1
|
Kovács P, Schwarcz S, Nyerges P, Bíró TI, Ujlaki G, Bai P, Mikó E. Anticarcinogenic effects of ursodeoxycholic acid in pancreatic adenocarcinoma cell models. Front Cell Dev Biol 2024; 12:1487685. [PMID: 39723238 PMCID: PMC11668698 DOI: 10.3389/fcell.2024.1487685] [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: 08/28/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Changes to the composition of the microbiome in neoplasia, is termed oncobiosis, may affect tumor behavior through the changes to the secretion of bacterial metabolites. In this study we show, that ursodeoxycholic acid (UDCA), a bacterial metabolite, has cytostatic properties in pancreatic adenocarcinoma cell (PDAC) models. UDCA in concentrations corresponding to the human serum reference range suppressed PDAC cell proliferation. UDCA inhibited the expression of epithelial mesenchymal transition (EMT)-related markers and invasion capacity of PDAC cells. UDCA treatment increased oxidative/nitrosative stress by reducing the expression of nuclear factor, erythroid 2-like 2 (NRF2), inducing inducible nitric oxide synthase (iNOS) and nitrotyrosine levels and enhancing lipid peroxidation. Furthermore, UDCA reduced the expression of cancer stem cell markers and the proportion of cancer stem cells. Suppression of oxidative stress by antioxidants, blunted the UDCA-induced reduction in cancer stemness. Finally, we showed that UDCA induced mitochondrial oxidative metabolism. UDCA did not modulate the effectiveness of chemotherapy agents used in the chemotherapy treatment of pancreatic adenocarcinoma. The antineoplastic effects of UDCA, observed here, may contribute to the induction of cytostasis in PDAC cell models by providing a more oxidative/nitrosative environment.
Collapse
Affiliation(s)
- Patrik Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szandra Schwarcz
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petra Nyerges
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Ingrid Bíró
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Belka M, Gostyńska-Stawna A, Stawny M, Krajka-Kuźniak V. Activation of Nrf2 and FXR via Natural Compounds in Liver Inflammatory Disease. Int J Mol Sci 2024; 25:11213. [PMID: 39456994 PMCID: PMC11508530 DOI: 10.3390/ijms252011213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Liver inflammation is frequently linked to oxidative stress and dysregulation of bile acid and fatty acid metabolism. This review focuses on the farnesoid X receptor (FXR), a critical regulator of bile acid homeostasis, and its interaction with the nuclear factor erythroid 2-related factor 2 (Nrf2), a key modulator of cellular defense against oxidative stress. The review explores the interplay between FXR and Nrf2 in liver inflammatory diseases, highlighting the potential therapeutic effects of natural FXR agonists. Specifically, compounds such as auraptene, cafestol, curcumin, fargesone A, hesperidin, lycopene, oleanolic acid, resveratrol, rutin, ursolic acid, and withaferin A are reviewed for their ability to modulate both the FXR and Nrf2 pathways. This article discusses their potential to alleviate liver inflammation, oxidative stress, and damage in diseases such as metabolic-associated fatty liver disease (MAFLD), cholestatic liver injury, and viral hepatitis. In addition, we address the molecular mechanisms driving liver inflammation, including oxidative stress, immune responses, and bile acid accumulation, while also summarizing relevant experimental models. This review emphasizes the promising therapeutic potential of targeting both the Nrf2 and FXR pathways using natural compounds, paving the way for future treatments for liver diseases. Finally, the limitations of the clinical application were indicated, and further research directions were proposed.
Collapse
Affiliation(s)
- Marta Belka
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Aleksandra Gostyńska-Stawna
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| |
Collapse
|
3
|
Liu X, Qiu X, Yang Y, Wang J, Wang Q, Liu J, Yang F, Liu Z, Qi R. Alteration of gut microbiome and metabolome by Clostridium butyricum can repair the intestinal dysbiosis caused by antibiotics in mice. iScience 2023; 26:106190. [PMID: 36895644 PMCID: PMC9988658 DOI: 10.1016/j.isci.2023.106190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/24/2022] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
This study evaluated the repair effects of Clostridium butyricum (CBX 2021) on the antibiotic (ABX)-induced intestinal dysbiosis in mice by the multi-omics method. Results showed that ABX eliminated more than 90% of cecal bacteria and also exerted adverse effects on the intestinal structure and overall health in mice after 10 days of the treatment. Of interest, supplementing CBX 2021 in the mice for the next 10 days colonized more butyrate-producing bacteria and accelerated butyrate production compared with the mice by natural recovery. The reconstruction of intestinal microbiota efficiently promoted the improvement of the damaged gut morphology and physical barrier in the mice. In addition, CBX 2021 significantly reduced the content of disease-related metabolites and meanwhile promoted carbohydrate digestion and absorption in mice followed the microbiome alternation. In conclusion, CBX 2021 can repair the intestinal ecology of mice damaged by the antibiotics through reconstructing gut microbiota and optimizing metabolic functions.
Collapse
Affiliation(s)
- Xin Liu
- Chongqing Academy of Animal Science, Chongqing 402460, China.,College of Animal Science and Technology, Southwest University, Chongqing 402460, China
| | - Xiaoyu Qiu
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Yong Yang
- College of Life Sciences, Southwest University of Science and Technology, Mianyang 621000, China
| | - Jing Wang
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Qi Wang
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Jingbo Liu
- College of Life Sciences, Southwest University of Science and Technology, Mianyang 621000, China
| | - Feiyun Yang
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Zuohua Liu
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| | - Renli Qi
- Chongqing Academy of Animal Science, Chongqing 402460, China.,National Pig Technology Innovation Center, Chongqing 402460, China
| |
Collapse
|
4
|
Weber S, Gerbes AL. Challenges and Future of Drug-Induced Liver Injury Research-Laboratory Tests. Int J Mol Sci 2022; 23:ijms23116049. [PMID: 35682731 PMCID: PMC9181520 DOI: 10.3390/ijms23116049] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
Drug-induced liver injury (DILI) is a rare but potentially severe adverse drug event, which is also a major cause of study cessation and market withdrawal during drug development. Since no acknowledged diagnostic tests are available, DILI diagnosis poses a major challenge both in clinical practice as well as in pharmacovigilance. Differentiation from other liver diseases and the identification of the causative agent in the case of polymedication are the main issues that clinicians and drug developers face in this regard. Thus, efforts have been made to establish diagnostic testing methods and biomarkers in order to safely diagnose DILI and ensure a distinguishment from alternative liver pathologies. This review provides an overview of the diagnostic methods used in differential diagnosis, especially with regards to autoimmune hepatitis (AIH) and drug-induced autoimmune hepatitis (DI-AIH), in vitro causality methods using individual blood samples, biomarkers for diagnosis and severity prediction, as well as experimental predictive models utilized in pre-clinical settings during drug development regimes.
