrent pericarditis, 17.6% vs. 35%, RR 0.50, 95% CI 0.41-0.61), CAD (reduced risk of cardiac death, myocardial infarction, stroke,coronary revascularization or hospitalization, 6.1% vs. 8.5%, RR 0.73, 95% CI 0.64-0.83), AF (reduced risk of arrhythmia recurrence, 14.2% vs. 22.7%, RR 0.62, 95% CI 0.44-0.88). Colchicine was associated with increased risk of gastrointestinal CRAEs (11.2% vs. 8.8%, RR 1.87, 95% CI 1.41-2.47) and drug discontinuation (5.4% vs. 3.7%, RR 1.58, 95% CI 1.25-1.99). In both cases, the risk was proportional to the daily dose or duration of treatment, possibly due to early drug discontinuation or tolerance. Other CRAEs (muscle-related, liver,hematologic,cutaneous, infections) were not increased by colchicine, as long as all-cause death (2.2% vs. 1.9%, RR 1.11, 95% CI 0.79-1.54) or non-cardiovascular death (1.5% vs. 1%, RR 1.43, 95% CI 0.93-2.19). Colchicine is efficacious and safe for the treatment of cardiovascular diseases. The risk of gastrointestinal CRAEs and drug discontinuation is not significant if colchicine is used at lower doses (0.5 mg daily) or for longer periods of time (> 6 months).
Collapse
Affiliation(s)
- Alessandro Andreis
- Departement of Medical Sciences, A.O.U. Città della Salute e della Scienza di Torino, University Cardiology, Corso Bramante 88, 10126, Turin, Italy
| | - Massimo Imazio
- Departement of Medical Sciences, A.O.U. Città della Salute e della Scienza di Torino, University Cardiology, Corso Bramante 88, 10126, Turin, Italy.
| | - Matteo Casula
- Departement of Medical Sciences, A.O.U. Città della Salute e della Scienza di Torino, University Cardiology, Corso Bramante 88, 10126, Turin, Italy
| | - Stefano Avondo
- Departement of Medical Sciences, A.O.U. Città della Salute e della Scienza di Torino, University Cardiology, Corso Bramante 88, 10126, Turin, Italy
| | - Gaetano Maria De Ferrari
- Departement of Medical Sciences, A.O.U. Città della Salute e della Scienza di Torino, University Cardiology, Corso Bramante 88, 10126, Turin, Italy
| |
Collapse
|
21
|
Shi Y, Cai H, Niu Z, Li J, Pan G, Tian H, Wei L, Chen L, Yang P, Wang J, Cao H, Gao L. Acute oral colchicine caused gastric mucosal injury and disturbance of associated microbiota in mice. Toxicology 2021; 461:152908. [PMID: 34453961 DOI: 10.1016/j.tox.2021.152908] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/22/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022]
Abstract
Colchicine (COL), an ancient and well-known drug, has been used in clinical practice for centuries. On the other hand, COL has also attracted extensive concerns for its potent toxic effects, especially gastrointestinal adverse reactions (nausea, vomiting, and diarrhea) before clinical symptoms relief. In this study, we used a rodent model to study the effects of COL on gastric mucosa and associated microbiota. The mice were exposed to various concentrations of COL (0.1, 0.5, and 2.5 mg kg-1 body weight per day) for 7 days, and the results showed that COL treatment caused severe gastric mucosal damage, accompanied by a significant decrease in gastric mucosal proinflammatory cytokines (IL-1β, IL-6, and TNF-α). The 16S rRNA gene sequencing revealed that COL significantly perturbed the gastric microbiota composition and reduced the gastric microbiota diversity in mice. Also, we identified bacterial biomarkers associated with diarrhea, including phylum Firmicutes, class Bacilli, order Lactobacillales, family Lactobacillaceae, genu Lactobacillus, and genu Blautia, suggesting that COL-triggered adverse reactions are closely related to gastric microbial perturbations. Our findings open new paths for understanding the mechanism of COL-related adverse gastrointestinal reactions, broadening the scientific view on the interaction between drugs and host gastrointestinal microbiota.
