1
|
Xiao Z, Gao S, Li S, Yang F, Zhang D, Niu Z, Zhang Y, Duan Z, Qi S, Ma S. Taohong Siwu Decoction Modulates Glutathione Metabolism to Suppress Hepatocyte Ferroptosis and Demonstrates Anti-Fibrotic Effects in the Liver. JOURNAL OF ETHNOPHARMACOLOGY 2025:120025. [PMID: 40414577 DOI: 10.1016/j.jep.2025.120025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/17/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The ameliorative effect of traditional Chinese medicine (TCM) on hepatic fibrosis has been widely recognized and researched, but studies on the mechanism of action have been hampered by its complex composition, which requires more in-depth studies to elucidate why and how TCM works. The theory of TCM believes that the liver is closely related to blood circulation, and hepatic fibrosis is caused by blood stagnation. Taohong Siwu Decoction (THSW) is a classic formula for nourishing and invigorating blood and has been used clinically for centuries. Current evidence has demonstrated its ameliorative effect on hepatic fibrosis, but the exact mechanism of action remains unclear. AIM OF THE STUDY Exploring the possible mechanism of the anti-hepatic fibrosis effect of THSW by proteomics and validating with in vivo and in vitro studies. MATERIALS AND METHODS The carbon tetrachloride (CCl4)-induced fibrosis model was conducted in mice and treated with THSW in vivo with colchicine as the positive control. Then serum biomarker alanine aminotransferase (ALT), aspartate aminotransferase (AST), and histopathological analysis were evaluated to examine the effects of THSW. And hepatic fibrosis indicators alpha-smooth muscle actin (α-SMA) and Collagen Ⅰ (Col-Ⅰ) were detected by western blotting, immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR) analysis. Additionally, the 4D Label-free quantitative proteomic analysis of liver samples was applied. In vitro, erastin-induced BRL-3A cells, a rat hepatocyte line, were performed as a hepatocyte ferroptosis model and treated with or without drug-containing serum of THSW. Finally, molecular docking was used to verify the binding ability of the main components of THSW to potential targets. RESULTS THSW treatment significantly ameliorated serum ALT, AST, hydroxyproline (Hyp) content, α-SMA and Col-Ⅰ mRNA expression in fibrosis mice. Further results showed that THSW decreased the malondialdehyde (MDA) and 4-Hydroxynonenal (4-HNE) content and increased the glutathione (GSH) content of liver tissue. Notably, proteomic analyses have identified 294 differentially expressed proteins in the THSW-treated group compared to the model group, with 97 proteins up-regulated and 197 down-regulated. Functional analysis of these differential proteins highlights the significant roles of inflammation and oxidative stress. Further validation in vivo and in vitro, THSW significantly improved the protein expression of glutathione S-transferase M1 (GSTM1), down-regulate the expression of transferrin receptor (TFRC), and kelch-like ECH-associated protein 1(Keap1) proteins, and promote the metabolism of GSH. Especially it reduced serum iron levels, increased total iron binding capacity, and up-regulated recombinant solute carrier family 7, member 11 (SLC7A11), nuclear factor erythroid 2-related factor 2 (Nrf2), and glutathione peroxidase 4 (GPX4) protein expression, suggesting the inhibition of hepatocyte ferroptosis. In addition, the molecular docking results showed that its main components, amygdalin, hydroxysafflor yellow A, paeoniflorin, and albiflorin, possessed good binding ability with Keap1. CONCLUSIONS THSW represents a novel therapeutic effect on hepatic fibrosis in mice, accompanied by inhibiting hepatocyte ferroptosis. Mechanically, THSW may regulate the glutathione metabolic pathway and TFRC expression through its main ingredients, such as amygdalin, hydroxysafflor yellow A, paeoniflorin, and albiflorin, thereby inhibiting hepatocyte ferroptosis.
Collapse
Affiliation(s)
- Zhun Xiao
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Siqi Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Shengsheng Li
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Fangming Yang
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Dingqi Zhang
- School of Pharmaceutical Sciences, School of TCM Research, Tsinghua University, Beijing 100084, China.
| | - Zhenyi Niu
- Department of Pathology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Yu Zhang
- Department of Pathology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Zhongping Duan
- Beijing Institute of Hepatology, Beijing Youan Hospital Capital Medical University, Beijing 100069, China.
| | - Shenglan Qi
- Institute of Chinese Materia Medica, Ministry of Education Key Laboratory for Standardization of Chinese Medicines, Shanghai Municipal Key Laboratory for Compound Chinese Medicines, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Suping Ma
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| |
Collapse
|
2
|
Naffaa ME, Hassan F, Jeries H, Dror D, Rozenberg V, Chodick G, Carmiel M. Long-term use of colchicine is associated with incident cirrhosis: a real-world cohort study. Scand J Gastroenterol 2025; 60:361-367. [PMID: 40047243 DOI: 10.1080/00365521.2025.2475488] [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: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Chronic effect of colchicine on the liver was not studied enough. We aimed to examine the association between long term colchicine use and incident cirrhosis among new colchicine initiators. STUDY Using database of Maccabi Healthcare Services (MHS), we included all patients aged ≥18 years old who initiated colchicine between 1 January 2000 and 31 December 2018 and followed them until the earliest of the following: incident cirrhosis, leaving MHS, death or 31 December 2020. Incident cirrhosis was diagnosed according to ICD-9 code. We defined incident decompensated cirrhosis as the first presentation of cirrhosis, once ascites, encephalopathy and/or variceal bleeding were diagnosed within a period of 90 days before until 90 days after incident cirrhosis. Exposure to colchicine was evaluated in two manners: proportion of months covered (PMC) and mean daily dose (MDD). RESULTS A total of 21,773 eligible patients were included. We identified 129 incident cases of cirrhosis. Seventy-six (59%) had decompensated cirrhosis, as a first clinical presentation of cirrhosis. Familial Mediterranean Fever (FMF), BMI > 40, FIBROSIS-4 (FIB-4) score and colchicine PMC were all significantly associated with incident cirrhosis. Patients in '60-80%' PMC group had the highest risk for developing cirrhosis (hazard ratio (HR) 3.68, 95% confidence interval (CI) 2.23-6.07). The risk was higher for patients on colchicine >60 months (HR 6.69, 95% CI 3.56-12.56). CONCLUSIONS long term colchicine use is associated with incident cirrhosis, mainly with decompensation at the time of diagnosis. Long term colchicine treatment should be limited to diseases with no other alternative, such as FMF.
Collapse
Affiliation(s)
- Mohammad E Naffaa
- Rheumatology Unit, Galilee Medical Center, Nahariya, Israel
- Azrieli's Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Fadi Hassan
- Rheumatology Unit, Galilee Medical Center, Nahariya, Israel
- Azrieli's Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Helana Jeries
- Rheumatology Unit, Galilee Medical Center, Nahariya, Israel
- Azrieli's Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Dikla Dror
- Azrieli's Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Liver Unit, Galilee Medical Center, Nahariya, Israel
| | - Vered Rozenberg
- Epidemiology and Database Research, Maccabi Healthcare Services, Tel Aviv, Israel
- Sackler's Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriel Chodick
- Epidemiology and Database Research, Maccabi Healthcare Services, Tel Aviv, Israel
- Sackler's Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michal Carmiel
- Azrieli's Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Liver Unit, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
3
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, De Sanctis JB, Martí-Amarista CE, Hidalgo R, Alegría-Barrero E, Riera Lizardo RJ, Correa-Pérez A. Colchicine for the primary prevention of cardiovascular events. Cochrane Database Syst Rev 2025; 2:CD015003. [PMID: 39927511 PMCID: PMC11808834 DOI: 10.1002/14651858.cd015003.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
BACKGROUND Atherosclerotic cardiovascular diseases (ACVDs), a condition characterised by lipid accumulation in arterial walls, which is often exacerbated by chronic inflammation disorders, is the major cause of mortality and morbidity worldwide. Colchicine, with its first medicinal use in ancient Egypt, is an inexpensive drug with anti-inflammatory properties. However, its role in primary prevention of ACVDs in the general population remains unknown. OBJECTIVES To assess the clinical benefits and harms of colchicine as primary prevention of cardiovascular outcomes in the general population. SEARCH METHODS We searched the Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, Web of Science, and LILACS. We searched ClinicalTrials.gov and WHO ICTRP for ongoing and unpublished studies. We also scanned the reference lists of relevant included studies, reviews, meta-analyses, and health technology reports to identify additional studies. There were no limitations on language, date of publication, or study setting. The search results were updated on 31 May 2023. SELECTION CRITERIA Randomised controlled trials (RCTs) in any setting, recruiting adults without pre-existing cardiovascular disease. We included trials that compared colchicine versus placebo, non-steroidal anti-inflammatory drugs, corticosteroids, immunomodulating drugs, or usual care. Our primary outcomes were all-cause mortality, non-fatal myocardial infarction, stroke, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors independently selected studies, extracted data, and performed risk of bias and GRADE assessments. MAIN RESULTS We identified 15 RCTs (1721 participants randomised; 1412 participants analysed) with follow-up periods ranging from 4 to 728 weeks. The intervention was oral colchicine compared with placebo, immunomodulating drugs, or usual care or no treatment. Due to biases and imprecision, the evidence was very uncertain for all outcomes. All trials but one had a high risk of bias. Five out of seven meta-analyses included fewer than six trials (71.4%). The objectives of the review were to assess cardiovascular outcomes in the general population, but many of the included trials focused on liver disease. Colchicine compared to placebo Colchicine may reduce all-cause mortality compared to placebo in primary prevention, but the evidence is very uncertain (risk ratio (RR) 0.68, 95% confidence interval (CI) 0.51 to 0.91; 6 studies, 463 participants; very low-certainty evidence; number needed to treat for an additional beneficial outcome (NNTB) 11, 95% CI 6 to 67). Colchicine may result in little to no difference in non-fatal myocardial infarction, but the evidence is very uncertain (RR 0.87, 95% CI 0.41 to 1.82; 1 study, 100 participants; very low-certainty evidence). Colchicine may not reduce the incidence of stroke, but the evidence is very uncertain (RR 2.43, 95% CI 0.67 to 8.86; 1 study, 100 participants; very low-certainty evidence). Regarding adverse events, colchicine may increase the incidence of diarrhoea (RR 3.99, 95% CI 1.44 to 11.06; 8 studies, 605 participants; very low-certainty evidence; number needed to treat for an additional harmful outcome (NNTH) 10, 95% CI 6 to 17), and may have little to no effect on neurological outcomes such as seizure or mental confusion (RR 0.72, 95% CI 0.31 to 1.66; 2 studies, 155 participants; very low-certainty evidence), but the evidence is very uncertain. The effect of colchicine on cardiovascular mortality is also very uncertain (RR 1.27, 95% CI 0.03 to 62.43; 2 studies, 160 participants; very low-certainty evidence). Colchicine may not reduce post-cardiac procedure atrial fibrillation, but the evidence is very uncertain (RR 0.74, 95% CI 0.25 to 2.19; 1 study, 100 participants). We found no trials reporting on pericardial effusion, peripheral artery disease, heart failure, or unstable angina. Colchicine compared to methotrexate (immunomodulating drug) Colchicine may result in little to no difference in all-cause mortality compared to methotrexate, but the evidence is very uncertain (RR 0.42, 95% CI 0.12 to 1.51; 1 study, 85 participants; very low-certainty evidence). We found no trials reporting other cardiovascular outcomes or adverse events for this comparison. Colchicine compared to usual care or no treatment The evidence is very uncertain about the effect of colchicine compared with usual care on all-cause mortality in primary prevention (RR 1.07, 95% CI 0.90 to 1.27; 2 studies, 729 participants; very low-certainty evidence). Regarding adverse events, colchicine may increase the incidence of diarrhoea compared to usual care, but the evidence is very uncertain (RR 3.32, 95% CI 1.56 to 7.03; 2 studies, 729 participants; very low-certainty evidence; NNTH 18, 95% CI 12 to 42). No trials reported other cardiovascular outcomes for this comparison. AUTHORS' CONCLUSIONS This Cochrane review evaluated the clinical benefits and harms of using colchicine for the primary prevention of cardiovascular events in the general population. Comparisons were made against placebo, immunomodulating medications, or usual care or no treatment. However, the certainty of the evidence for the predefined outcomes was very low, highlighting the pressing need for high-quality, rigorous studies to ascertain colchicine's clinical impact definitively. We identified numerous biases and inaccuracies in the included studies, limiting their generalisability and precluding a conclusive determination of colchicine's efficacy in preventing cardiovascular events. The existing evidence regarding colchicine's potential cardiovascular benefits or harms for primary prevention is inconclusive owing to the limitations inherent in the current studies. More robust clinical trials are needed to bridge this evidence gap effectively.