Collapse
|
5
|
Xiao R, Lei K, Kuok H, Deng W, Zhuang Y, Tang Y, Guo Z, Qin H, Bai L, Li T. Synthesis and identification of lithocholic acid 3‐sulfate as RORγt ligand to inhibit Th17 cell differentiation. J Leukoc Biol 2022; 112:835-843. [DOI: 10.1002/jlb.1ma0122-513r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/27/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022] Open
Affiliation(s)
- Riping Xiao
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Kawai Lei
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Hioha Kuok
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Wende Deng
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Yuxin Zhuang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Yanqing Tang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Zhengyang Guo
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
| | - Hongyan Qin
- Department of Pharmacy First Hospital of Lanzhou University Lanzhou China
| | - Li‐Ping Bai
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
- Guangdong‐Hong Kong‐Macao Joint Laboratory of Respiratory Infectious Disease Macau University of Science and Technology Macau China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health Macau University of Science and Technology Macau China
- Guangdong‐Hong Kong‐Macao Joint Laboratory of Respiratory Infectious Disease Macau University of Science and Technology Macau China
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China Macau University of Science and Technology Macau China
| |
Collapse
|
6
|
Zhang P, Zheng L, Duan Y, Gao Y, Gao H, Mao D, Luo Y. Gut microbiota exaggerates triclosan-induced liver injury via gut-liver axis. JOURNAL OF HAZARDOUS MATERIALS 2022; 421:126707. [PMID: 34315018 DOI: 10.1016/j.jhazmat.2021.126707] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/26/2021] [Accepted: 07/18/2021] [Indexed: 06/13/2023]
Abstract
Triclosan (TCS) is an antimicrobial ingredient that has been widely incorporated in consumer products. TCS can cause hepatic damage by disturbing lipid metabolism, which is often accompanied with gut microbiota dysbiosis. However, the effects of gut microbiota on the TCS-induced liver injury are still unknown. Therefore, we constructed a mouse model based on five-week-old male C57BL/6 mice to investigate the effects of dietary TCS exposure (40 ppm) on liver injury. We found that TCS treatment for 4 weeks dramatically disturbed gut microbiota homeostasis, resulting in overproduction of lipopolysaccharides (LPS) and deficiency of secondary bile acids such as deoxycholic acid (DCA) and lithocholic acid (LCA). In addition, TCS considerably increased intestinal permeability by reducing mucus excretion and expression of tight junction proteins (ZO-1, occludin and claudin 4), which facilitated translocation of LPS. The LPS accumulation in blood contributed to liver injury by triggering the inflammatory response via TLR4 pathway. In summary, this study provides novel insights into the underlying mechanisms of TCS-associated liver injury induced by gut microbiota via the gut-liver axis, and contributes to better interpretation of the health impact of the environmentally emerging contaminant TCS.
Collapse
Affiliation(s)
- Peng Zhang
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210046, China
| | - Liyang Zheng
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yitao Duan
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China
| | - Yuting Gao
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China
| | - Huihui Gao
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin 300071, China.
| | - Yi Luo
- College of Environmental Sciences and Engineering, Nankai University, Tianjin 300350, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210046, China.
| |
Collapse
|
7
|
Koelfat KV, Picot D, Chang X, Desille‐Dugast M, van Eijk HM, van Kuijk SM, Lenicek M, Layec S, Carsin M, Dussaulx L, Seynhaeve E, Trivin F, Lacaze L, Thibault R, Schaap FG, Olde Damink SW. Chyme Reinfusion Restores the Regulatory Bile Salt-FGF19 Axis in Patients With Intestinal Failure. Hepatology 2021; 74:2670-2683. [PMID: 34133768 PMCID: PMC8596508 DOI: 10.1002/hep.32017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/20/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Automated chyme reinfusion (CR) in patients with intestinal failure (IF) and a temporary double enterostomy (TDE) restores intestinal function and protects against liver injury, but the mechanisms are incompletely understood. The aim was to investigate whether the beneficial effects of CR relate to functional recovery of enterohepatic signaling through the bile salt-FGF19 axis. APPROACH AND RESULTS Blood samples were collected from 12 patients, 3 days before, at start, and 1, 3, 5, and 7 weeks after CR initiation. Plasma FGF19, total bile salts (TBS), 7-α-hydroxy-4-cholesten-3-one (C4; a marker of bile salt synthesis), citrulline (CIT), bile salt composition, liver tests, and nutritional risk indices were determined. Paired small bowel biopsies prior to CR and after 21 days were taken, and genes related to bile salt homeostasis and enterocyte function were assessed. CR induced an increase in plasma FGF19 and decreased C4 levels, indicating restored regulation of bile salt synthesis through endocrine FGF19 action. TBS remained unaltered during CR. Intestinal farnesoid X receptor was up-regulated after 21 days of CR. Secondary and deconjugated bile salt fractions were increased after CR, reflecting restored microbial metabolism of host bile salts. Furthermore, CIT and albumin levels gradually rose after CR, while abnormal serum liver tests normalized after CR, indicating restored intestinal function, improved nutritional status, and amelioration of liver injury. CR increased gene transcripts related to enterocyte number, carbohydrate handling, and bile salt homeostasis. Finally, the reciprocal FGF19/C4 response after 7 days predicted the plasma CIT time course. CONCLUSIONS CR in patients with IF-TDE restored bile salt-FGF19 signaling and improved gut-liver function. Beneficial effects of CR are partly mediated by recovery of the bile salt-FGF19 axis and subsequent homeostatic regulation of bile salt synthesis.