Collapse
Affiliation(s)
- Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hui Cai
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
| | - Zhanyu Niu
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jiande Li
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Gaowei Pan
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Huanbing Tian
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Li Wei
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Linchi Chen
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Pengfei Yang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Ji Wang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
22
|
Kofler T, Kurmann R, Lehnick D, Cioffi GM, Chandran S, Attinger-Toller A, Toggweiler S, Kobza R, Moccetti F, Cuculi F, Jolly SS, Bossard M. Colchicine in Patients With Coronary Artery Disease: A Systematic Review and Meta-Analysis of Randomized Trials. J Am Heart Assoc 2021; 10:e021198. [PMID: 34369166 PMCID: PMC8475038 DOI: 10.1161/jaha.121.021198] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Inflammation plays a pivotal role in coronary artery disease (CAD). The anti‐inflammatory drug colchicine seems to reduce ischemic events in patients with CAD. So far there is equipoise about its safety and impact on mortality. Methods and Results To evaluate the utility of colchicine in patients with acute and chronic CAD, we performed a systematic review and meta‐analysis. MEDLINE, EMBASE, Cochrane CENTRAL and conference abstracts were searched from January 1975 to October 2020. Randomized trials assessing colchicine compared with placebo/standard therapy in patients with CAD were included. Data were combined using random‐effects models. The reliability of the available data was tested using trial sequential analyses . Of 3108 citations, 13 randomized trials (n=13 125) were included. Colchicine versus placebo/standard therapy in patients with CAD reduced risk of myocardial infarction (odds ratio [OR] 0.64; 95% CI, 0.46–0.90; P=0.01; I2 41%) and stroke/transient ischemic attack (OR 0.50; 95% CI, 0.31–0.81; P=0.005; I2 0%). But treatment with colchicine compared with placebo/standard therapy had no influence on all‐cause and cardiovascular mortality (OR 0.96; 95% CI, 0.65–1.41; P=0.83; I2 24%; and OR 0.82; 95% CI, 0.55–1.22; P=0.45; I2 0%, respectively). Colchicine increased the risk for gastrointestinal side effects (P<0.001). According to trial sequential analyses, there is only sufficient evidence for a myocardial infarction risk reduction with colchicine. Conclusions Among patients with CAD, colchicine reduces the risk of myocardial infarction and stroke, but has a higher rate of gastrointestinal upset with no influence on all‐cause mortality.
Collapse
Affiliation(s)
- Thomas Kofler
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| | - Reto Kurmann
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| | - Dirk Lehnick
- Department of Biostatistics and Methodology CTU-CS University of Lucerne Lucerne Switzerland
| | | | - Sujay Chandran
- Royal Sussex County Hospital Sussex Worthing United Kingdom
| | | | - Stefan Toggweiler
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| | - Richard Kobza
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| | - Federico Moccetti
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| | - Florim Cuculi
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| | - Sanjit S Jolly
- McMaster UniversityHamilton Health Sciences Hamilton Ontario Canada
| | - Matthias Bossard
- Cardiology Division Heart Center Luzerner Kantonsspital Lucerne Switzerland
| |
Collapse
|
23
|
Lu X, Liu Y, Wang C, Dong J, Bai L, Zhang C, Zhang R, Sun C, Qiu Z. Pathogenic characteristics and treatment in 43 cases of acute colchicine poisoning. Toxicol Res (Camb) 2021; 10:885-892. [PMID: 34484680 PMCID: PMC8403592 DOI: 10.1093/toxres/tfab074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 07/05/2021] [Accepted: 07/10/2021] [Indexed: 11/12/2022] Open
Abstract
Colchicine poisoning is complicated and has a high mortality rate. The aim of this study was to identify the pathogenic characteristics of colchicine poisoning cases and to propose a comprehensive treatment procedure. A total of 43 patients were divided into survival (n = 32) and death groups (n = 11) according to prognosis. The clinical data (basic information, clinical manifestations, laboratory tests, examination results, therapeutic schedule, response evaluation, and prognosis) were analyzed, and the comprehensive treatment was proposed. The ingestion doses were ≤0.5, 0.5-0.8, and ≥0.8 mg/kg, and the survival rates were 100, 83.33, and 28.60%. The causes of death were cardiovascular and bone marrow hematopoietic failures. We found that the order of organ damage was digestive tract, coagulation, muscle, heart, hematopoietic, lung, liver, and kidney, while the recovery order was digestive tract, coagulation, heart, hematopoietic, lung, muscle, kidney, and liver. Different doses of recombinant human granulocyte colony-stimulating factor and recombinant human thrombopoietin can shorten the severity and duration of neutropenia and thrombocytopenia. Plasma exchange combined with continuous veno-venous hemodialysis filtration treatment can increase survival time. The prognosis is positively correlated with the dose. Early removal of toxicants from the digestive tract and blood is essential. It is vital to give comprehensive treatment of multiple organ injuries, include the use of recombinant human granulocyte colony-stimulating factor, recombinant human thrombopoietin, plasma exchange, and continuous veno-venous hemodialysis filtration.