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador, Universidad UTE, Quito, Ecuador
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Mario A Gemmato-Valecillos
- Department of Medicine, NYC Health + Hospitals / Elmhurst, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine, Palacky University Olomouc, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | | | - Ricardo Hidalgo
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador., Universidad UTE, Quito, Ecuador
| | | | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
- Medicine Department, Rheumatology Unit, Universidad de Carabobo, Valencia, Venezuela
| | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Department of Hospital Pharmacy and Medical Devices, Hospital Central de la Defensa Gómez Ulla, Madrid, Spain
| |
Collapse
|
4
|
Delplanque M, Amiot X, Wendum D, Rodrigues F, Aknouche Z, Bourguiba R, Terris B, Duvoux C, Bedossa P, Lebrec D, Sogni P, Parlati L, Charlotte F, Ratziu V, Mouly S, Augustin J, Calderaro J, Scoazec G, Michel Vignaud J, Arnaud Seyrig J, Grateau G, Savey L, Georgin-Lavialle S. Liver Disease Complicating Familial Mediterranean Fever: A Study on 66 Patients Out of 533 Adult From the JIR Cohort. Liver Int 2025; 45:e16232. [PMID: 39790043 DOI: 10.1111/liv.16232] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease, associated with MEFV mutations. FMF patients can experience liver involvement, potentially leading to cirrhosis. OBJECTIVES This study aimed to evaluate liver involvement in FMF patients at a French tertiary centre for adult FMF. METHODS We conducted an observational study with FMF patients displaying 2 pathogenic MEFV mutations at the National Reference Center for Autoinflammatory Diseases and Inflammatory Amyloidosis (CEREMAIA) in Paris and included in the JIR cohort. MEFV heterozygous patients and those with other liver disease causes were excluded. RESULTS Among 533 FMF patients 12.4% had chronic liver abnormalities, with 30% who developed cirrhosis 54 years [36-57] in median after disease onset. Forty-seven per cent were colchicine resistant, and 41% received interleukin-1 inhibitors. Cirrhotic patients experienced delayed hepatopathy diagnosis, prolonged FMF diagnosis delay and late-onset treatment initiation compared to those with only liver function test abnormalities. Colchicine resistance and interleukin-1 inhibitor use were more common in cirrhotic patients. Body mass index and AA amyloidosis rates did not differ significantly between groups. Twenty-one patients had undergone liver biopsies including 14 cirrhotic patients revealing steatohepatitis in 12 cases and probable steatohepatitis in 4. Other lesions, like iron overload and sinusoidal dilatation, were sporadically observed. CONCLUSION FMF patients are at risk of chronic liver disease. Regular liver function monitoring is crucial, particularly in case of persistent inflammation, due to the risk of progression to cirrhosis and its associated morbidity and mortality.
Collapse
Affiliation(s)
- Marion Delplanque
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Xavier Amiot
- Sorbonne Université, Service de Gastroenterologie, Assistance Publique des hôpitaux de Paris Hôpital Tenon, Paris, France
| | - Dominique Wendum
- Sorbonne Université, Service d'anatomopathologie, Assistance Publique des hôpitaux de Paris Hôpital Saint Antoine, Paris, France
| | - François Rodrigues
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Zohra Aknouche
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Rim Bourguiba
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Benoit Terris
- Université de Paris, Service d'anatomopathologie, Assistance Publique des hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Christophe Duvoux
- Service d'hépatogastroenterologie, Assistance Publique des hôpitaux de Paris, Hôpital Mondor, Créteil, France
| | - Pierre Bedossa
- Université Paris Cité, Service d'anatomopathologie, Assistance Publique des hôpitaux de Paris, Hôpital Beaujon, Clichy, France
| | - Didier Lebrec
- Université Paris Cité, Service d'hépatogastroenterologie, Assistance Publique des hôpitaux de Paris, Hôpital Beaujon, Clichy, France
| | - Philippe Sogni
- Université Paris Cité, Service d'hépatogastroenterologie Assistance Publique des hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Lucia Parlati
- Université Paris Cité, Service d'hépatogastroenterologie Assistance Publique des hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Frederic Charlotte
- Sorbonne Université, Service d'anatomopathologie, Assistance Publique des hôpitaux de Paris, Hôpital Pitié Salpêtrière, Paris, France
| | - Vlad Ratziu
- Sorbonne Université, Service d'hépatogastroenterologie, Assistance Publique des hôpitaux de Paris, Hôpital Pitié Salpêtrière, Paris, France
| | - Stéphane Mouly
- Université Paris Cité, DMU INVICTUS, Département de Médecine Interne, Assistance Publique des hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Jeremy Augustin
- Service d'anatomopathologie, Assistance Publique des hôpitaux de Paris, Hôpital Mondor, Créteil, France
| | - Julien Calderaro
- Service d'anatomopathologie, Assistance Publique des hôpitaux de Paris, Hôpital Mondor, Créteil, France
| | - Giovanna Scoazec
- Service d'hépatogastroenterologie, Assistance Publique des hôpitaux de Paris, Hôpital Mondor, Créteil, France
| | | | | | - Gilles Grateau
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Léa Savey
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Sophie Georgin-Lavialle
- Sorbonne Université, Service Médecine Interne, Centre de référence des maladies autoinflammatoires et des amyloses (CEREMAIA), Assistance Publique des hôpitaux de Paris, Hôpital Tenon, Paris, France
| |
Collapse
|
5
|
Xue W, Liu H, Su Z, Wang S, Cheng J, Pan Y, Zhang L. Qinggan Yipi capsule ameliorates hepatic fibrosis in rats by down-regulating the TGF-β1/Smad2/3 signaling pathway and improving gut microbiota imbalance. Front Pharmacol 2025; 16:1525914. [PMID: 39925848 PMCID: PMC11802500 DOI: 10.3389/fphar.2025.1525914] [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: 11/10/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Background and objective Qinggan Yipi Capsule (QgYp) is a hospital preparation that has been used for many years in the treatment of chronic liver diseases. However, the mechanism of QgYp in ameliorating hepatic fibrosis (HF) remains unclear. This study aims to clarify the anti-liver fibrosis effect of QgYp and its mechanism of action. Methods This study uses a carbon tetrachloride (CCl4) induced HF rat model and TGF-β1 stimulated HSC-T6 cell line (rat HSCs) as experimental models. The therapeutic effects were evaluated through pathology, biochemical tests, and ELISA. The therapeutic mechanism of QgYp for HF was predicted through network pharmacology. The expression of TGF-β1/Smad2/3 related proteins was detected by qPCR analysis and Western blot analysis. The composition of the gut microbiota was analyzed using 16S rRNA gene sequencing. Results Histopathological analysis, serum biochemical tests, and ELISA measurements showed that QgYp effectively decreased the levels of ALT, AST, HA, LN, PCIII, and IV-C while improving collagen deposition and hepatocyte necrosis. Protein-protein interaction (PPI) network analysis screened HF-related genes, including peroxisome proliferator-activated receptor gamma (PPARG), tumor necrosis factor (TNF), and TGF-β1. GO and KEGG analyses indicated that QgYp significantly affects TGF-β signaling pathway. In addition, the results of qPCR and Western blot analysis from both in vitro and in vivo experiments indicated that QgYp significantly downregulated the expression of proteins and mRNA associated with the TGF-β1/Smad2/3 pathway. The 16S rDNA gene sequencing results showed that QgYp can increase the diversity and richness of the gut microbiota in HF rats and alter the composition of the gut microbiota. Conclusion QgYp could effectively ameliorate HF, and this effect might be connected to the downregulation of the TGF-β1/Smad2/3 pathway, the suppression of HSCs activation, and regulation of gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Wenjing Xue
- Key Laboratory for Evaluation and Transformation of Wu Men Medical School’s Empirical Prescriptions, Suzhou Traditional Chinese Medicine Hospital, Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Haiqing Liu
- Key Laboratory for Evaluation and Transformation of Wu Men Medical School’s Empirical Prescriptions, Suzhou Traditional Chinese Medicine Hospital, Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Ziheng Su
- Department of Pharmacy, Affiliated Hospital of Hebei University, Baoding, China
| | - Siqi Wang
- Department of Pharmacy, Suzhou Fifth People’s Hospital, Suzhou, China
| | - Junping Cheng
- Key Laboratory for Evaluation and Transformation of Wu Men Medical School’s Empirical Prescriptions, Suzhou Traditional Chinese Medicine Hospital, Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yunzhi Pan
- Department of Pharmacy, Suzhou Fifth People’s Hospital, Suzhou, China
| | - Lurong Zhang
- Key Laboratory for Evaluation and Transformation of Wu Men Medical School’s Empirical Prescriptions, Suzhou Traditional Chinese Medicine Hospital, Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| |
Collapse
|
6
|
Wu WH, Yang YL, Wang T, Sun XM, Wei MG, Zhou XY, Zhu LZ, Ma G, Liu B, Qi LW, Liu Q. Ginsenoside compound K restrains hepatic fibrotic response by dual-inhibition of GLS1 and LDHA. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156223. [PMID: 39561660 DOI: 10.1016/j.phymed.2024.156223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/05/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Liver fibrosis is a dynamic process marked by the accumulation of extracellular matrix due to hepatic stellate cells (HSCs) activation. Ginsenoside compound K (CK), a rare derivative of its parent ginsenosides, is known to significantly ameliorate metabolic disorders. PURPOSE The aim of this study was to elucidate the protective effects of CK against liver fibrosis with a focus on metabolic regulation. METHODS We established liver fibrosis models in mice using carbon tetrachloride (CCl4) challenge, bile duct ligation, or a methionine-choline deficient diet, with continuous oral administration of CK at specified doses and intervals. Simultaneously, we examined the impact of CK on metabolic regulation in cultured HSCs and investigated the associated mechanisms. RESULTS CK was found to alleviate liver injury and curb fibrotic responses in mouse models, as well as decrease elevated levels of liver enzyme. Metabolomic analysis in vitro highlighted the crucial roles of pyruvate and glutamine metabolism in metabolic remodeling. Immunohistochemical staining indicated significantly elevated expressions of lactate dehydrogenase A (LDHA) (p = 0.014) and glutaminase 1 (GLS1) (p = 0.024) in liver cirrhosis patients. Comparable alterations were noted in the liver of model mice and in cultured HSCs. Molecular docking and bio-layer interferometry demonstrated that CK interacts with and inhibits the activities of LDHA and GLS1. As expected, CK attenuated glycolysis and glutaminolysis, reducing HSC growth dependently on lactate and α-ketoglutarate (α-KG). Upon HSC activation, metabolism is reprogrammed with Myc as a key regulator, transcriptionally controlling LDHA, GLS1, and glutamine transporters SLC1A5 and SLC38A5. CK inhibited Myc induction, integrating glycolysis and glutaminolysis regulation to counteract the fibrotic response. CONCLUSION CK inhibited LDHA and GLS1 activities, thereby inhibiting hepatic fibrosis. These findings offer new insights into the role of ginsenosides in liver protection, especially regarding metabolic disorders.
Collapse
Affiliation(s)
- Wen-Hui Wu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Ya-Lan Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Ting Wang
- Department of Pathology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210009, China
| | - Xiao-Meng Sun
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Meng-Guang Wei
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Xin-Yue Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Zeng Zhu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Gaoxiang Ma
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Baolin Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China
| | - Lian-Wen Qi
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing 210009, China.
| | - Qun Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
7
|
de Bulhões FV, Assis GE, Cazé AB, Barros-Pereira JP, Laguna GGDC, Improta-Caria AC, Aras-Júnior R. The Action of Colchicine in Patients with Metabolic Syndrome and Obesity: Perspectives and Challenges. Metabolites 2024; 14:629. [PMID: 39590865 PMCID: PMC11596755 DOI: 10.3390/metabo14110629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/27/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Colchicine is an alkaloid traditionally used to treat inflammatory conditions such as gout and familial Mediterranean fever. Currently, there are proposals for the use of this drug in several other situations, such as cardiovascular and liver diseases and diabetes. In this study, the current literature on the potential of colchicine in the treatment of obesity and metabolic syndrome (MS) was evaluated. The inhibitory action of the NLRP3 inflammasome and other processes, such as reductions in the migration and activation of immune system cells, are effects observed in both in vitro studies and animal models related to colchicine, as well as the promotion of mechanisms of the intensification of lipid metabolism, the reduction of tissue fibrosis, and the reduction of serum glucose and triglycerides. These factors are associated with changes in the prognoses of patients with MS, which, together with obesity, has a high association with inflammatory mechanisms for its maintenance and secondary impairments to homeostasis. In humans, clinical research has rarely addressed the use of colchicine in obesity and MS, with only one pilot randomized clinical trial having been conducted, which identified a beneficial anti-inflammatory effect on endothelial function and the process of insulin resistance in this population. However, it is not yet possible to extrapolate its findings and apply its results to a broader context. Given the potential of this "ancient drug" in various pathological contexts and its good tolerability, it is important that its properties continue to be investigated and that more clinical studies be conducted to expand the therapeutic applications of this low-cost substance in patients with obesity and MS.