Collapse
Affiliation(s)
- Kiran V.K. Koelfat
- Department of SurgeryMaastricht University Medical CenterMaastrichtthe Netherlands,NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Denis Picot
- Department of Nutritional and Digestive RehabilitationClinique Saint YvesRennesFrance
| | - Xinwei Chang
- Department of SurgeryMaastricht University Medical CenterMaastrichtthe Netherlands,NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Mireille Desille‐Dugast
- INSERM, INRAEUniversity of RennesNutrition Metabolisms and CancerNuMeCanNutrition UnitCRB SantéCHU RennesRennesFrance
| | - Hans M. van Eijk
- Department of SurgeryMaastricht University Medical CenterMaastrichtthe Netherlands,NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Sander M.J. van Kuijk
- Department of Clinical Epidemiology and Medical Technology AssessmentMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Martin Lenicek
- Institute of Medical Biochemistry and Laboratory Diagnostics1st Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Sabrina Layec
- Department of Nutritional and Digestive RehabilitationClinique Saint YvesRennesFrance
| | - Marie Carsin
- Department of Nutritional and Digestive RehabilitationClinique Saint YvesRennesFrance
| | - Laurence Dussaulx
- Department of Nutritional and Digestive RehabilitationClinique Saint YvesRennesFrance
| | - Eloi Seynhaeve
- Department of Nutritional and Digestive RehabilitationClinique Saint YvesRennesFrance
| | - Florence Trivin
- Department of Nutritional and Digestive RehabilitationClinique Saint YvesRennesFrance
| | - Laurence Lacaze
- INSERM, INRAEUniversity of RennesNutrition Metabolisms and CancerNuMeCanNutrition UnitCRB SantéCHU RennesRennesFrance
| | - Ronan Thibault
- INSERM, INRAEUniversity of RennesNutrition Metabolisms and CancerNuMeCanNutrition UnitCRB SantéCHU RennesRennesFrance
| | - Frank G. Schaap
- Department of SurgeryMaastricht University Medical CenterMaastrichtthe Netherlands,NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands,Department of General, Visceral and Transplantation SurgeryRWTH University Hospital AachenAachenGermany
| | - Steven W.M. Olde Damink
- Department of SurgeryMaastricht University Medical CenterMaastrichtthe Netherlands,NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands,Department of General, Visceral and Transplantation SurgeryRWTH University Hospital AachenAachenGermany
| |
Collapse
|
8
|
Santos Silva E, Moreira Silva H, Catarino C, Dias CC, Santos-Silva A, Lopes AI. Neonatal cholestasis: development of a diagnostic decision algorithm from multivariate predictive models. Eur J Pediatr 2021; 180:1477-1486. [PMID: 33410939 DOI: 10.1007/s00431-020-03886-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/06/2020] [Accepted: 11/20/2020] [Indexed: 11/28/2022]
Abstract
Despite the recent advances involving molecular studies, the neonatal cholestasis (NC) diagnosis still relays on the expertise of medical teams. Our aim was to develop models of etiological diagnosis and unfavourable prognosis which may support a rationale diagnostic approach. We retrospectively analysed 154 patients born between January 1985 and October 2019. The cohort was divided into two main groups: (A) transient cholestasis and (B) other diagnosis (with subgroups) and also in two groups of outcomes: (I) unfavourable and (II) favourable. Multivariate logistic regression analysis identified the lower gestational age as the only variable independently associated with an increased risk of transient cholestasis and signs and/or symptoms of sepsis with infectious or metabolic diseases. Gamma-glutamyl transferase serum levels > 300 IU/L had a positive predictive value for both diagnosis of biliary atresia and for alpha-1-antitrypsin deficiency (A1ATD) and for unfavourable prognosis. A model of diagnosis for A1ATD (n = 34) showed an area under the ROC curve = 0.843 [confidence interval (CI): 0.773-0.912].Conclusion: This study identified some predictors of diagnosis and prognosis which helped to build a diagnostic decision algorithm. The unusually large subgroup of patients with A1ATD in this cohort emphasizes its predictive diagnostic model. What Is Known • The etiological diagnosis of neonatal cholestasis (NC) requires a step-by-step guided approach, and diagnostic models have been developed only for biliary atresia. • Current algorithms neither address the epidemiology changes nor the application of the new molecular diagnostic tools. What Is New • This study provides diagnostic predictive models for patients with A1ATD, metabolic/infectious diseases, and transient cholestasis, and two models of unfavourable prognosis for NC. • A diagnostic decision algorithm is proposed based on this study, authors expertise and the literature.
Collapse
Affiliation(s)
- Ermelinda Santos Silva
- Gastroenterology Unit, Paediatrics Division, Child and Adolescent Department, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, n° 45, 4050-651, Porto, Portugal. .,Integrated Master in Medicine, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313, Porto, Portugal. .,UCIBIO, REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313, Porto, Portugal.