Collapse
Affiliation(s)
- Xiaoxia Lu
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
- Academy of Military Medical Sciences, Academy of Military Sciences, No. 27 North Taiping Road, Beijing 100850, China
| | - Yanqing Liu
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Chunyan Wang
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Jianguang Dong
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Lili Bai
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Chengcheng Zhang
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Renzheng Zhang
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Chengwen Sun
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| | - Zewu Qiu
- Poisoning Treatment Department, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, No. 8 Dong da Street, Fengtai District, Beijing 100071, China
| |
Collapse
|
24
|
Andreis A, Imazio M, Avondo S, Casula M, Paneva E, Piroli F, De Ferrari GM. Adverse events of colchicine for cardiovascular diseases: a comprehensive meta-analysis of 14 188 patients from 21 randomized controlled trials. J Cardiovasc Med (Hagerstown) 2021; 22:637-644. [PMID: 33399344 DOI: 10.2459/jcm.0000000000001157] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS Colchicine has an emerging role in the cardiovascular field, although, concerns for side effects, especially gastrointestinal, limit its prescription. We aimed at evaluating reported side effects of colchicine for cardiovascular indications. METHODS We performed a meta-analysis of published randomized controlled trials on colchicine for the treatment of cardiovascular diseases. Random-effects meta-analysis was used to assess the risk of adverse events and drug withdrawal. Publication bias was assessed using the Egger test, and meta-regression was performed to assess sources of heterogeneity. RESULTS Among 14 188 patients, 7136 patients received colchicine while the other 7052 received placebo. The occurrence of any adverse event with colchicine was reported in 15.3 vs. 13.9% patients [relative risk (RR) 1.26, 95% confidence interval (CI) 0.96-1.64, P = 0.09]. Gastrointestinal events were reported in 16.1 vs. 12.2% (RR 2.16, 95% CI 1.50-3.12, P < 0.001), while diarrhea was reported in 12.5 vs. 8.1% (RR 2.77, 95% CI 1.55-4.94, P < 0.001). The risk of gastrointestinal events increased with daily dose and shorter treatment duration. Myalgias were observed in 21 vs. 18% patients (RR 1.16, 95% CI 1.02-1.32, P = 0.03). Other adverse events such as myotoxicity, hepatic adverse events, hematologic adverse events, cutaneous adverse events, infection or death were not increased by colchicine treatment. Colchicine discontinuation was reported in 4.8 vs. 3.4% patients (RR 1.54, 95% CI 1.20-1.99, P < 0.001). CONCLUSION Colchicine is associated with increased risk of gastrointestinal events and myalgias, but not of other adverse events. The risk of gastrointestinal events may be avoided with lower dose (0.5 mg/daily) and is inversely related to treatment duration, possibly due to early drug discontinuation or drug tolerance.