Collapse
Affiliation(s)
- Fábio Vieira de Bulhões
- Faculty of Medicine, Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (F.V.d.B.); (G.E.A.); (A.B.C.); (J.P.B.-P.)
| | - Gabriele Eliza Assis
- Faculty of Medicine, Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (F.V.d.B.); (G.E.A.); (A.B.C.); (J.P.B.-P.)
| | - Ana Beatriz Cazé
- Faculty of Medicine, Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (F.V.d.B.); (G.E.A.); (A.B.C.); (J.P.B.-P.)
| | - Jackson Pedro Barros-Pereira
- Faculty of Medicine, Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (F.V.d.B.); (G.E.A.); (A.B.C.); (J.P.B.-P.)
| | | | - Alex Cleber Improta-Caria
- Laboratory of Biochemistry and Molecular Biology of the Exercise, Physical Education and Sport School, University of Sao Paulo (USP), Sao Paulo 05508-030, SP, Brazil
| | - Roque Aras-Júnior
- Faculty of Medicine, Federal University of Bahia (UFBA), Salvador 40110-060, BA, Brazil; (F.V.d.B.); (G.E.A.); (A.B.C.); (J.P.B.-P.)
| |
Collapse
|
8
|
Meroueh C, Warasnhe K, Tizhoosh HR, Shah VH, Ibrahim SH. Digital pathology and spatial omics in steatohepatitis: Clinical applications and discovery potentials. Hepatology 2024:01515467-990000000-00815. [PMID: 38517078 PMCID: PMC11669472 DOI: 10.1097/hep.0000000000000866] [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: 01/27/2024] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
Steatohepatitis with diverse etiologies is the most common histological manifestation in patients with liver disease. However, there are currently no specific histopathological features pathognomonic for metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, or metabolic dysfunction-associated steatotic liver disease with increased alcohol intake. Digitizing traditional pathology slides has created an emerging field of digital pathology, allowing for easier access, storage, sharing, and analysis of whole-slide images. Artificial intelligence (AI) algorithms have been developed for whole-slide images to enhance the accuracy and speed of the histological interpretation of steatohepatitis and are currently employed in biomarker development. Spatial biology is a novel field that enables investigators to map gene and protein expression within a specific region of interest on liver histological sections, examine disease heterogeneity within tissues, and understand the relationship between molecular changes and distinct tissue morphology. Here, we review the utility of digital pathology (using linear and nonlinear microscopy) augmented with AI analysis to improve the accuracy of histological interpretation. We will also discuss the spatial omics landscape with special emphasis on the strengths and limitations of established spatial transcriptomics and proteomics technologies and their application in steatohepatitis. We then highlight the power of multimodal integration of digital pathology augmented by machine learning (ML)algorithms with spatial biology. The review concludes with a discussion of the current gaps in knowledge, the limitations and premises of these tools and technologies, and the areas of future research.
Collapse
Affiliation(s)
- Chady Meroueh
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Khaled Warasnhe
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - H. R. Tizhoosh
- Department of Artificial Intelligence & Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H. Shah
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samar H. Ibrahim
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Pediatric Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Wang S, Li W, Liu W, Yu L, Peng F, Qin J, Pu L, Tang Y, Xie X, Peng C. Total flavonoids extracted from Penthorum chinense Pursh mitigates CCl 4-induced hepatic fibrosis in rats via inactivation of TLR4-MyD88-mediated NF-κB pathways and regulation of liver metabolism. Front Pharmacol 2023; 14:1253013. [PMID: 38074148 PMCID: PMC10701287 DOI: 10.3389/fphar.2023.1253013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/10/2023] [Indexed: 05/16/2025] Open
Abstract
Background: Penthorum chinense Pursh (PCP) is widely utilized in China to treat a variety of liver diseases. It has been shown that flavonoids inhibit inflammation and have the potential to attenuate tissue damage and fibrosis. However, the mechanisms underlying how total flavonoids isolated from PCP (TFPCP) exert their anti-fibrotic effects remain unclear. Methods: The chemical composition of TFPCP was determined using UHPLC-Q-Orbitrap HRMS. Subsequently, rats were randomly assigned to a control group (Control), a carbon tetrachloride (CCl4)-induced hepatic fibrosis model group (Model), a positive control group [0.2 mg/(kg∙day)] of Colchicine), and three TFPCP treatment groups [50, 100, and 150 mg/(kg∙day)]. All substances were administered by gavage and treatments lasted for 9 weeks. Simultaneously, rats were intraperitoneally injected with 10%-20% CCl4 for 9 weeks to induce liver fibrosis. At the end of the experiment, the liver ultrasound, liver histomorphological, biochemical indicators, and inflammatory cytokine levels were tested respectively. The underlying mechanisms were assessed using Western blot, immunohistochemistry, immunofluorescence, RT-qPCR, and metabolomics. Results: Fourteen flavonoids were identified in TFPCP. Compared with control animals, CCl4-treated rats demonstrated obvious liver injury and fibrosis, manifested as increases in gray values, distal diameter of portal vein (DDPV) and a decrease in blood flow velocity (VPV) in the ultrasound analysis; increased biochemical index values (serum levels of ALT, AST, TBIL, and ALP); marked increases in the contents of fibrotic markers (PC III, COL4, LN, HA) and inflammatory factors (serum TNF-α, IL-6, and IL-1β); and significant pathological changes. However, compared with the Model group, the ultrasound parameters were significantly improved and the serum levels of inflammatory cytokines were reduced in the TFPCP group. In contrast, the expression of TGF-β1, TLR4, and MyD88, as well as the p-P65/P65 and p-IκBα/IκBα ratios, were considerably reduced following TFPCP treatment. In addition, we identified 32 metabolites exhibiting differential abundance in the Model group. Interestingly, TFPCP treatment resulted in the restoration of the levels of 20 of these metabolites. Conclusion: Our findings indicated that TFPCP can ameliorate hepatic fibrosis by improving liver function and morphology via the inactivation of the TLR4/MyD88-mediated NF-κB pathway and the regulation of liver metabolism.
Collapse
Affiliation(s)
- Sujuan Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenqing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenxiu Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Junyuan Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Pu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunli Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Korkmaz C, Cansu DÜ, Cansu GB. A Hypothesis Regarding Neurosecretory Inhibition of Stress Mediators by Colchicine in Preventing Stress-Induced Familial Mediterranean Fever Attacks. Front Immunol 2022; 13:834769. [PMID: 35251026 PMCID: PMC8891608 DOI: 10.3389/fimmu.2022.834769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 12/31/2022] Open
Abstract
Familial Mediterranean fever (FMF) is a monogenic autoinflammatory disease characterized by recurrent episodes of fever and serositis. Colchicine (Col) has a crucial role in the prevention of amyloidosis and FMF attacks. The effect of Col on innate immune cells is based on the inhibition of the microtubule system. The microtubule system is also very important for neurosecretory functions. The inhibitory effect of Col on neurosecretory functions is an overlooked issue. Considering that the neuroimmune cross-talk process plays a role in the development of inflammatory diseases, the effect of Col on the neuronal system becomes important. FMF attacks are related to emotional stress. Therefore, the effect of Col on stress mediators is taken into consideration. In this hypothetical review, we discuss the possible effects of Col on the central nervous systems (CNS) and peripheral nervous systems (PNS) in light of mostly experimental study findings using animal models. Studies to be carried out on this subject will shed light on the pathogenesis of FMF attacks and the other possible mechanisms of action of Col apart from the anti-inflammatory features.
Collapse
Affiliation(s)
- Cengiz Korkmaz
- Department of Internal Medicine, Division of Rheumatology, School of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Döndü Üsküdar Cansu
- Department of Internal Medicine, Division of Rheumatology, School of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Güven Barıs Cansu
- Department of Endocrinology, School of Medicine, Kutahya Health Science University, Kutahya, Turkey
| |
Collapse
|
11
|
Mu P, Feng J, Hu Y, Xiong F, Ma X, Tian L. Botanical Drug Extracts Combined With Biomaterial Carriers for Osteoarthritis Cartilage Degeneration Treatment: A Review of 10 Years of Research. Front Pharmacol 2022; 12:789311. [PMID: 35173609 PMCID: PMC8841352 DOI: 10.3389/fphar.2021.789311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a long-term chronic arthrosis disease which is usually characterized by pain, swelling, joint stiffness, reduced range of motion, and other clinical manifestations and even results in disability in severe cases. The main pathological manifestation of OA is the degeneration of cartilage. However, due to the special physiological structure of the cartilage, once damaged, it is unable to repair itself, which is one of the challenges of treating OA clinically. Abundant studies have reported the application of cartilage tissue engineering in OA cartilage repair. Among them, cell combined with biological carrier implantation has unique advantages. However, cell senescence, death and dedifferentiation are some problems when cultured in vitro. Botanical drug remedies for OA have a long history in many countries in Asia. In fact, botanical drug extracts (BDEs) have great potential in anti-inflammatory, antioxidant, antiaging, and other properties, and many studies have confirmed their effects. BDEs combined with cartilage tissue engineering has attracted increasing attention in recent years. In this review, we will explain in detail how cartilage tissue engineering materials and BDEs play a role in cartilage repair, as well as the current research status.
Collapse
Affiliation(s)
- Panyun Mu
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Feng
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yimei Hu
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Yimei Hu,
| | - Feng Xiong
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xu Ma
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linling Tian
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Li JJ, Jiang HC, Wang A, Bu FT, Jia PC, Zhu S, Zhu L, Huang C, Li J. Hesperetin derivative-16 attenuates CCl 4-induced inflammation and liver fibrosis by activating AMPK/SIRT3 pathway. Eur J Pharmacol 2022; 915:174530. [PMID: 34902361 DOI: 10.1016/j.ejphar.2021.174530] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022]
Abstract
Liver fibrosis, a chronic inflammatory healing reaction, progresses to hepatocirrhosis without effective intervention. Hesperetin derivative (HD-16), a monomer compound derived from hesperitin, exerts anti-inflammatory and hepatoprotective effects against a spectrum of liver diseases. However, the anti-fibrotic potential of HD-16 in liver fibrosis and its underlying mechanism have not yet been elucidated. In this study, we investigated the anti-fibrotic effect of HD-16 on mouse liver fibrosis induced by CCl4 and on LX-2 cells (human immortalized HSCs) stimulated by TGF-β1, in vivo and in vitro. HD-16 exerted an anti-fibrotic effect via regulation of the AMPK/SIRT3 pathway. Pharmacodynamic results showed that HD-16 alleviated the degree of injury and inflammation in CCl4-induced mouse liver fibrosis. Consistently, HD-16 also effectively inhibited the expression of α-SMA, Col1α1, Col3α1, and TIMP-1 in TGF-β1-activated LX-2 cells. Mechanistically, HD-16 promoted SIRT3 expression and activity in fibrotic liver and activated LX-2 cells. Furthermore, SIRT3 depletion attenuated the anti-fibrotic effects of HD-16. Intriguingly, HD-16 increased AMPK phosphorylation, whereas inhibition of SIRT3 expression did not affect AMPK phosphorylation. In contrast, AMPK silencing suppressed SIRT3 expression, suggesting that SIRT3 is a downstream target of AMPK in liver fibrosis. Overall, HD-16 attenuated CCl4-induced liver inflammation and fibrosis by activating the AMPK/SIRT3 pathway, and HD-16 may be a potential anti-fibrotic compound in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Juan-Juan Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - He-Chun Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; The First Affiliated Hospital of USTC Anhui Provincial Hospital, China
| | - Ao Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Fang-Tian Bu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Peng-Cheng Jia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Sai Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Lin Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Abstract
Fibrosis is not a unidirectional, linear process, but a dynamic one resulting from an interplay of fibrogenesis and fibrolysis depending on the extent and severity of a biologic insult, or lack thereof. Regression of fibrosis has been documented best in patients treated with phlebotomies for hemochromatosis, and after successful suppression and eradication of chronic hepatitis B and C infections. This evidence mandates a reconsideration of the term "cirrhosis," which implies an inevitable progression towards liver failure. Furthermore, it also necessitates a staging system that acknowledges the bidirectional nature of evolution of fibrosis, and has the ability to predict if the disease process is progressing or regressing. The Beijing classification attempts to fill this gap in contemporary practice. It is based on microscopic features termed "the hepatic repair complex," defined originally by Wanless and colleagues. The elements of the hepatic repair complex represent the 3 processes of fragmentation and regression of scar, vascular remodeling (resolution), and parenchymal regeneration. However, regression of fibrosis does not imply resolution of cirrhosis, which is more than just a stage of fibrosis. So far, there is little to no evidence to suggest that large regions of parenchymal extinction can be repopulated by regenerating hepatocytes. Similarly, the vascular lesions of cirrhosis persist, and there is no evidence of complete return to normal microcirculation in cirrhotic livers. In addition, the risk of hepatocellular carcinoma is higher compared with the general population and these patients need continued screening and surveillance.
Collapse
|
14
|
Inhibiting NLRP3 Inflammasome Activity in Acute Myocardial Infarction: A Review of Pharmacologic Agents and Clinical Outcomes. J Cardiovasc Pharmacol 2020; 74:297-305. [PMID: 31356538 DOI: 10.1097/fjc.0000000000000701] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The NLRP3 inflammasome is an intracellular, multimeric protein complex that initiates a potent inflammatory response to danger signals. After acute myocardial infarction, NLRP3 inflammasome-dependent inflammation promotes adverse left ventricular remodeling and recurrent atherosclerotic events. Selective and nonselective inhibitors of the NLRP3 inflammasome or its downstream effectors (interleukin-1β and interleukin-18) may prevent adverse left ventricular remodeling and recurrent atherosclerotic events. In this review, we highlight strategies to inhibit NLRP3 inflammasome activity and their potential roles in the management of acute myocardial infarction.