| | - Helena Moreira Silva
- Gastroenterology Unit, Paediatrics Division, Child and Adolescent Department, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, n° 45, 4050-651, Porto, Portugal
| | - Cristina Catarino
- UCIBIO, REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313, Porto, Portugal
| | - Cláudia Camila Dias
- MEDCIDS (Departamento de Medicina da Comunidade, Informação e Decisão em Saúde) and CINTESIS (Centro de Investigação em Tecnologias e em Serviços de Saúde), Faculdade de Medicina da Universidade do Porto, Rua Dr Plácido da Costa, s/n, 4200-450, Porto, Portugal
| | - Alice Santos-Silva
- UCIBIO, REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313, Porto, Portugal
| | - Ana-Isabel Lopes
- Paediatric Gastroenterology Unit, Paediatrics Department, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Av. Prof. Egas Moniz, 1600-190, Lisboa, Portugal.,Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| |
Collapse
|
9
|
Jee A, Sernoskie SC, Uetrecht J. Idiosyncratic Drug-Induced Liver Injury: Mechanistic and Clinical Challenges. Int J Mol Sci 2021; 22:ijms22062954. [PMID: 33799477 PMCID: PMC7998339 DOI: 10.3390/ijms22062954] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (IDILI) remains a significant problem for patients and drug development. The idiosyncratic nature of IDILI makes mechanistic studies difficult, and little is known of its pathogenesis for certain. Circumstantial evidence suggests that most, but not all, IDILI is caused by reactive metabolites of drugs that are bioactivated by cytochromes P450 and other enzymes in the liver. Additionally, there is overwhelming evidence that most IDILI is mediated by the adaptive immune system; one example being the association of IDILI caused by specific drugs with specific human leukocyte antigen (HLA) haplotypes, and this may in part explain the idiosyncratic nature of these reactions. The T cell receptor repertoire likely also contributes to the idiosyncratic nature. Although most of the liver injury is likely mediated by the adaptive immune system, specifically cytotoxic CD8+ T cells, adaptive immune activation first requires an innate immune response to activate antigen presenting cells and produce cytokines required for T cell proliferation. This innate response is likely caused by either a reactive metabolite or some form of cell stress that is clinically silent but not idiosyncratic. If this is true it would make it possible to study the early steps in the immune response that in some patients can lead to IDILI. Other hypotheses have been proposed, such as mitochondrial injury, inhibition of the bile salt export pump, unfolded protein response, and oxidative stress although, in most cases, it is likely that they are also involved in the initiation of an immune response rather than representing a completely separate mechanism. Using the clinical manifestations of liver injury from a number of examples of IDILI-associated drugs, this review aims to summarize and illustrate these mechanistic hypotheses.
Collapse
Affiliation(s)
- Alison Jee
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | | | - Jack Uetrecht
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada;
- Correspondence:
| |
Collapse
|
10
|
Mann S, Ramsay JD, Wakshlag JJ, Stokol T, Reed S, Divers TJ. Investigating the pathogenesis of high-serum gamma-glutamyl transferase activity in Thoroughbred racehorses: A series of case-control studies. Equine Vet J 2021; 54:39-51. [PMID: 33555643 DOI: 10.1111/evj.13435] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND High-serum γ-Glutamyl Transferase (GGT) activity has been associated with and thought to be a marker of maladaptation to training and possibly poor performance in racehorses, but the cause is unknown. OBJECTIVES To investigate possible metabolic and infectious causes for the high GGT syndrome. STUDY DESIGN Pilot case-control study and nested case-control study. METHODS The case-control study in 2017 included 16 horses (8 cases and 8 controls with median [range] serum GGT 82 [74-148] and 22 [19-28] IU/L, respectively) from the same stable. In 2018, similar testing was performed in a nested case-control study that identified 27 case (serum GGT 50 ≥ IU/L)-control pairs from three stables for further testing. Serum liver chemistries, selenium measurements, viral PCR and metabolomics were performed. RESULTS No differences were found in frequency of detection of viral RNA/DNA or copy numbers for equine hepacivirus (EqHV) and parvovirus-hepatitis (EqPV-H) between cases and controls. Mild increases in hepatocellular injury and cholestatic markers in case vs control horses suggested a degree of liver disease in a subset of cases. Metabolomic and individual bile acid testing showed differences in cases compared with controls, including increased abundance of pyroglutamic acid and taurine-conjugated bile acids, and reduced abundance of Vitamin B6. Selenium concentrations, although within or above the reference intervals, were also lower in case horses in both studies. MAIN LIMITATIONS Observational study design did not allow us to make causal inferences. CONCLUSIONS We conclude that high GGT syndrome is likely a complex metabolic disorder and that viral hepatitis was not identified as a cause for this syndrome in this cohort of racehorses. Our results support a contribution of oxidative stress and cholestasis in its pathophysiology.
Collapse
Affiliation(s)
- Sabine Mann
- Department of Population Medicine and Diagnostic Sciences, Cornell College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Joshua D Ramsay
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Joseph J Wakshlag
- Department of Clinical Sciences, Cornell College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Tracy Stokol
- Department of Population Medicine and Diagnostic Sciences, Cornell College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Steven Reed
- Rood & Riddle Equine Hospital, Lexington, KY, USA
| | - Thomas J Divers
- Department of Clinical Sciences, Cornell College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
11
|
Xu WF, Zhang Q, Ding CJ, Sun HY, Che Y, Huang H, Wang Y, Wu JW, Hao HP, Cao LJ. Gasdermin E-derived caspase-3 inhibitors effectively protect mice from acute hepatic failure. Acta Pharmacol Sin 2021; 42:68-76. [PMID: 32457417 PMCID: PMC7921426 DOI: 10.1038/s41401-020-0434-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 05/09/2020] [Indexed: 12/15/2022] Open
Abstract
Programmed cell death (PCD), including apoptosis, apoptotic necrosis, and pyroptosis, is involved in various organ dysfunction syndromes. Recent studies have revealed that a substrate of caspase-3, gasdermin E (GSDME), functions as an effector for pyroptosis; however, few inhibitors have been reported to prevent pyroptosis mediated by GSDME. Here, we developed a class of GSDME-derived inhibitors containing the core structure of DMPD or DMLD. Ac-DMPD-CMK and Ac-DMLD-CMK could directly bind to the catalytic domains of caspase-3 and specifically inhibit caspase-3 activity, exhibiting a lower IC50 than that of Z-DEVD-FMK. Functionally, Ac-DMPD/DMLD-CMK substantially inhibited both GSDME and PARP cleavage by caspase-3, preventing apoptotic and pyroptotic events in hepatocytes and macrophages. Furthermore, in a mouse model of bile duct ligation that mimics intrahepatic cholestasis-related acute hepatic failure, Ac-DMPD/DMLD-CMK significantly alleviated liver injury. Together, this study not only identified two specific inhibitors of caspase-3 for investigating PCD but also, more importantly, shed light on novel lead compounds for treating liver failure and organ dysfunctions caused by PCD.