Collapse
Affiliation(s)
- Alessandro Andreis
- Department of Medical Sciences, University of Torino, University Cardiology, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Weaver JL. The brain-gut axis: A prime therapeutic target in traumatic brain injury. Brain Res 2020; 1753:147225. [PMID: 33359374 DOI: 10.1016/j.brainres.2020.147225] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 01/10/2023]
Abstract
Traumatic brain injury (TBI) is a significant cause of morbidity and mortality in trauma patients. The primary focus of treating TBI is to prevent additional injury to the damaged brain tissue, known as secondary brain injury. This treatment can include treating the body's inflammatory response. Despite promise in animal models, anti-inflammatory therapy has failed to improve outcomes in human patients, suggesting a more targeted and precise approach may be needed. There is a bidirectional axis between the intestine and the brain that contributes to this inflammation in acute and chronic injury. The mechanisms for this interaction are not completely understood, but there is evidence that neural, inflammatory, endocrine, and microbiome signals all participate in this process. Therapies that target the intestine as a source of inflammation have potential to lessen secondary brain injury and improve outcomes in TBI patients, but to develop these treatments we need to better understand the mechanisms behind this intestinal inflammatory response.
Collapse
Affiliation(s)
- Jessica L Weaver
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego School of Medicine, 200 W Arbor Drive #8896, San Diego, CA 92103-8896, United States.
| |
Collapse
|
26
|
Igarashi S, Nozu T, Ishioh M, Kumei S, Saito T, Toki Y, Hatayama M, Yamamoto M, Shindo M, Tanabe H, Okumura T. Centrally administered orexin prevents lipopolysaccharide and colchicine induced lethality via the vagal cholinergic pathway in a sepsis model in rats. Biochem Pharmacol 2020; 182:114262. [DOI: 10.1016/j.bcp.2020.114262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022]
|
27
|
Mutual Interplay of Host Immune System and Gut Microbiota in the Immunopathology of Atherosclerosis. Int J Mol Sci 2020; 21:ijms21228729. [PMID: 33227973 PMCID: PMC7699263 DOI: 10.3390/ijms21228729] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is the key for the initiation and progression of atherosclerosis. Accumulating evidence has revealed that an altered gut microbiome (dysbiosis) triggers both local and systemic inflammation to cause chronic inflammatory diseases, including atherosclerosis. There have been some microbiome-relevant pro-inflammatory mechanisms proposed to link the relationships between dysbiosis and atherosclerosis such as gut permeability disruption, trigger of innate immunity from lipopolysaccharide (LPS), and generation of proatherogenic metabolites, such as trimethylamine N-oxide (TMAO). Meanwhile, immune responses, such as inflammasome activation and cytokine production, could reshape both composition and function of the microbiota. In fact, the immune system delicately modulates the interplay between microbiota and atherogenesis. Recent clinical trials have suggested the potential of immunomodulation as a treatment strategy of atherosclerosis. Here in this review, we present current knowledge regarding to the roles of microbiota in contributing atherosclerotic pathogenesis and highlight translational perspectives by discussing the mutual interplay between microbiota and immune system on atherogenesis.