Collapse
|
15
|
Zhang B, Lai L, Tan Y, Liang Q, Bai F, Mai W, Huang Q, Ye Y. Hepatoprotective effect of total flavonoids of Mallotus apelta (Lour.) Muell.Arg. leaf against carbon tetrachloride-induced liver fibrosis in rats via modulation of TGF-β1/Smad and NF-κB signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112714. [PMID: 32105750 DOI: 10.1016/j.jep.2020.112714] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 10/06/2019] [Accepted: 02/23/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Mallotus apelta (Lour.) Muell.Arg. is a well-known traditional Chinese medicine (TCM) used for anti-inflammatory, hemostasis and chronic hepatitis. AIM The purpose of this study was to explore the antifibrotic effect of total flavonoids of Mallotus apelta leaf (TFM) and its potential mechanism. METHODS Hepatic fibrosis was induced by carbon tetrachloride (CCl4) in rats. The CCl4-induced rats received intragastric administration of colchicine (0.2 mg/kg per day), TFM (25, 50, 100 mg/kg per day) and the equal vehicle was given to normal rats. Pathological evaluation in hepatic tissue were examined by hematoxylin and eosin (HE) staining. And the levels of serum biochemical parameters were detected by automatic biochemical analysis. Meanwhile, the collagen deposition in liver was observed by staining with Masson's trichrome. Collagenic parameters and inflammatory factors were measured by enzyme-linked immunosorbent assay (ELISA) kits. Additionally, corresponding assay kit was used to estimate the antioxidant enzyme and lipid peroxidation. In order to explore the potential mechanism of anti-fibrotic effects in TFM, the expressions of liver fibrosis related gene and protein were analyzed by real-time quantitative reverse transcription polymerase chain reaction (RT-PCR) and Western blot. RESULTS The CCl4-induced hepatic fibrosis were inhibited dose-dependently in rats by TFM. The results showed that the key hallmarks of liver injury including aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), albumin (ALB) and total protein (TP) in the serum were reversed in CCl4-induced hepatic fibrosis rats which were treated by TFM. Furthermore, TFM significantly alleviates collagen accumulation and reduces the contents of hydroxyproline (Hyp), Type III precollagen (PC-III), collagen I (Col I), hyaluronic acid (HA) and laminin (LN). RT-PCR and Western blot results showed that TFM markedly inhibits liver fibrosis hallmark factor α-smooth muscle actin (α-SMA) expressions in CCl4-induced hepatic fibrosis rats. Moreover, TFM alleviated the oxidative stress and lipid peroxidation in rats induced by CCl4. TFM also attenuated the pro-inflammatory cytokines including interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) via inhibiting nuclear factor-κB (NF-κB) activation. Meanwhile, transforming growth factor-β1 (TGF-β1)/Smad signaling pathway was inhibited by TFM treatment. CONCLUSIONS TFM can alleviate CCl4-induced hepatic fibrosis in rats, which potential mechanism may be due to its ability of reducing ECM accumulation, improving antioxidant and regulating TGF-β1/Smad signaling pathways and NF-κB-dependent inflammatory response.
Collapse
Affiliation(s)
- Bo Zhang
- Guangxi Medical University, Guangxi, China.
| | - Ling Lai
- Guangxi Medical University, Guangxi, China.
| | - Yanjun Tan
- Guangxi Medical University, Guangxi, China.
| | | | | | | | - Qiujie Huang
- Guangxi University of Chinese Medicine, Guangxi, China.
| | - Yong Ye
- Guangxi Medical University, Guangxi, China.
| |
Collapse
|
16
|
Adverse events during oral colchicine use: a systematic review and meta-analysis of randomised controlled trials. Arthritis Res Ther 2020; 22:28. [PMID: 32054504 PMCID: PMC7020579 DOI: 10.1186/s13075-020-2120-7] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/06/2020] [Indexed: 12/26/2022] Open
Abstract
Background Colchicine is a widely used drug to treat inflammatory diseases. Due to its long historical use in medicine, controlled clinical trials have been small and there remains some caution with the use of this drug in patients with co-morbidities. The aim of the study is to systematically examine the side effect profile of colchicine in controlled clinical trials across all published indications. Methods A systematic review was conducted in accordance with PRISMA methodology. The Cochrane Library, MEDLINE and EMBASE were searched for double-blind controlled trials of oral colchicine in adult patients that reported adverse event data. Meta-analyses were used to determine the relative risk (RR) of adverse events in colchicine users compared to comparator groups. Results A total of 4915 studies were initially identified and after exclusions, 35 randomised controlled trials with placebo (n = 35) or active comparators (n = 5) were included. The most common diseases studied were gout, liver cirrhosis and pericarditis. There were a total of 8659 pooled participants, 4225 participants were randomised to receive colchicine, 3956 to placebo and 411 to an active comparator. Diarrhoea was reported in 17.9% of colchicine users versus 13.1% in comparator groups (RR 2.4, 95% confidence interval (CI) 1.6, 3.7). Any gastrointestinal event was reported in 17.6% of colchicine users and 13.1% of comparators (RR 1.7, 95% CI 1.3, 2.3). Adverse liver events were reported in 1.9% of colchicine users versus 1.1% in the comparator groups (RR 1.6, 95% CI 0.9, 3.0). Muscle events were reported in 4.2% of colchicine users and 3.3% in the comparator groups (RR 1.3, 95% CI 0.8, 1.9). Haematology events were reported in 0.6% of colchicine users and 0.4% of comparator groups (RR 1.34 (0.64, 2.82). No study reported neuropathy events. Other sensory events were reported in 1.1% of colchicine users and 1.5% of comparator groups (RR 1.4, 95% CI 0.3, 6.7). Infectious events were reported in 0.4% of colchicine users and 2.1% of comparator groups (RR 1.0, 95% CI 0.7, 1.5). No study reported death as an adverse event. Conclusion Colchicine increases the rate of diarrhoea and gastrointestinal adverse events but does not increase the rate of liver, sensory, muscle, infectious or haematology adverse events or death.
Collapse
|
17
|
Janssen L, Allard NAE, Saris CGJ, Keijer J, Hopman MTE, Timmers S. Muscle Toxicity of Drugs: When Drugs Turn Physiology into Pathophysiology. Physiol Rev 2019; 100:633-672. [PMID: 31751166 DOI: 10.1152/physrev.00002.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Drugs are prescribed to manage or prevent symptoms and diseases, but may sometimes cause unexpected toxicity to muscles. The symptomatology and clinical manifestations of the myotoxic reaction can vary significantly between drugs and between patients on the same drug. This poses a challenge on how to recognize and prevent the occurrence of drug-induced muscle toxicity. The key to appropriate management of myotoxicity is prompt recognition that symptoms of patients may be drug related and to be aware that inter-individual differences in susceptibility to drug-induced toxicity exist. The most prevalent and well-documented drug class with unintended myotoxicity are the statins, but even today new classes of drugs with unintended myotoxicity are being discovered. This review will start off by explaining the principles of drug-induced myotoxicity and the different terminologies used to distinguish between grades of toxicity. The main part of the review will focus on the most important pathogenic mechanisms by which drugs can cause muscle toxicity, which will be exemplified by drugs with high risk of muscle toxicity. This will be done by providing information on key clinical and laboratory aspects, muscle electromyography patterns and biopsy results, and pathological mechanism and management for a specific drug from each pathogenic classification. In addition, rather new classes of drugs with unintended myotoxicity will be highlighted. Furthermore, we will explain why it is so difficult to diagnose drug-induced myotoxicity, and which tests can be used as a diagnostic aid. Lastly, a brief description will be given of how to manage and treat drug-induced myotoxicity.
Collapse
Affiliation(s)
- Lando Janssen
- Departments of Physiology, Hematology, and Neurology, Radboud University Medical Center, Nijmegen, The Netherlands; and Human and Animal Physiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Neeltje A E Allard
- Departments of Physiology, Hematology, and Neurology, Radboud University Medical Center, Nijmegen, The Netherlands; and Human and Animal Physiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Christiaan G J Saris
- Departments of Physiology, Hematology, and Neurology, Radboud University Medical Center, Nijmegen, The Netherlands; and Human and Animal Physiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Jaap Keijer
- Departments of Physiology, Hematology, and Neurology, Radboud University Medical Center, Nijmegen, The Netherlands; and Human and Animal Physiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Maria T E Hopman
- Departments of Physiology, Hematology, and Neurology, Radboud University Medical Center, Nijmegen, The Netherlands; and Human and Animal Physiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Silvie Timmers
- Departments of Physiology, Hematology, and Neurology, Radboud University Medical Center, Nijmegen, The Netherlands; and Human and Animal Physiology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
18
|
Guo X, Chen Y, Li Q, Yang X, Zhao G, Peng Y, Zheng J. Studies on hepatotoxicity and toxicokinetics of colchicine. J Biochem Mol Toxicol 2019; 33:e22366. [DOI: 10.1002/jbt.22366] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 04/29/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Xiucai Guo
- Wuya College of InnovationShenyang Pharmaceutical University Shenyang China
| | - Yan Chen
- Wuya College of InnovationShenyang Pharmaceutical University Shenyang China
| | - Qingmei Li
- Wuya College of InnovationShenyang Pharmaceutical University Shenyang China
| | - Xiaojing Yang
- Wuya College of InnovationShenyang Pharmaceutical University Shenyang China
| | - Guode Zhao
- School of Pharmaceutical EngineeringShenyang Pharmaceutical University Shenyang China
| | - Ying Peng
- Wuya College of InnovationShenyang Pharmaceutical University Shenyang China
| | - Jiang Zheng
- Wuya College of InnovationShenyang Pharmaceutical University Shenyang China
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical University Guiyang Guizhou China
- Key Laboratory of Pharmaceutics of Guizhou ProvinceGuizhou Medical University Guiyang Guizhou China
| |
Collapse
|
19
|
Extraction, development and validation of HPLC-UV method for rapid and sensitive determination of colchicine from Colchicum autumnale L. Bulbs. Saudi J Biol Sci 2018; 26:345-351. [PMID: 31485175 PMCID: PMC6717106 DOI: 10.1016/j.sjbs.2018.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 09/03/2018] [Accepted: 10/01/2018] [Indexed: 11/20/2022] Open
Abstract
Colchicum autumnale L. also known as the autumn crocus, contains colchicine alkaloid having antifungal properties. The tuber of this plant is rich in terms of colchicine. In this research an ultrasound-assisted extraction (UAE) method was optimized for the extraction of colchicine from Colchicum autumnale L. bulbs before high-performance liquid chromatography with UV detection (HPLC-UV). Optimization of various extraction parameters was performed using response surface methodology (RSM) to evaluate the maximum colchicine yield from Colchicum autumnale L. bulbs. The Box-Behnken design (BBD) and RSM were used to investigate the effect of three key parameters (extraction time (20–60 min), extraction temperature (40–80 °C) and ultrasound power (500–700 W) on extraction efficiency. The variance analysis suggested that the dependent response variable of yield of colchicine may be expressed by a quadratic polynomial model. The optimal theoretical extraction conditions were found to be an ultrasonication power of 602.4 W, an extraction time of 42 min and a temperature of 64 °C. Under these conditions, the optimum foreseen yield was 0.237%. The experimental colchicine yield obtained by following the optimized conditions was found to be 0.238%. These values are very well compatible with each other.
Collapse
|
20
|
Liu H, Dong F, Li G, Niu M, Zhang C, Han Y, He L, Yin P, Wang B, Sang X, Li R, Wang J, Bai Z, Xiao X. Liuweiwuling tablets attenuate BDL-induced hepatic fibrosis via modulation of TGF-β/Smad and NF-κB signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2018; 210:232-241. [PMID: 28864168 DOI: 10.1016/j.jep.2017.08.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liuweiwuling (LWWL) tablets contain a six-herb Chinese formula and are commonly prescribed to facilitate nourishment of the liver and kidneys, clear away toxic materials and activate blood circulation. Administration of LWWL is well known for its protective effects on the liver and its capacity to confer long-term stability in patients exhibiting reduced transaminase levels. Clinical studies have reported that LWWL can also be used for the treatment of liver fibrosis with associated treatment regimens resulting in a concomitant reduction in transforming growth factor β1 (TGF-β1) levels in the serum of patients with hepatic fibrosis. TGF-β1 plays a prominent role in stimulating liver fibrogenesis and this effect is mediated by myofibroblasts (MFB) derived from hepatic stellate cells (HSCs). It is likely that this phenomenon underpins the antifibrotic effects associated with LWWL. AIM The purpose of this study was to investigate the antifibrotic effects and mechanisms pertaining to LWWL. METHODS Hepatic fibrosis was induced in rats following bile duct ligation (BDL). Rats that underwent BDL received daily gavage administration of colchicine (0.2mg/kg per day), LWWL (0.4, 1.6, 6.4g/kg per day) or PBS (for the control group). Pathological changes in hepatic tissue were examined using hematoxylin and eosin (HE) and sirius red staining. Immunohistochemical analysis was performed to monitor α-SMA and type I collagen (Collagen I) protein expression. Real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot analyses were used to monitor the expression of genes and proteins in the TGF-β/Smad signaling pathway, including TGF-β1, bone morphogenic protein and activin membrane-bound inhibitor (Bambi), Smad3, phosphorylated Smad3 (p-Smad3) and Smad7. We also monitored the expression of genes and proteins in the nuclear factor-κB (NF-κB) signaling pathway, including NF-κB p65, IκBα and phosphorylation of IκBα (p-IκBα), tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6) and interleukin 1β (IL-1β). RESULTS LWWL dose-dependently inhibited BDL-induced liver injury and hepatic fibrosis in rats. Furthermore, LWWL reduced liver tissue collagen deposition, hydroxyproline content, liver dysfunction and α-SMA expression in BDL-induced hepatic fibrosis rats. Moreover, LWWL markedly prevented activation of the TGF-β/Smad signaling pathway by inhibiting expression of Smad2/3 and phosphorylation of Smad3, and upregulating the expression of Bambi and Smad7. In addition, LWWL regulated the expression of the inflammatory cytokines IL-1β, TNF-α and IL-6 by inhibiting the activation of NF-κB p65 and the phosphorylation of IκBα, and increasing the expression of IκBα. CONCLUSIONS LWWL can attenuate BDL-induced hepatic fibrosis in rats, and this effect may be due to modulation of the NF-κB-dependent inflammatory response and activation of HSC and TGF-β/Smad-mediated synthesis and degradation of Collagen I.