Collapse
Affiliation(s)
- Wan-Feng Xu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Quan Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Chu-Jie Ding
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Hui-Yong Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuan Che
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Hai Huang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Yun Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Jia-Wei Wu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China
| | - Hai-Ping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| | - Li-Juan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
12
|
Moore R, Anturaniemi J, Velagapudi V, Nandania J, Barrouin-Melo SM, Hielm-Björkman A. Targeted Metabolomics With Ultraperformance Liquid Chromatography-Mass Spectrometry (UPLC-MS) Highlights Metabolic Differences in Healthy and Atopic Staffordshire Bull Terriers Fed Two Different Diets, A Pilot Study. Front Vet Sci 2020; 7:554296. [PMID: 33195525 PMCID: PMC7653775 DOI: 10.3389/fvets.2020.554296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/22/2020] [Indexed: 01/09/2023] Open
Abstract
Background: While anecdotal evidence has long claimed that a raw meat-based diet (RMBD) improves the metabolic health of canines, no rigorous scientific study has clarified this issue. Canine atopic dermatitis (CAD) has also been linked to metabolic health, but its relation to diet remains poorly understood. This study investigates whether dietary choice is linked to metabolic health in healthy and CAD-diagnosed canines via targeted serum and urine metabolomic analysis of polar, non-ionic metabolites, as well as whether the underlying CAD condition modulates the response to nutritional intake. Materials and Methods: Serum metabolites of client-owned Staffordshire bull terriers, divided into CAD-diagnosed (n = 14) and healthy (n = 6) cohorts, were studied. Urine metabolites of a subset of the CAD-diagnosed canines (n = 8) were also studied. The canines were split into two cohorts based on diet. The first cohort were fed a commercially available high-fat, moderate-protein, low-carbohydrate RMBD (n = 11, CAD diagnosed n = 8, healthy n = 3). Those in the second cohort were fed a commercially available moderate-fat, moderate-protein, high-carbohydrate kibble diet (KD) (n = 9: CAD diagnosed n = 6, healthy n = 3). The diet intervention period lasted approximately 4.5 months (median 135 days). Statistical analyses of the serum profiles across all dogs (n = 20) and the urine profiles of the CAD-diagnosed subset (n = 8) were performed. Results and Discussion: The KD cohort was found to have higher concentrations of methionine than the RMBD cohort, both in serum (all dogs, p < 0.0001) and in urine (CAD-only cohort, p < 0.0002), as well as cystathionine and 4-pyridoxic acid. Methionine plays important roles in homocysteine metabolism, and elevated levels have been implicated in various pathologies. The CAD (n = 14) cohort dogs showed starker metabolic changes in response to diet regarding these pathways compared to the healthy (n = 6) cohort. However, there was no significant change in CAD severity as a result of either diet. Likely due to the higher meat content of the RMBD, higher concentrations of several carnitines and creatine were found in the RMBD cohort. Citrulline was found in higher concentrations in the KD cohort. Our findings provide insight into the relationship between diet and the serum and urine metabolite profiles of canines. They also suggest that neither diet significantly affected CAD severity.
Collapse
Affiliation(s)
- Robin Moore
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Anturaniemi
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Vidya Velagapudi
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jatin Nandania
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Stella Maria Barrouin-Melo
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Anna Hielm-Björkman
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Xiao Q, Zhang S, Ren H, Du R, Li J, Zhao J, Gao Y, Zhu Y, Huang W. Ginsenoside Rg1 alleviates ANIT-induced intrahepatic cholestasis in rats via activating farnesoid X receptor and regulating transporters and metabolic enzymes. Chem Biol Interact 2020; 324:109062. [PMID: 32198087 DOI: 10.1016/j.cbi.2020.109062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 12/12/2022]
Abstract
Ginsenoside Rg1 is an active ingredient extracted from the roots of ginsenoside, and an α-naphthylisothiocyanate (ANIT)-induced rat model of intrahepatic cholestasis was used to investigate the protective effect of Rg1 on cholestasis. 48 SD male rats were randomly divided into 6 groups: control group, model group, UDCA group (ursodeoxycholic acid), low-dose Rg1 group (10 mg/kg), medium-dose Rg1 group (20 mg/kg) and high-dose Rg1 group (40 mg/kg). The model group, the UDCA group and all the Rg1 group were then intragastrically administered with 80 mg/kg ANIT, and the control group were given equal volume of olive oil. Then the pathological changes in liver tissue were observed, the secretion of bile in the bile duct was measured, and the biochemical markers in serum were quantified, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), glutamyl transfer peptidase (GTP) and the content of total bilirubin (TBIL), direct bilirubin (DBIL), total bile acid (TBA). The contents of inflammatory mediators in serum were quantified, including tumor necrosis factor (TNF-α), γ-interferon (IFN-γ) and interleukin-1β (IL-1β). The contents of superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) in liver homogenate were quantified. Expression of farnesoid X receptor (FXR), transporters and metabolic enzymes in liver tissue was monitored. Rg1 treatment improved liver tissue pathological damage, promoted bile secretion and significantly reduced serum levels of the intrahepatic cholestasis markers ALT, AST, ALP, GTP, TBIL, DBIL and TBA. Rg1 increased the activity of SOD and GSH-Px in liver homogenate, while, reducing the serum levels of MDA and inflammatory mediators. Rg1 also regulated the expression of FXR, bile acid transporters and metabolic enzymes. Overall, Rg1 alleviated liver injury by improving secretion of bile and normalizing the activity of enzymes in the serum. The protective mechanism appeared to be related to the activation of FXR and regulation of liver transporters and metabolic enzymes.