Collapse
|
28
|
Dey P. Targeting gut barrier dysfunction with phytotherapies: Effective strategy against chronic diseases. Pharmacol Res 2020; 161:105135. [PMID: 32814166 DOI: 10.1016/j.phrs.2020.105135] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The intestinal epithelial layer serves as a physical and functional barrier between the microbe-rich lumen and immunologically active submucosa; it prevents systemic translocation of microbial pyrogenic products (e.g. endotoxin) that elicits immune activation upon translocation to the systemic circulation. Loss of barrier function has been associated with chronic 'low-grade' systemic inflammation which underlies pathogenesis of numerous no-communicable chronic inflammatory disease. Thus, targeting gut barrier dysfunction is an effective strategy for the prevention and/or treatment of chronic disease. This review intends to emphasize on the beneficial effects of herbal formulations, phytochemicals and traditional phytomedicines in attenuating intestinal barrier dysfunction. It also aims to provide a comprehensive understanding of intestinal-level events leading to a 'leaky-gut' and systemic complications mediated by endotoxemia. Additionally, a variety of detectable markers and diagnostic criteria utilized to evaluate barrier improving capacities of experimental therapeutics has been discussed. Collectively, this review provides rationale for targeting gut barrier dysfunction by phytotherapies for treating chronic diseases that are associated with endotoxemia-induced systemic inflammation.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| |
Collapse
|
29
|
Colchicine increases intestinal permeability, suppresses inflammatory responses, and alters gut microbiota in mice. Toxicol Lett 2020; 334:66-77. [PMID: 33002524 DOI: 10.1016/j.toxlet.2020.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/26/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022]
Abstract
Although colchicine (COL) has been used to treat gout for more than a thousand years, it has been shrouded in a dark history for a long time due to its high toxicity, especially for the gastrointestinal tract. With the widespread clinical application of COL, COL's toxicity to the gastrointestinal tract has raised concerns. This study's objective was to address the exact intestinal toxicity of COL, with particular attention to the effects of COL on gut microbiota homeostasis. The mice were exposed to various dosages of COL (0.1, 0.5, and 2.5 mg kg-1 body weight per day) for a week, and the results showed that COL exposure caused serious intestinal injuries, reducing the relative expression levels of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) and tight junction proteins (zo-1, claudin-1, and occludin) in the ileum and colon tissue. The 16S rRNA gene sequencing analysis of mice feces samples revealed that the composition and diversity of intestinal microbiome underwent a profound remodeling at the dosage of 2.5 mg kg-1 body weight per day, which may increase the toxic load in the gut. In addition, elevated levels of diamine oxidase (DAO) and lipopolysaccharide (LPS) in serum indicated that COL increased intestinal permeability, impairing intestinal barrier. In conclusion, our results demonstrate that COL's toxicity to the gut microbiome is compatible with intestinal injuries, inflammatory pathway inhibition, and increased intestinal permeability; our results also represent a novel insight to uncover the adverse reactions of COL in the gastrointestinal tract.
Collapse
|
30
|
Ghosh SS, Wang J, Yannie PJ, Ghosh S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J Endocr Soc 2020; 4:bvz039. [PMID: 32099951 PMCID: PMC7033038 DOI: 10.1210/jendso/bvz039] [Citation(s) in RCA: 388] [Impact Index Per Article: 77.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/05/2020] [Indexed: 12/24/2022] Open
Abstract
The intestinal barrier is complex and consists of multiple layers, and it provides a physical and functional barrier to the transport of luminal contents to systemic circulation. While the epithelial cell layer and the outer/inner mucin layer constitute the physical barrier and are often referred to as the intestinal barrier, intestinal alkaline phosphatase (IAP) produced by epithelial cells and antibacterial proteins secreted by Panneth cells represent the functional barrier. While antibacterial proteins play an important role in the host defense against gut microbes, IAP detoxifies bacterial endotoxin lipopolysaccharide (LPS) by catalyzing the dephosphorylation of the active/toxic Lipid A moiety, preventing local inflammation as well as the translocation of active LPS into systemic circulation. The causal relationship between circulating LPS levels and the development of multiple diseases underscores the importance of detailed examination of changes in the “layers” of the intestinal barrier associated with disease development and how this dysfunction can be attenuated by targeted interventions. To develop targeted therapies for improving intestinal barrier function, it is imperative to have a deeper understanding of the intestinal barrier itself, the mechanisms underlying the development of diseases due to barrier dysfunction (eg, high circulating LPS levels), the assessment of intestinal barrier function under diseased conditions, and of how individual layers of the intestinal barrier can be beneficially modulated to potentially attenuate the development of associated diseases. This review summarizes the current knowledge of the composition of the intestinal barrier and its assessment and modulation for the development of potential therapies for barrier dysfunction-associated diseases.
Collapse
Affiliation(s)
| | - Jing Wang
- Department of Internal Medicine, VCU Medical Center, Richmond, Virginia
| | - Paul J Yannie
- Hunter Homes McGuire VA Medical Center, Richmond, Virginia
| | - Shobha Ghosh
- Department of Internal Medicine, VCU Medical Center, Richmond, Virginia.,Hunter Homes McGuire VA Medical Center, Richmond, Virginia
| |
Collapse
|