Collapse
Affiliation(s)
- Huimin Liu
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China; Chengde Medical University Chengde, Hebei, People's Republic of China.
| | - Fang Dong
- Department of Health Statistics, Taishan Medical University, Taian, Shandong, People's Republic of China
| | - Guangquan Li
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Ming Niu
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Congen Zhang
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Yanzhong Han
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Lanzhi He
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Ping Yin
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Bin Wang
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Xiuxiu Sang
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China; Chengde Medical University Chengde, Hebei, People's Republic of China
| | - Ruishen Li
- Animal Laboratory Center, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Jiabo Wang
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Zhaofang Bai
- Department of Pharmacy, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| | - Xiaohe Xiao
- China Military Institute of Chinese Medicine, 302 Hospital of People's Liberation Army, Beijing, People's Republic of China
| |
Collapse
|
21
|
Zhang F, Lu S, He J, Jin H, Wang F, Wu L, Shao J, Chen A, Zheng S. Ligand Activation of PPARγ by Ligustrazine Suppresses Pericyte Functions of Hepatic Stellate Cells via SMRT-Mediated Transrepression of HIF-1α. Am J Cancer Res 2018; 8:610-626. [PMID: 29344293 PMCID: PMC5771080 DOI: 10.7150/thno.22237] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/22/2017] [Indexed: 12/31/2022] Open
Abstract
Rationale: Hepatic stellate cells (HSCs) are liver-specific pericytes regulating vascular remodeling during hepatic fibrosis. Here, we investigated how ligustrazine affects HSC pericyte functions. Methods: Rat HSC-T6 and human HSC-LX2 cells were cultured, and multiple molecular experiments including real-time PCR, Western blot, flow cytometry, immunofluorescence, electrophoretic mobility shift assay and co-immunoprecipitation were used to elucidate the underlying mechanisms. Molecular simulation and site-directed mutagenesis were performed to uncover the target molecule of ligustrazine. Rats were intoxicated with CCl4 for evaluating ligustrazine's effects in vivo. Results: Ligustrazine inhibited angiogenic cytokine production, migration, adhesion and contraction in HSCs, and activated PPARγ. Selective PPARγ inhibitor GW9662 potently abrogated ligustrazine suppression of HSC pericyte functions. Additionally, HIF-1α inhibitor PX-478 repressed HSC pericyte functions, and ligustrazine inhibited the transcription of HIF-1α, which was diminished by GW9662. Moreover, ligustrazine downregulation of HIF-1α was rescued by knockdown of SMRT, and ligustrazine increased PPARγ physical interaction with SMRT, which was abolished by GW9662. These findings collectively indicated that activation of PPARγ by ligustrazine led to transrepression of HIF-1α via a SMRT-dependent mechanism. Furthermore, molecular docking evidence revealed that ligustrazine bound to PPARγ in a unique double-molecule manner via hydrogen bonding with the residues Ser289 and Ser342. Site-directed mutation of Ser289 and/or Ser342 resulted in the loss of ligustrazine transrepression of HIF-1α in HSCs, indicating that interactions with both the residues were indispensable for ligustrazine effects. Finally, ligustrazine improved hepatic injury, angiogenesis and vascular remodeling in CCl4-induced liver fibrosis in rats. Conclusions: We discovered a novel ligand activation pattern for PPARγ transrepression of the target gene with therapeutic implications in HSC pericyte biology and liver fibrosis.
Collapse
|
22
|
Chinese medicine CGA formula ameliorates DMN-induced liver fibrosis in rats via inhibiting MMP2/9, TIMP1/2 and the TGF-β/Smad signaling pathways. Acta Pharmacol Sin 2016; 37:783-93. [PMID: 27133300 DOI: 10.1038/aps.2016.35] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 01/05/2016] [Indexed: 02/07/2023]
Abstract
AIM Chinese medicine CGA formula consists of polysaccharide from Cordyceps sinensis mycelia (CS-PS), gypenosides and amygdalin, which is derived from Fuzheng Huayu (FZHY) capsule for treating liver fibrosis. In this study we attempted to confirm the therapeutic effects of CGA formula in dimethylnitrosamine (DMN)-induced liver fibrosis in rats, and to identify the mechanisms of anti-fibrotic actions. METHODS Rats were injected with DMN (10 mg·kg(-1)·d(-1), ip) for 3 consecutive days per week over a 4-week period. The rats then were orally administered with CGA formula (CS-PS 60 mg·kg(-1)·d(-1), gypenosides 50 mg·kg(-1)·d(-1) and amygdalin 80 mg·kg(-1)·d(-1)) daily in the next 2 weeks. CS-PS, gypenosides or amygdalin alone were administered as individual component controls, whereas colchicine and FZHY were used as positive controls. Serum biomarkers were measured. Hepatic injury, collagen deposition and stellate cell activation were examined. The MMP activities, expression of TIMP protein and proteins involved in the TGF-β1/Smad signaling pathways in liver tissues were assayed. RESULTS In DMN-treated rats, administration of CGA formula significantly decreased serum ALT, AST and total bilirubin and hepatic hydroxyproline levels, increased serum albumin level, and attenuated liver fibrosis as shown by histological examination. Furthermore, these effects were comparable to those caused by administration of FZHY, and superior to those caused by administration of colchicine or the individual components of CGA formula. Moreover, administration of CGA formula significantly decreased the protein levels of α-SMA, TGF-β1, TGF-β1 receptor (TβR-I), p-TβR-I, p-TβR-II, p-Smad2, p-Smad3, TIMP1 and TIMP2, as well as MMP2 and MMP9 activities in liver tissues of DMN-treated rats. CONCLUSION Chinese medicine CGA formula ameliorates DMN-induced liver fibrosis in rats, and this effect was likely associated with the down-regulation of MMP2/9 activities, TIMP1/2 protein expression and the TGF-β1/Smad signaling pathways in the liver.
Collapse
|
23
|
Guo X, Lin D, Li W, Wang K, Peng Y, Zheng J. Electrophilicities and Protein Covalent Binding of Demethylation Metabolites of Colchicine. Chem Res Toxicol 2016; 29:296-302. [PMID: 26845511 DOI: 10.1021/acs.chemrestox.5b00461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Colchicine, an alkaloid existing in plants of Liliaceous colchicum, has been widely used in the treatment of gout and familial Mediterranean fever. The administration of colchicine was found to cause liver injury in humans. The mechanisms of colchicine-induced liver toxicity remain unknown. The objectives of this study were to determine the electrophilicities of demethylation metabolites of colchicine and investigate the protein adductions derived from the reactive metabolites of colchicine. Four demethylated colchicine (1-, 2-, 3-, and 10-DMCs), namely, M1-M4, were detected in colchicine-fortified microsomal incubations. Four N-acetyl cysteine (NAC) conjugates (M5-M8) derived from colchicine were detected in the microsomes in the presence of NAC. M5 and M6 were derived from 10-DMC. M7 resulted from the reaction of 2-DMC or 3-DMC with NAC, and M8 originated from 10-DMC. Microsomal protein covalent binding was observed after exposure to colchicine. Two cysteine adducts (CA-1 and CA-2) derived from 10-DMC were found in proteolytically digested microsomal protein samples after incubation with colchicine. The findings allow us to define the chemical property of demethylation metabolites of colchicine and the interaction between protein and the reactive metabolites of colchicine generated in situ.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiang Zheng
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Division of Gastroenterology and Hepatology, Department of Pediatrics, University of Washington School of Medicine , Seattle, Washington 98101, United States
| |
Collapse
|
24
|
Hemkens LG, Ewald H, Gloy VL, Arpagaus A, Olu KK, Nidorf M, Glinz D, Nordmann AJ, Briel M. Cardiovascular effects and safety of long-term colchicine treatment: Cochrane review and meta-analysis. Heart 2016; 102:590-6. [PMID: 26830663 DOI: 10.1136/heartjnl-2015-308542] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/25/2015] [Indexed: 11/04/2022] Open
Abstract
Colchicine is an old anti-inflammatory drug that has shown substantial cardiovascular benefits in recent trials. We systematically reviewed cardiovascular benefits and harms of colchicine in any population and specifically in patients with high cardiovascular risk. We evaluated randomised controlled trials comparing colchicine over at least 6 months versus any control in any adult population. Primary outcomes were all-cause mortality, myocardial infarction and adverse events. Cardiovascular mortality was a secondary outcome. We included 39 trials with 4992 patients. The quality of evidence for mortality outcomes and myocardial infarction was moderate but lower for adverse events. Colchicine had no effect on all-cause mortality (RR 0.94, 95% CI 0.82 to 1.09; I(2)=27%; 30 trials). Cardiovascular mortality was reduced in some but not all meta-analytical models (random-effects RR 0.34, 0.09 to 1.21, I(2)=9%; Peto's OR 0.24, 0.09 to 0.64, I(2)=15%; Mantel-Haenszel fixed-effect RR 0.20, 0.06 to 0.68, I(2)=0%; 7 trials). The risk for myocardial infarction was reduced (RR 0.20, 0.07 to 0.57; 2 trials). There was no effect on total adverse events (RR 1.52, 0.93 to 2.46, I(2)=45%; 11 trials) but gastrointestinal intolerance was increased (RR 1.83, 1.03 to 3.26, I(2)=74%; 11 trials). Reporting of serious adverse events was inconsistent; no event occurred over 824 patient-years (4 trials). Effects in high cardiovascular risk populations were similar (4 trials; 1230 patients). We found no evidence supporting colchicine doses above 1 mg/day. Colchicine may have substantial cardiovascular benefits; however, there is sufficient uncertainty about its benefit and harm to indicate the need for large-scale trials to further evaluate this inexpensive, promising treatment in cardiovascular disease.
Collapse
Affiliation(s)
- Lars G Hemkens
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Hannah Ewald
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Viktoria L Gloy
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Armon Arpagaus
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Kelechi K Olu
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Mark Nidorf
- Heart Care Western Australia, Perth, Australia
| | - Dominik Glinz
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Alain J Nordmann
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland
| | - Matthias Briel
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Basel, Switzerland Department of Clinical Research, University of Basel, Basel, Switzerland Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Rosato V, Abenavoli L, Federico A, Masarone M, Persico M. Pharmacotherapy of alcoholic liver disease in clinical practice. Int J Clin Pract 2016; 70:119-31. [PMID: 26709723 DOI: 10.1111/ijcp.12764] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIMS Alcohol is the most commonly used addictive substance and alcoholic liver disease (ALD) is a major cause of chronic liver disease worldwide, responsible for 47.9% of all liver chronic deaths. Despite ALD has a significant burden on the health, few therapeutic advances have been made in the last 40 years, particularly in the long-term management of these patients. METHODS we searched in PubMed, Scopus, Google Scholar, and MEDLINE databases to identify relevant English language publications focused on long-term therapy of ALD. RESULTS From the huge literature on this topic, including about 755 studies, 75 were selected as eligible including clinical trials and meta-analysis. CONCLUSIONS Abstinence remains the cornerstone of ALD therapy but it is also the most difficult therapeutic target to achieve and the risk of recidivism is very high at any time. Several drugs (disulfiram, naltrexone, acamprosate, sodium oxybate) have proven to be effective to prevent alcohol relapse and increase the abstinence, although the psychotherapeutic support remains crucial. Baclofen seems to be effective to improve abstinence, showing an excellent safety and tolerability. ALD is often complicated by a state of malnutrition, which is related to a worst mortality. A nutritional therapy may improve survival in cirrhotic patients, reversing muscle wasting, weight loss and specific nutritional deficiencies. While in aggressive forms of alcoholic hepatitis are recommended specific drug treatments, including glucocorticoids or pentoxifylline, for the long-term treatment of ALD, specific treatments aimed at stopping the progression of fibrosis are not yet approved, but there are some future perspective in this field, including probiotics and antibiotics, caspase inhibitors, osteopontin and endocannabinoids.