Collapse
Affiliation(s)
- Qing Xiao
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shujun Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huina Ren
- Department of General Medicine, People's Hospital of Chongqing Bishan District, Chongqing, China
| | - Ruoyang Du
- Department of Urology, Chongqing Emergency Medical Center, Chongqing, China
| | - Jiajun Li
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinqiu Zhao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Gao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yali Zhu
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenxiang Huang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
14
|
Jia ET, Liu ZY, Pan M, Lu JF, Ge QY. Regulation of bile acid metabolism-related signaling pathways by gut microbiota in diseases. J Zhejiang Univ Sci B 2020; 20:781-792. [PMID: 31489798 DOI: 10.1631/jzus.b1900073] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the past decade, there has been increasing attention on the interaction between microbiota and bile acid metabolism. Bile acids are not only involved in the metabolism of nutrients, but are also important in signal transduction for the regulation of host physiological activities. Microbial-regulated bile acid metabolism has been proven to affect many diseases, but there have not been many studies of disease regulation by microbial receptor signaling pathways. This review considers findings of recent research on the core roles of farnesoid X receptor (FXR), G protein-coupled bile acid receptor (TGR5), and vitamin D receptor (VDR) signaling pathways in microbial-host interactions in health and disease. Studying the relationship between these pathways can help us understand the pathogenesis of human diseases, and lead to new solutions for their treatments.
Collapse
Affiliation(s)
- Er-Teng Jia
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhi-Yu Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Pan
- School of Medicine, Southeast University, Nanjing 210097, China
| | - Jia-Feng Lu
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, China
| | - Qin-Yu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
15
|
Blackmore D, Li L, Wang N, Maksymowych W, Yacyshyn E, Siddiqi ZA. Metabolomic profile overlap in prototypical autoimmune humoral disease: a comparison of myasthenia gravis and rheumatoid arthritis. Metabolomics 2020; 16:10. [PMID: 31902059 DOI: 10.1007/s11306-019-1625-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Myasthenia gravis (MG) and rheumatoid arthritis (RA) are examples of antibody-mediated chronic, progressive autoimmune diseases. Phenotypically dissimilar, MG and RA share common immunological features. However, the immunometabolomic features common to humoral autoimmune diseases remain largely unexplored. OBJECTIVES The aim of this study was to reveal and illustrate the metabolomic profile overlap found between these two diseases and describe the immunometabolomic significance. METHODS Metabolic analyses using acid- and dansyl-labelled was performed on serum from adult patients with seropositive MG (n = 46), RA (n = 23) and healthy controls (n = 49) presenting to the University of Alberta Hospital specialty clinics. Chemical isotope labelling liquid chromatography mass spectrometry (CIL LC-MS) methods were utilized to assess the serum metabolome in patients; 12C/13C-dansyl chloride (DnsCl) was used to label amine/phenol metabolites and 12C/13C-p-dimethylaminophenacyl bromide (DmPA) was used for carboxylic acids. Metabolites matching our criteria for significance were selected if they were present in both groups. Multivariate statistical analysis [including principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA)] and biochemical pathway analysis was then conducted to gain understanding of the principal pathways involved in antibody-mediated pathogenesis. RESULTS We found 20 metabolites dysregulated in both MG and RA when compared to healthy controls. Most prominently, observed changes were related to pathways associated with phenylalanine metabolism, tyrosine metabolism, ubiquinone and other terpenoid-quinone biosynthesis, and pyruvate metabolism. CONCLUSION From these results it is evident that many metabolites are common to humoral disease and exhibit significant immunometabolomic properties. This observation may lead to an enhanced understanding of the metabolic underpinnings common to antibody-mediated autoimmune disease. Further, contextualizing these findings within a larger clinical and systems biology context could provide new insights into the pathogenesis and management of these diseases.
Collapse
Affiliation(s)
- Derrick Blackmore
- Division of Neurology, University of Alberta, 7th Floor, Clinical Sciences Building, 11350 - 83 Ave NW, Edmonton, AB, T6G 2G3, Canada
| | - Liang Li
- Department of Chemistry, University of Alberta, Chemistry Centre Room W3-39C, Edmonton, AB, T6G 2G2, Canada
| | - Nan Wang
- Department of Chemistry, University of Alberta, Chemistry Centre Room W3-39C, Edmonton, AB, T6G 2G2, Canada
| | - Walter Maksymowych
- 568A Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Elaine Yacyshyn
- Division of Rheumatology, University of Alberta, 8-130 Clinical Sciences Building, 11350 - 83 Ave NW, Edmonton, AB, Canada
| | - Zaeem A Siddiqi
- Division of Neurology, University of Alberta, 7th Floor, Clinical Sciences Building, 11350 - 83 Ave NW, Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
16
|
Chen Y, Zhou J, Xu S, Liu M, Wang M, Ma Y, Zhao M, Wang Z, Guo Y, Zhao L. Association between the perturbation of bile acid homeostasis and valproic acid-induced hepatotoxicity. Biochem Pharmacol 2019; 170:113669. [DOI: 10.1016/j.bcp.2019.113669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/14/2019] [Indexed: 02/08/2023]
|
17
|
Hegade VS, Pechlivanis A, McDonald JAK, Rees D, Corrigan M, Hirschfield GM, Taylor-Robinson SD, Holmes E, Marchesi JR, Kendrick S, Jones DE. Autotaxin, bile acid profile and effect of ileal bile acid transporter inhibition in primary biliary cholangitis patients with pruritus. Liver Int 2019; 39:967-975. [PMID: 30735608 DOI: 10.1111/liv.14069] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/19/2019] [Accepted: 01/23/2019] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Pruritus is a common symptom in patients with primary biliary cholangitis (PBC) for which ileal bile acid transporter (IBAT) inhibition is emerging as a potential therapy. We explored the serum metabonome and gut microbiota profile in PBC patients with pruritus and investigated the effect of GSK2330672, an IBAT inhibitor. METHODS We studied fasting serum bile acids (BAs), autotaxin and faecal microbiota in 22 PBC patients with pruritus at baseline and after 2 weeks of GSK2330672 treatment. Control group included 31 asymptomatic PBC patients and 18 healthy volunteers. BA profiling was done by ultra performance liquid chromatography coupled to a mass spectrometry (UPLC-MS). Faecal microbiomes were analysed by 16S ribosomal RNA gene sequencing. RESULTS In PBC patients with pruritus, serum levels of total and glyco-conjugated primary BAs and autotaxin were significantly elevated. Autotaxin activity correlated significantly with tauro- and glyco-conjugated cholic acid (CA) and chenodeoxycholic acid (CDCA), both at baseline and after GSK2330672. GSK2330672 significantly reduced autotaxin and all tauro- and glyco- conjugated BAs and increased faecal levels of CA (P = 0.048) and CDCA (P = 0.027). Gut microbiota of PBC patients with pruritus was similar to control groups. GSK2330672 increased the relative abundance of Firmicutes (P = 0.033) and Clostridia (P = 0.04) and decreased Bacteroidetes (P = 0.033) and Bacteroidia (P = 0.04). CONCLUSIONS Pruritus in PBC does not show a distinct gut bacterial profile but is associated with elevated serum bile acid and autotaxin levels which decrease after IBAT inhibition. In cholestatic pruritus, a complex interplay between BAs and autotaxin is likely and may be modified by IBAT inhibition.