Collapse
Affiliation(s)
- V Rosato
- Internal Medicine and Hepatology Department, Second University of Naples, Naples, Italy
| | - L Abenavoli
- Department of Health Science, University Magna Graecia, Catanzaro, Italy
| | - A Federico
- Gastroenterology and Endoscopy Unit, Second University of Naples, Naples, Italy
| | - M Masarone
- Internal Medicine and Hepatology Unit, University of Salerno, Baronissi, Italy
| | - M Persico
- Internal Medicine and Hepatology Unit, University of Salerno, Baronissi, Italy
| |
Collapse
|
26
|
Hemkens LG, Ewald H, Gloy VL, Arpagaus A, Olu KK, Nidorf M, Glinz D, Nordmann AJ, Briel M. Colchicine for prevention of cardiovascular events. Cochrane Database Syst Rev 2016; 2016:CD011047. [PMID: 26816301 PMCID: PMC6956668 DOI: 10.1002/14651858.cd011047.pub2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Colchicine is an anti-inflammatory drug that is used for a wide range of inflammatory diseases. Cardiovascular disease also has an inflammatory component but the effects of colchicine on cardiovascular outcomes remain unclear. Previous safety analyses were restricted to specific patient populations. OBJECTIVES To evaluate potential cardiovascular benefits and harms of a continuous long-term treatment with colchicine in any population, and specifically in people with high cardiovascular risk. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, ClinicalTrials.gov, WHO International Clinical Trials Registry, citations of key papers, and study references in January 2015. We also contacted investigators to gain unpublished data. SELECTION CRITERIA Randomised controlled trials (parallel-group or cluster design or first phases of cross-over studies) comparing colchicine over at least six months versus any control in any adult population. DATA COLLECTION AND ANALYSIS Primary outcomes were all-cause mortality, myocardial infarction, and adverse events. Secondary outcomes were cardiovascular mortality, stroke, heart failure, non-scheduled hospitalisations, and non-scheduled cardiovascular interventions. We conducted predefined subgroup analyses, in particular for participants with high cardiovascular risk. . MAIN RESULTS We included 39 randomised parallel-group trials with 4992 participants. Colchicine had no effect on all-cause mortality (RR 0.94, 95% CI 0.82 to 1.09; participants = 4174; studies = 30; I² = 27%; moderate quality of evidence). There is uncertainty surrounding the effect of colchicine in reducing cardiovascular mortality (RR 0.34, 95% CI 0.09 to 1.21, I² = 9%; participants = 1132; studies = 7; moderate quality of evidence). Colchicine reduced the risk for total myocardial infarction (RR 0.20, 95% CI 0.07 to 0.57; participants = 652; studies = 2; moderate quality of evidence). There was no effect on total adverse events (RR 1.52, 95% CI 0.93 to 2.46; participants = 1313; studies = 11; I² = 45%; very low quality of evidence) but gastrointestinal intolerance was increased (RR 1.83, 95% CI 1.03 to 3.26; participants = 1258; studies = 11; I² = 74%; low quality of evidence). Colchicine showed no effect on heart failure (RR 0.62, 95% CI 0.10 to 3.88; participants = 462; studies = 3; I² = 45%; low quality of evidence) and no effect on stroke (RR 0.38, 95% CI 0.09 to 1.70; participants = 874; studies = 3; I² = 45%; low quality of evidence). Reporting of serious adverse events was inconsistent; no event occurred over 824 patient-years (4 trials). Effects on other outcomes were very uncertain. Summary effects of RCTs specifically focusing on participants with high cardiovascular risk were similar (4 trials; 1230 participants). AUTHORS' CONCLUSIONS There is much uncertainty surrounding the benefits and harms of colchicine treatment. Colchicine may have substantial benefits in reducing myocardial infarction in selected high-risk populations but uncertainty about the size of the effect on survival and other cardiovascular outcomes is high, especially in the general population from which most of the studies in our review were drawn. Colchicine is associated with gastrointestinal side effects based on low-quality evidence. More evidence from large-scale randomised trials is needed.
Collapse
Affiliation(s)
- Lars G Hemkens
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, Spitalstrasse 12, Basel, Switzerland, CH-4031
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Weiskirchen R. Hepatoprotective and Anti-fibrotic Agents: It's Time to Take the Next Step. Front Pharmacol 2016; 6:303. [PMID: 26779021 PMCID: PMC4703795 DOI: 10.3389/fphar.2015.00303] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022] Open
Abstract
Hepatic fibrosis and cirrhosis cause strong human suffering and necessitate a monetary burden worldwide. Therefore, there is an urgent need for the development of therapies. Pre-clinical animal models are indispensable in the drug discovery and development of new anti-fibrotic compounds and are immensely valuable for understanding and proofing the mode of their proposed action. In fibrosis research, inbreed mice and rats are by far the most used species for testing drug efficacy. During the last decades, several hundred or even a thousand different drugs that reproducibly evolve beneficial effects on liver health in respective disease models were identified. However, there are only a few compounds (e.g., GR-MD-02, GM-CT-01) that were translated from bench to bedside. In contrast, the large number of drugs successfully tested in animal studies is repeatedly tested over and over engender findings with similar or identical outcome. This circumstance undermines the 3R (Replacement, Refinement, Reduction) principle of Russell and Burch that was introduced to minimize the suffering of laboratory animals. This ethical framework, however, represents the basis of the new animal welfare regulations in the member states of the European Union. Consequently, the legal authorities in the different countries are halted to foreclose testing of drugs in animals that were successfully tested before. This review provides a synopsis on anti-fibrotic compounds that were tested in classical rodent models. Their mode of action, potential sources and the observed beneficial effects on liver health are discussed. This review attempts to provide a reference compilation for all those involved in the testing of drugs or in the design of new clinical trials targeting hepatic fibrosis.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy, and Clinical Chemistry, RWTH University Hospital Aachen Aachen, Germany
| |
Collapse
|
28
|
Gasparyan AY, Ayvazyan L, Yessirkepov M, Kitas GD. Colchicine as an anti-inflammatory and cardioprotective agent. Expert Opin Drug Metab Toxicol 2015; 11:1781-94. [PMID: 26239119 DOI: 10.1517/17425255.2015.1076391] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Colchicine has been successfully used for the treatment of neutrophilic disorders such as familial Mediterranean fever (FMF), Behçet disease (BD) and gout. There is a growing interest in its cardiovascular effects. AREAS COVERED A MEDLINE/PubMed search for English articles published from January 1972 to June 2015 was completed using the following terms: therapy, pharmacokinetics, efficiency, side effects, toxicity, heart, colchicine, inflammation, FMF, amyloidosis, BD, gout, cardiovascular disorders, pericarditis, arrhythmias, inflammation, neutrophils, platelets. EXPERT OPINION By targeting neutrophils, endothelial cells and platelets, inhibiting mitosis, vascular hyperplasia and fibrosis, colchicine improves outcomes of pericarditis, myocardial ischemia and coronary interventions. Studies in neutrophilic rheumatic diseases and cardiovascular disorders demonstrated that oral colchicine at doses of 0.5 - 2.5 mg/daily is useful for treating pericarditis, myocardial ischemia and coronary occlusion. In rheumatic and cardiovascular disorders, therapeutic doses of the drug reduce C-reactive protein to levels below 2 mg/L, prevent myocardial damage and preserve normal values of atrial and ventricular impulse generation. One of the drug's frequent side effects is diarrhea, which is treated by diet modification or temporary discontinuation of the therapy. Certain drugs (macrolides, statins), comorbidities and certain genetic factors increase risk of colchicine toxicity.
Collapse
Affiliation(s)
- Armen Yuri Gasparyan
- a 1 Dudley Group NHS Foundation Trust (Teaching Trust of University of Birmingham), Russells Hall Hospital, Departments of Rheumatology and Research & Development , DY1 2HQ, Dudley, UK +44 138 424 4842 ; +44 138 424 4808 ;
| | - Lilit Ayvazyan
- b 2 Yerevan State Medical University, Department of Medical Chemistry , Yerevan, Armenia
| | - Marlen Yessirkepov
- c 3 South Kazakhstan State Pharmaceutical Academy, Department of Biochemistry, Biology and Microbiology , Shymkent, Kazakhstan
| | - George D Kitas
- a 1 Dudley Group NHS Foundation Trust (Teaching Trust of University of Birmingham), Russells Hall Hospital, Departments of Rheumatology and Research & Development , DY1 2HQ, Dudley, UK +44 138 424 4842 ; +44 138 424 4808 ; .,d 4 University of Manchester, Arthritis Research UK Epidemiology Unit , Manchester, UK
| |
Collapse
|
29
|
Leung YY, Yao Hui LL, Kraus VB. Colchicine--Update on mechanisms of action and therapeutic uses. Semin Arthritis Rheum 2015; 45:341-50. [PMID: 26228647 DOI: 10.1016/j.semarthrit.2015.06.013] [Citation(s) in RCA: 594] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/19/2015] [Accepted: 06/19/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To review the literature and provide an update on the mechanisms of action and therapeutic uses of oral colchicine in arthritis and inflammatory conditions. METHODS We performed PubMed database searches through June 2014 for relevant studies in the English literature published since the last update of colchicine in 2008. Searches encompassed colchicine mechanisms of action and clinical applications in medical conditions. A total of 381 articles were reviewed. RESULTS The primary mechanism of action of colchicine is tubulin disruption. This leads to subsequent down regulation of multiple inflammatory pathways and modulation of innate immunity. Newly described mechanisms include various inhibitory effects on macrophages including the inhibition of the NACHT-LRRPYD-containing protein 3 (NALP3) inflammasome, inhibition of pore formation activated by purinergic receptors P2X7 and P2X2, and stimulation of dendritic cell maturation and antigen presentation. Colchicine also has anti-fibrotic activities and various effects on endothelial function. The therapeutic use of colchicine has extended beyond gouty arthritis and familial Mediterranean fever, to osteoarthritis, pericarditis, and atherosclerosis. CONCLUSION Further understanding of the mechanisms of action underlying the therapeutic efficacy of colchicine will lead to its potential use in a variety of conditions.
Collapse
Affiliation(s)
- Ying Ying Leung
- Department of Rheumatology & Immunology, Singapore General Hospital, The Academia, Level 4, 20 College Rd, Singapore 169856; Department of Clinical Sciences, Duke-NUS Graduate Medical School, Singapore.
| | - Laura Li Yao Hui
- Department of Rheumatology & Immunology, Singapore General Hospital, The Academia, Level 4, 20 College Rd, Singapore 169856
| | - Virginia B Kraus
- Duke Molecular Physiology Institute and Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Durham, NC
| |
Collapse
|
30
|
|
31
|
Sağıroğlu T, Sayhan MB, Yağcı MA, Yalta T, Sağıroğlu G, Çopuroğlu E, Oğuz S. Comparison of sirolimus and colchicine treatment on the development of peritoneal fibrozis in rats having peritoneal dialysis. Balkan Med J 2015; 32:101-6. [PMID: 25759780 DOI: 10.5152/balkanmedj.2015.15183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 11/04/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Continuous ambulatory peritoneal dialysis is a successful treatment modality for patients with end-stage renal disease. Peritoneal fibrosis (PF) is the most critical complication of long-term peritoneal dialysis (PD). AIMS In our study, we aimed to compare the effects of colchicine and sirolimus on PF induced by hypertonic peritoneal dialysis solutions in rats. STUDY DESIGN Animal experiment. METHODS Twenty-four rats were randomly divided into three groups. The control group received an intraperitoneal injection (ip) of saline. The sirolimus group received the PD solution, plus 1.0 mg/kg/day Rapamune®. The colchicine group received the PD solution ip plus 1.0 mg/kg/day of colchicine. Blood samples were taken to measure the serum levels of VEGF, TGF-β, and TNF-α. Peritoneal tissue samples were taken for histopathological evaluation. RESULTS TGF-β and TNF-α values in the sirolimus group were found to be statistically significantly lower than in the control and colchicine groups, but the differences between the control and colchicine groups were not statistically significant. No statistically significant differences were found between the groups regarding the VEGF values. Vascular neogenesis and peritoneal thickness were compared; the values in the sirolimus group were statistically reduced compared to the values in the control group. Mild fibrosis developed in 75% of all animals in the sirolimus group; there was no moderate or severe fibrosis observed. Fibrosis developed to varying degrees in 100% of the animals in the control and colchicine groups. CONCLUSION The present study demonstrates that sirolimus might be beneficial for preventing or delaying the progression of PF and neoangiogenesis. These alterations in the peritoneal membrane may be connected with reduced TNF-α and TGF-β levels.
Collapse
Affiliation(s)
- Tamer Sağıroğlu
- Deptartment of General Surgery, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Mustafa Burak Sayhan
- Deparment of Emergency Medicine, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Mehmet A Yağcı
- Department of General Surgery, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Tülin Yalta
- Department of Pathology, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Gönül Sağıroğlu
- Department of Pathology, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Elif Çopuroğlu
- Department of Anesthesiology, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Serhat Oğuz
- Deptartment of General Surgery, Trakya University Faculty of Medicine, Edirne, Turkey
| |
Collapse
|
32
|
Suk KT, Kim MY, Baik SK. Alcoholic liver disease: Treatment. World J Gastroenterol 2014; 20:12934-12944. [PMID: 25278689 PMCID: PMC4177474 DOI: 10.3748/wjg.v20.i36.12934] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 03/30/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
The excess consumption of alcohol is associated with alcoholic liver diseases (ALD). ALD is a major healthcare problem, personal and social burden, and significant reason for economic loss worldwide. The ALD spectrum includes alcoholic fatty liver, alcoholic hepatitis, cirrhosis, and the development of hepatocellular carcinoma. The diagnosis of ALD is based on a combination of clinical features, including a history of significant alcohol intake, evidence of liver disease, and laboratory findings. Abstinence is the most important treatment for ALD and the treatment plan varies according to the stage of the disease. Various treatments including abstinence, nutritional therapy, pharmacological therapy, psychotherapy, and surgery are currently available. For severe alcoholic hepatitis, corticosteroid or pentoxifylline are recommended based on the guidelines. In addition, new therapeutic targets are being under investigation.
Collapse
|
33
|
Atasever A, Yaman D. The effects of grape seed and colchicine on carbon tetrachloride induced hepatic damage in rats. ACTA ACUST UNITED AC 2014; 66:361-5. [PMID: 24925249 DOI: 10.1016/j.etp.2014.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/26/2014] [Accepted: 04/29/2014] [Indexed: 11/15/2022]
Abstract
This study aims to determine the effects of grape seed and colchicine on carbon tetrachloride (CCl4) induced hepatic damage and on some serum biochemical parameters. Sixty male Wistar albino rats (200-250 g) were randomly divided into six groups (ten rats/group) and included the control group the group were given isotonic sodium chloride (1 mL/kg b.w) intraperitonealy (i.p.), group 2 the group treated i.p. injection of CCl4 (1.0 mL/kg b.w) in corn oil twice in the first week, Groups 3 and 4 injected with CCl4 as described for group 2 and the rats were orally given (100 mg/kg b.w) GSE and i.p. injected (10 μg/rat) with colchicine for four weeks, respectively and groups 5 and 6 were the grape seed and colchicine control groups in which rats were orally given grape seed (100 mg/kg b.w) and i.p. injected with colchicine (10 μg/rat), respectively. Anorexia, weight loss, motionlessness and hepatic colour variation at necropsy were observed in groups 2, 3, and 4. Hyperemia, focal bleeding, fat degeneration, changes ranging from degenerative to necrotic, increase in connective tissue elements, pronounced in portal sites in particular, and infiltration of lymphoid series cell observed in the livers of the rats in group 2, treated with CCl4. Histological hepatic changes in the rats in group 3 and 4 were similar to those in group 2. The levels of serum total protein, albumin and globulin decreased in groups 2, 3, and 4, compared with groups 1, 5 and 6; aspartate transaminase (ALT) activities increased. The lowest alkaline phosphatase (ALP) activities were in groups 4 and 5. We concluded that GSE and colchicine have not sufficient ameliorative effects to CCl4 induced acute hepatic damage.
Collapse
Affiliation(s)
- Ayhan Atasever
- Erciyes University Faculty of Veterinary Medicine, Department of Pathology, Kayseri, Turkey
| | - Duygu Yaman
- Erciyes University Faculty of Veterinary Medicine, Department of Pathology, Kayseri, Turkey.
| |
Collapse
|
34
|
Pandey DK, Malik T, Dey A, Singh J, Banik RM. Improved growth and colchicine concentration in Gloriosa superba on mycorrhizal inoculation supplemented with phosphorus-fertilizer. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2014; 11:439-446. [PMID: 25435630 PMCID: PMC4202654 DOI: 10.4314/ajtcam.v11i2.30] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Gloriosa superba produces an array of alkaloids including colchicine, a compound of interest in the treatment of various diseases. The tuber of Gloriosa superba is a rich source of colchicine which has shown anti-gout, anti-inflammatory, and anti-tumor activity. However, this promising compound remains expensive and Gloriosa superba is such a good source in global scale. Increase in yield of naturally occurring colchicine is an important area of investigation. MATERIALS AND METHODS The effects of inoculation by four arbuscular mycorrhizal (AM), fungi, Glomus mossae, Glomus fasciculatum, Gigaspora margarita and Gigaspora gilmorei either alone or supplemented with P-fertilizer, on colchicine concentration in Gloriosa superba were studied. The concentration of colchicine was determined by high-performance thin layer chromatography. RESULTS The four fungi significantly increased concentration of colchicine in the herb. Although there was significant increase in concentration of colchicine in non-mycorrhizal P-fertilized plants as compared to control, the extent of the increase was less compared to mycorrhizal plants grown with or without P-fertilization. This suggests that the increase in colchicine concentration may not be entirely attributed to enhanced P-nutrition and improved growth. Among the four AM fungi Glomus mossae was found to be best. The total colchicine content of plant (mg / plant) was significantly high in plants inoculated with Glomus mossae and 25 mg kg(-1)phosphorus fertilizer (348.9 mg /plant) while the control contain least colchicine (177.87 mg / plant). CONCLUSION The study suggests a potential role of AM fungi in improving the concentration of colchicine in Gloriosa superba tuber.
Collapse
Affiliation(s)
- Devendra Kumar Pandey
- Department of Biotechnology, School of Biosciences and Biotechnology, Lovely Professional University.Phagwara144402, India
| | - Tabarak Malik
- Department of Biotechnology, School of Biosciences and Biotechnology, Lovely Professional University.Phagwara144402, India
| | - Abhijit Dey
- Department of Botany, Presidency University, Kolkata
| | - Joginder Singh
- Department of Biotechnology, School of Biosciences and Biotechnology, Lovely Professional University.Phagwara144402, India
| | - R M Banik
- School of Biochemical Engineering, Institute of Technology, Banaras Hindu University, Varanasi - 221005, India
| |
Collapse
|
35
|
Novel analogue of colchicine induces selective pro-death autophagy and necrosis in human cancer cells. PLoS One 2014; 9:e87064. [PMID: 24466327 PMCID: PMC3900699 DOI: 10.1371/journal.pone.0087064] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 12/19/2013] [Indexed: 02/06/2023] Open
Abstract
Colchicine, a natural product of Colchicum autumnae currently used for gout treatment, is a tubulin targeting compound which inhibits microtubule formation by targeting fast dividing cells. This tubulin-targeting property has lead researchers to investigate the potential of colchicine and analogs as possible cancer therapies. One major study conducted on an analogue of allocolchicine, ZD 6126, was halted in phase 2 clinical trials due to severe cardio-toxicity associated with treatment. This study involves the development and testing of novel allocolchicine analogues that hold non-toxic anti-cancer properties. Currently we have synthesized and evaluated the anti-cancer activities of two analogues; N-acetyl-O-methylcolchinol (NSC 51046 or NCME), which is structurally similar to ZD 6126, and (S)-3,8,9,10-tetramethoxyallocolchicine (Green 1), which is a novel derivative of allocolchicine that is isomeric in the A ring. NSC 51046 was found to be non-selective as it induced apoptosis in both BxPC-3 and PANC-1 pancreatic cancer cells and in normal human fibroblasts. Interestingly, we found that Green 1 was able to modestly induce pro-death autophagy in these pancreatic cancer cells and E6-1 leukemia cells but not in normal human fibroblasts. Unlike colchicine and NSC 51046, Green 1 does not appear to affect tubulin polymerization indicating that it has a different molecular target. Green 1 also caused increased reactive oxygen species (ROS) production in mitochondria isolated from pancreatic cancer cells. Furthermore, in vivo studies revealed that Green 1 was well tolerated in mice. Our findings suggest that a small change in the structure of colchicine has apparently changed the mechanism of action and lead to improved selectivity. This may lead to better selective treatments in cancer therapy.
Collapse
|
36
|
Peng J, Li X, Feng Q, Chen L, Xu L, Hu Y. Anti-fibrotic effect of Cordyceps sinensis polysaccharide: Inhibiting HSC activation, TGF-β1/Smad signalling, MMPs and TIMPs. Exp Biol Med (Maywood) 2013; 238:668-77. [PMID: 23918878 DOI: 10.1177/1535370213480741] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cordyceps sinensis has been used to treat liver disease in traditional Chinese medicine for thousands of years. Polysaccharide extracted from cultured Cordyceps sinensis mycelia (CS-PS) is the major active components of cordyceps sinensis with anti-liver injury effects. In the present study, the effects of CS-PS on hepatic stellate cell (HSC) activation, transforming growth factor-β1 (TGF-β1)/Smad pathway, as well as matrix metalloproteinase (MMP) 2, MMP9 and tissue inhibitor of metalloproteinase (TIMP) 1, TIMP2, were investigated in liver fibrosis in rats induced by carbon tetrachloride (CCl4). Colchicine was used as a positive control. The effect of CS-PS inhibition liver injury and fibrosis was confirmed by decreasing serum alanine aminotransferase, aspartate aminotransferase, total bilirubin, hepatic hydroxyproline and increasing serum albumin, as well as alleviation of histological changes, which was comparable to that of colchicine. With CS-PS treatment, hepatic α-smooth muscle actin, TGF-β1, TGF-β1 receptor (TβR)-I, TβR-II, p-Smad2, p-Smad3 and TIMP2 proteins expression were down-regulated comparing to that in CCl4 group. The activities of MMP2 and MMP9 in liver tissue were also inhibited in CS-PS-treated group. It is indicated that the effects of CS-PS anti-liver fibrosis are probably associated with the inhibition on HSC activation, TGF-β1/Smads signalling pathway, as well as MMP2, MMP9 activity and TIMP2 expression.
Collapse
Affiliation(s)
- Jinghua Peng
- Institute of Liver diseases, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Cogger VC, Roessner U, Warren A, Fraser R, Le Couteur DG. A Sieve-Raft Hypothesis for the regulation of endothelial fenestrations. Comput Struct Biotechnol J 2013; 8:e201308003. [PMID: 24688743 PMCID: PMC3962122 DOI: 10.5936/csbj.201308003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 07/31/2013] [Accepted: 08/11/2013] [Indexed: 01/13/2023] Open
Affiliation(s)
- Victoria C Cogger
- Centre for Education and Research on Ageing and ANZAC Research Institute, Concord Hospital and University of Sydney, Sydney NSW, Australia ; Charles Perkins Centre, University of Sydney NSW Australia
| | - Ute Roessner
- Metabolomics Australia and Australian Centre for Plant Functional Genomics, The University of Melbourne, 3010 Victoria, Australia
| | - Alessandra Warren
- Centre for Education and Research on Ageing and ANZAC Research Institute, Concord Hospital and University of Sydney, Sydney NSW, Australia ; Charles Perkins Centre, University of Sydney NSW Australia
| | - Robin Fraser
- Christchurch School of Medicine, University of Otago, Christchurch NZ
| | - David G Le Couteur
- Centre for Education and Research on Ageing and ANZAC Research Institute, Concord Hospital and University of Sydney, Sydney NSW, Australia ; Charles Perkins Centre, University of Sydney NSW Australia
| |
Collapse
|
39
|
Translating an understanding of the pathogenesis of hepatic fibrosis to novel therapies. Clin Gastroenterol Hepatol 2013; 11:224-31.e1-5. [PMID: 23305825 PMCID: PMC4151461 DOI: 10.1016/j.cgh.2013.01.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The response to injury is one of wound healing and fibrogenesis, which ultimately leads to fibrosis. The fibrogenic response to injury is a generalized one across virtually all organ systems. In the liver, the injury response, typically occurring over a prolonged period of time, leads to cirrhosis (although it should be pointed out that not all patients with liver injury develop cirrhosis). The fact that many different diseases result in cirrhosis suggests a common pathogenesis. The study of hepatic fibrogenesis over the past 2 decades has been remarkably active, leading to a considerable understanding of this process. It clearly has been shown that the hepatic stellate cell is a central component in the fibrogenic process. It also has been recognized that other effector cells are important in the fibrogenic process, including resident fibroblasts, bone marrow-derived cells, fibrocytes, and even perhaps cells derived from epithelial cells (ie, through epithelial to mesenchymal transition). A key aspect of the biology of fibrogenesis is that the fibrogenic process is dynamic; thus, even advanced fibrosis (or cirrhosis) is reversible. Together, an understanding of the cellular basis for liver fibrogenesis, along with multiple aspects of the basic pathogenesis of fibrosis, have highlighted many exciting potential therapeutic opportunities. Thus, although the most effective antifibrotic therapy is simply treatment of the underlying disease, in situations in which this is not possible, specific antifibrotic therapy is likely not only to become feasible, but will soon become a reality. This review highlights the mechanisms underlying fibrogenesis that may be translated into future antifibrotic therapies and to review the current state of clinical development.
Collapse
|
40
|
Abstract
Alcoholic liver disease is a major cause of morbidity and mortality worldwide. Patients with cirrhosis caused by alcohol are at risk for developing complications associated with a failing liver. The long-term management of alcoholic liver disease stresses the following: (1) Abstinence of alcohol (Grade 1A), with referral to an alcoholic rehabilitation program; (2) Adequate nutritional support (Grade 1B), emphasizing multiple feedings and a referral to a nutritionist; (3) Routine screening in alcoholic cirrhosis to prevent complications; (4) Timely referral to a liver transplant program for those with decompensated cirrhosis; (5) Avoid pharmacologic therapies, as these medications have shown no benefit.