Collapse
Affiliation(s)
- Vinod S Hegade
- Institute of Cellular Medicine and NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Alexandros Pechlivanis
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Douglas Rees
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Margaret Corrigan
- NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| | - Gideon M Hirschfield
- NIHR Biomedical Research Unit, Centre for Liver Research, University of Birmingham, Birmingham, UK
| | - Simon D Taylor-Robinson
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Elaine Holmes
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Julian R Marchesi
- Department of Surgery and Cancer, Imperial College London, London, UK.,School of Biosciences, Cardiff University, Cardiff, UK
| | - Stuart Kendrick
- GlaxoSmithKline (GSK), Research and Development, Hertfordshire, UK
| | - David E Jones
- Institute of Cellular Medicine and NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
18
|
Zhang Y, Lu Y, Ji H, Li Y. Anti-inflammatory, anti-oxidative stress and novel therapeutic targets for cholestatic liver injury. Biosci Trends 2019; 13:23-31. [PMID: 30814402 DOI: 10.5582/bst.2018.01247] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cholestasis is a pathological process in which bile drainage is poor for a variety of reasons. Many studies have shown that cholestatic liver injury is a neutrophil-mediated inflammatory response, and oxidative stress induced by neutrophils is the main mechanism of liver cell death. The literature summarizes the bile acid signaling pathway, the neutrophil chemotaxis recruitment process during cholestasis, and the oxidative stress damage produced by neutrophil activation, summarizes the latest research progress. Sphingosine-1-phosphate receptor (S1PR) is a potential therapeutic target for cholestasis that reduces neutrophil aggregation without inhibiting systemic immune status. Early growth response factor 1 (Egr-1) may play a central role in the inflammation induced by cholestasis, and it is also a potential therapeutic target to inhibit the inflammation induced by cholestasis. Strengthening the antioxidant system of hepatocytes to cope with oxidative stress of neutrophils is a feasible treatment for cholestatic liver injury.
Collapse
Affiliation(s)
- Yafei Zhang
- Department of General Surgery, Second Affiliated Hospital of Xi'an Jiaotong University
| | - Yuxuan Lu
- The High School Affiliated to xi'an Jiaotong University
| | - Hong Ji
- Department of General Surgery, Second Affiliated Hospital of Xi'an Jiaotong University
| | - Yiming Li
- Department of General Surgery, Second Affiliated Hospital of Xi'an Jiaotong University
| |
Collapse
|
19
|
Burban A, Sharanek A, Humbert L, Eguether T, Guguen-Guillouzo C, Rainteau D, Guillouzo A. Predictive Value of Cellular Accumulation of Hydrophobic Bile Acids As a Marker of Cholestatic Drug Potential. Toxicol Sci 2019; 168:474-485. [DOI: 10.1093/toxsci/kfz009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Audrey Burban
- INSERM U1241, Numecan, Rennes, France
- University of Rennes 1, Rennes, France
| | - Ahmad Sharanek
- INSERM U1241, Numecan, Rennes, France
- University of Rennes 1, Rennes, France
| | - Lydie Humbert
- ERL INSERM U1157/UMR7203, Faculty of Medicine Pierre et Marie Curie Saint Antoine, Paris, France
| | - Thibaut Eguether
- ERL INSERM U1157/UMR7203, Faculty of Medicine Pierre et Marie Curie Saint Antoine, Paris, France
| | | | - Dominique Rainteau
- ERL INSERM U1157/UMR7203, Faculty of Medicine Pierre et Marie Curie Saint Antoine, Paris, France
| | - André Guillouzo
- INSERM U1241, Numecan, Rennes, France
- University of Rennes 1, Rennes, France
| |
Collapse
|
20
|
Malinen MM, Kauttonen A, Beaudoin JJ, Sjöstedt N, Honkakoski P, Brouwer KLR. Novel in Vitro Method Reveals Drugs That Inhibit Organic Solute Transporter Alpha/Beta (OSTα/β). Mol Pharm 2019; 16:238-246. [PMID: 30481467 PMCID: PMC6465078 DOI: 10.1021/acs.molpharmaceut.8b00966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Drug interactions with the organic solute transporter alpha/beta (OSTα/β) are understudied even though OSTα/β is an important transporter that is expressed in multiple human tissues including the intestine, kidneys, and liver. In this study, an in vitro method to identify novel OSTα/β inhibitors was first developed using OSTα/β-overexpressing Flp-In 293 cells. Incubation conditions were optimized using previously reported OSTα/β inhibitors. A method including a 10 min preincubation step with the test compound was used to screen for OSTα/β inhibition by 77 structurally diverse compounds and fixed-dose combinations. Seven compounds and one fixed-dose combination (100 μM final concentration) inhibited OSTα/β-mediated dehydroepiandrosterone sulfate (DHEAS) uptake by >25%. Concentration-dependent OSTα/β inhibition was evaluated for all putative inhibitors (atorvastatin, ethinylestradiol, fidaxomicin, glycochenodeoxycholate, norgestimate, troglitazone, and troglitazone sulfate). Ethinylestradiol, fidaxomicin, and troglitazone sulfate yielded a clear concentration-inhibition response with IC50 values <200 μM. Among all tested compounds, there was no clear association between physicochemical properties, the severity of hepatotoxicity, and the degree of OSTα/β inhibition. This study utilized a novel in vitro method to identify OSTα/β inhibitors and, for the first time, provided IC50 values for OSTα/β inhibition. These data provide evidence that several drugs, some of which are associated with cholestatic drug-induced liver injury, may impair the function of the OSTα/β transporter.