Collapse
Affiliation(s)
- Garmen A Woo
- Center for Liver Diseases, Miller School of Medicine, University of Miami, 1500 Northwest 12th Avenue, Miami, FL 33136, USA.
| | | |
Collapse
|
41
|
Abstract
Alcoholic hepatitis is a form of severe, cholestatic liver disease that results from consumption of large amount of alcohol during a sustained period of time in a subset of alcoholics. Symptoms could be mild and nonspecific to more severe. The diagnosis of alcoholic hepatitis can be made with a thorough history, physical examination, and review of laboratory results. Liver biopsy is confirmatory but generally not indicated for the diagnosis. Abstinence is the key form of therapeutic intervention. Despite variable results in clinical trials, corticosteroids and pentoxifylline seem to provide moderate survival benefit. Liver transplantation in acute alcoholic hepatitis is contentious.
Collapse
Affiliation(s)
- Umair Sohail
- University of Tennessee Health sciences center,1211 Union Avenue, Memphis, TN 38104, USA
| | | |
Collapse
|
42
|
Crittenden DB, Lehmann RA, Schneck L, Keenan RT, Shah B, Greenberg JD, Cronstein BN, Sedlis SP, Pillinger MH. Colchicine use is associated with decreased prevalence of myocardial infarction in patients with gout. J Rheumatol 2012; 39:1458-64. [PMID: 22660810 DOI: 10.3899/jrheum.111533] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The ability of antiinflammatory strategies to alter cardiovascular risk has not been rigorously examined. Colchicine is an antiinflammatory agent that affects macrophages, neutrophils, and endothelial cells, all of which are implicated in the pathogenesis of cardiovascular disease. We examined whether colchicine use was associated with a reduced risk of myocardial infarction (MI) in patients with gout. METHODS We conducted a retrospective, cross-sectional study of all patients with an International Classification of Diseases, 9th ed, code for gout in the electronic medical record (EMR) of the New York Harbor Healthcare System Veterans Affairs network and ≥ 1 hospital visit between August 2007 and August 2008. Hospital pharmacy data were used to identify patients who had filled at least 1 colchicine prescription versus those who had not. Demographics and CV comorbidities were collected by EMR review. The primary outcome was diagnosis of MI. Secondary outcomes included all-cause mortality and C-reactive protein (CRP) level. RESULTS In total, 1288 gout patients were identified. Colchicine (n = 576) and no colchicine (n = 712) groups had similar baseline demographics and serum urate levels. Prevalence of MI was 1.2% in the colchicine versus 2.6% in the no-colchicine group (p = 0.03). Colchicine users also had fewer deaths and lower CRP levels, although these did not achieve statistical significance. Colchicine effects persisted when allopurinol users were excluded from the analysis. CONCLUSION In this hypothesis-generating study, gout patients who took colchicine had a significantly lower prevalence of MI and exhibited trends toward reduced all-cause mortality and lower CRP level versus those who did not take colchicine.
Collapse
Affiliation(s)
- Daria B Crittenden
- Crystal Diseases Study Group, Division of Rheumatology, New York University School of Medicine, New York, New York 10003, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Zheng B, Tan L, Mo X, Yu W, Wang Y, Tucker-Kellogg L, Welsch RE, So PTC, Yu H. Predicting in vivo anti-hepatofibrotic drug efficacy based on in vitro high-content analysis. PLoS One 2011; 6:e26230. [PMID: 22073152 PMCID: PMC3206809 DOI: 10.1371/journal.pone.0026230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 09/22/2011] [Indexed: 01/11/2023] Open
Abstract
Background/Aims Many anti-fibrotic drugs with high in vitro efficacies fail to produce significant effects in vivo. The aim of this work is to use a statistical approach to design a numerical predictor that correlates better with in vivo outcomes. Methods High-content analysis (HCA) was performed with 49 drugs on hepatic stellate cells (HSCs) LX-2 stained with 10 fibrotic markers. ∼0.3 billion feature values from all cells in >150,000 images were quantified to reflect the drug effects. A systematic literature search on the in vivo effects of all 49 drugs on hepatofibrotic rats yields 28 papers with histological scores. The in vivo and in vitro datasets were used to compute a single efficacy predictor (Epredict). Results We used in vivo data from one context (CCl4 rats with drug treatments) to optimize the computation of Epredict. This optimized relationship was independently validated using in vivo data from two different contexts (treatment of DMN rats and prevention of CCl4 induction). A linear in vitro-in vivo correlation was consistently observed in all the three contexts. We used Epredict values to cluster drugs according to efficacy; and found that high-efficacy drugs tended to target proliferation, apoptosis and contractility of HSCs. Conclusions The Epredict statistic, based on a prioritized combination of in vitro features, provides a better correlation between in vitro and in vivo drug response than any of the traditional in vitro markers considered.
Collapse
Affiliation(s)
- Baixue Zheng
- Computation and Systems Biology Program, Singapore-MIT Alliance, National University of Singapore, Singapore, Singapore
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Looling Tan
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, Singapore
| | - Xuejun Mo
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, Singapore
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Weimiao Yu
- Imaging Informatics Division, Bioinformatics Institute, A*STAR, Singapore, Singapore
- Central Imaging Facility, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Yan Wang
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, Singapore
- Department of Hepatobiliary Surgery, Southern Medical University Affiliated Zhujiang Hospital, Guangzhou, China
| | - Lisa Tucker-Kellogg
- Computation and Systems Biology Program, Singapore-MIT Alliance, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Roy E. Welsch
- Computation and Systems Biology Program, Singapore-MIT Alliance, National University of Singapore, Singapore, Singapore
- Engineering Systems Division, Sloan School of Management, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Peter T. C. So
- Computation and Systems Biology Program, Singapore-MIT Alliance, National University of Singapore, Singapore, Singapore
- Singapore-MIT Alliance for Research and Technology, BioSyM, Singapore, Singapore
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Hanry Yu
- Computation and Systems Biology Program, Singapore-MIT Alliance, National University of Singapore, Singapore, Singapore
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Singapore-MIT Alliance for Research and Technology, BioSyM, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences, National University of Singapore, Singapore, Singapore
- NUS Tissue-Engineering Programme, National University of Singapore, Singapore, Singapore
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
45
|
Cohen-Naftaly M, Friedman SL. Current status of novel antifibrotic therapies in patients with chronic liver disease. Therap Adv Gastroenterol 2011; 4:391-417. [PMID: 22043231 PMCID: PMC3187682 DOI: 10.1177/1756283x11413002] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Fibrosis accumulation is a dynamic process resulting from a wound-healing response to acute or chronic liver injury of all causes. The cascade starts with hepatocyte necrosis and apoptosis, which instigate inflammatory signaling by chemokines and cytokines, recruitment of immune cell populations, and activation of fibrogenic cells, culminating in the deposition of extracellular matrix. These key elements, along with pathways of transcriptional and epigenetic regulation, represent fertile therapeutic targets. New therapies include drugs specifically designed as antifibrotics, as well as drugs already available with well-established safety profiles, whose mechanism of action may also be antifibrotic. At the same time, the development of noninvasive fibrogenic markers, and techniques (e.g. fibroscan), as well as combined scoring systems incorporating serum and clinical features will allow improved assessment of therapy response. In aggregate, the advances in the elucidation of the biology of fibrosis, combined with improved technologies for assessment will provide a comprehensive framework for design of antifibrotics and their analysis in well-designed clinical trials. These efforts may ultimately yield success in halting the progression of, or reversing, liver fibrosis.
Collapse
Affiliation(s)
| | - Scott L. Friedman
- Fishberg Professor of Medicine, Division of Liver Diseases, Box 1123, Mount Sinai School of Medicine, 1425 Madison Avenue, Room 11-70C, New York, NY 10029-6574, USA
| |
Collapse
|
46
|
Cocco G, Chu DCC, Pandolfi S. Colchicine in clinical medicine. A guide for internists. Eur J Intern Med 2010; 21:503-8. [PMID: 21111934 DOI: 10.1016/j.ejim.2010.09.010] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/07/2010] [Accepted: 09/15/2010] [Indexed: 12/24/2022]
Abstract
Colchicine (COL) has been used in medicine for a long time. It is well recognized as a valid therapy in acute flares of gouty arthritis, familial Mediterranean fever (FMF), Behçet's disease, and recurring pericarditis with effusion. It has also been used to treat many inflammatory disorders prone to fibrosis, mostly with disappointing therapeutic results. The pharmacotherapeutic mechanism of action of COL in diverse diseases is not fully understood, thought it is known that the drug accumulates preferentially in neutrophils, and this effect is useful in FMF. COL shows a large interindividual bioavailability. Furthermore, interactions with drugs interfering with CYP3A4 dependent enzymes and P-glycoprotein occur and are clinically important. The dosage of COL must be reduced in patients with relevant hepatic and/or renal dysfunction. However, when appropriately used and contraindications have been excluded, oral COL is a safe treatment.
Collapse
|
47
|
Use of colchicine to prevent recurrence of ear keloids. A new approach. J Plast Reconstr Aesthet Surg 2010; 63:e650-2. [DOI: 10.1016/j.bjps.2010.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Revised: 03/03/2010] [Accepted: 03/05/2010] [Indexed: 11/21/2022]
|
48
|
Muntoni S, Rojkind M, Muntoni S. Colchicine reduces procollagen III and increases pseudocholinesterase in chronic liver disease. World J Gastroenterol 2010; 16:2889-94. [PMID: 20556834 PMCID: PMC2887584 DOI: 10.3748/wjg.v16.i23.2889] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To test whether colchicine would be an effective antifibrotic agent for treatment of chronic liver diseases in patients who could not be treated with α-interferon.
METHODS: Seventy-four patients (46 males, 28 females) aged 40-66 years (mean 53 ± 13 years) participated in the study. The patients were affected by chronic liver diseases with cirrhosis which was proven histologically (n = 58); by chronic active hepatitis C (n = 4), chronic active hepatitis B (n = 2), and chronic persistent hepatitis C (n = 6). In the four patients lacking histology, cirrhosis was diagnosed from anamnesis, serum laboratory tests, esophageal varices and ascites. Patients were assigned to colchicine (1 mg/d) or standard treatment as control in a randomized, double-blind trial, and followed for 4.4 years with clinical and laboratory evaluation.
RESULTS: Survival at the end of the study was 94.6% in the colchicine group and 78.4% in the control group (P = 0.001). Serum N-terminal peptide of type III procollagen levels fell from 34.0 to 18.3 ng/mL (P = 0.0001), and pseudocholinesterase levels rose from 4.900 to 5.610 mU/mL (P = 0.0001) in the colchicine group, while no significant change was seen in controls. Best results were obtained in patients with chronic hepatitis C and in alcoholic cirrhotics.
CONCLUSION: Colchicine is an effective and safe antifibrotic drug for long-term treatment of chronic liver disease in which fibrosis progresses towards cirrhosis.
Collapse
|
49
|
Abstract
Fibrosis is a pathological process that includes scar formation and overproduction of extracellular matrix by the connective tissue as a response to tissue damage. The fibrotic process involves multiple organs and results in progressive life-threatening diseases. Today, we know more about the molecular mechanism that leads to fibrosis involving different type of cells, cytokines, chemokines, and tissue enzymes. Fibrosis was considered an irreversible process, at least clinically, and is still usually treated by anti-inflammatory and immunosuppressive agents. No proven antifibrotic therapy has shown efficacy in ameliorating the clinical course of fibrotic diseases, but our current understanding led to the development of different drugs with promising results, like: mycophenolate mofetil, interferon, relaxin, and intravenous immunoglobulin. This review will provide a glance to this heavily investigated subject.
Collapse
Affiliation(s)
- Ziv Paz
- Department of Medicine B, Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
| | | |
Collapse
|
50
|
Shorbagi A, Bayraktar Y. Experience of a single center with congenital hepatic fibrosis: A review of the literature. World J Gastroenterol 2010; 16:683-90. [PMID: 20135715 PMCID: PMC2817055 DOI: 10.3748/wjg.v16.i6.683] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Congenital hepatic fibrosis (CHF) is an autosomal recessive inherited malformation defined pathologically by a variable degree of periportal fibrosis and irregularly shaped proliferating bile ducts. It is one of the fibropolycystic diseases, which also include Caroli disease, autosomal dominant polycystic kidney disease, and autosomal recessive polycystic kidney disease. Clinically it is characterized by hepatic fibrosis, portal hypertension, and renal cystic disease. CHF is known to occur in association with a range of both inherited and non-inherited disorders, with multiorgan involvement, as a result of ductal plate malformation. Because of the similarities in the clinical picture, it is necessary to differentiate CHF from idiopathic portal hypertension and early liver cirrhosis, for which a liver biopsy is essential. Radiological tests are important for recognizing involvement of other organ systems. With regards to our experience at Hacettepe University, a total of 26 patients have been diagnosed and followed-up between 1974 and 2009 with a diagnosis of CHF. Presentation with Caroli syndrome was the most common diagnosis, with all such patients presenting with symptoms of recurrent cholangitis and symptoms related to portal hypertension. Although portal fibrosis is known to contribute to the ensuing portal hypertension, it is our belief that portal vein cavernous transformation also plays an important role in its pathogenesis. In all patients with CHF portal vein morphology should be evaluated by all means since portal vein involvement results in more severe and complicated portal hypertension. Other associations include the Joubert and Bardet-Biedl syndromes.
Collapse
|