Collapse
Affiliation(s)
- Melina M. Malinen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Antti Kauttonen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - James J. Beaudoin
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Noora Sjöstedt
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paavo Honkakoski
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kim L. R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
21
|
Becerra-Verdín EM, Morales Ávila ÚM, García-Galindo HS, Montalvo-González R, Castañeda-Martínez A, Montalvo-González E. Evaluation of biochemical markers in diabetic rats fed diets supplemented with fruit purees. CYTA - JOURNAL OF FOOD 2019. [DOI: 10.1080/19476337.2019.1578267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Eduardo Mendeleev Becerra-Verdín
- Laboratorio de Investigación Clínica e Histología, Unidad Académica de Ciencias Químico Bilógicas y Farmaceúticas, Universidad Autónoma de Nayarit, Tepic Nayarit, Mexico
| | - Úrsula Mireya Morales Ávila
- Laboratorio Integral de Investigación en Alimentos, División de Estudios de Posgrado e Invesgación, Tecnológico Nacional de México/Campus Instituto Tecnológico de Tepic, Tepic Nayarit, Mexico
| | - Hugo Sergio García-Galindo
- UNIDA, División de estudios de Posgrado e Investigación, Tecnológico Nacional de México/Campus Instituto Tecnológico de Veracruz, Veracruz, Mexico
| | - Rubén Montalvo-González
- Laboratorio de Investigación Clínica e Histología, Unidad Académica de Ciencias Químico Bilógicas y Farmaceúticas, Universidad Autónoma de Nayarit, Tepic Nayarit, Mexico
| | - Alfonso Castañeda-Martínez
- Laboratorio de Investigación Clínica e Histología, Unidad Académica de Ciencias Químico Bilógicas y Farmaceúticas, Universidad Autónoma de Nayarit, Tepic Nayarit, Mexico
| | - Efigenia Montalvo-González
- Laboratorio Integral de Investigación en Alimentos, División de Estudios de Posgrado e Invesgación, Tecnológico Nacional de México/Campus Instituto Tecnológico de Tepic, Tepic Nayarit, Mexico
| |
Collapse
|
22
|
Gabbia D, Pozzo L, Zigiotto G, Roverso M, Sacchi D, Dalla Pozza A, Carrara M, Bogialli S, Floreani A, Guido M, De Martin S. Dexamethasone counteracts hepatic inflammation and oxidative stress in cholestatic rats via CAR activation. PLoS One 2018; 13:e0204336. [PMID: 30252871 PMCID: PMC6155538 DOI: 10.1371/journal.pone.0204336] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/06/2018] [Indexed: 12/19/2022] Open
Abstract
Glucocorticoids (GCs) are currently used for the therapeutic management of cholestatic diseases, but their use and molecular mechanism remain controversial. The aims of this study were 1) to assess the therapeutic effect of a 2-week treatment with the GC dexamethasone on hepatic damage in bile duct-ligated rats; 2) to investigate its effect on the activation of the nuclear receptors (NRs) pregnane X receptor (PXR), constitutive androstane receptor (CAR) and GC receptor (GR), and NF-kB, as well as on oxidative stress and bile acid (BA) hepatic composition. Cholestasis was induced by ligation of bile duct (BDL animals) in 16 male Wistar-Kyoto rats, and eight of them were daily treated by oral gavage with 0.125 mg/ml/kg DEX for 14 days. Eight Sham-operated rats were used as controls. Severity of cholestasis was assessed histologically and on plasma biochemical parameters. The nuclear expression of NF-kB (p65), GR, PXR and CAR was measured in hepatic tissue by Western Blot. Oxidative stress was evaluated by measuring malondialdehyde, carbonylated proteins, GHS and ROS content in rat livers. LC-MS was used to measure the plasma and liver concentration of 7 BAs. Histological findings and a significant drop in several markers of inflammation (p65 nuclear translocation, mRNA expressions of TNF-α, IL-1β, IL-6) showed that DEX treatment reversed cholestasis-induced inflammation, and similar results have been obtained with oxidative stress markers. The nuclear expression of p65 and CAR were inversely correlated, with the latter increasing significantly after DEX treatment (p<0.01 vs vehicle). Hepatic BA levels tended to drop in the untreated cholestatic rats, whereas they were similar to those of healthy rats in DEX-treated animals. Plasma BAs decreased significantly in DEX-treated animals with respect to untreated cholestatic rats. In conclusion, DEX reduces inflammation and oxidative stress in BDL rats, and probably CAR is responsible for this effect. Therefore, this NR represents a promising pharmacological target for managing cholestatic and inflammatory liver diseases.
Collapse
Affiliation(s)
- Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Luisa Pozzo
- Institute of Agricultural Biology and Biotechnology, CNR, Pisa, Italy
| | - Giorgia Zigiotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Marco Roverso
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Diana Sacchi
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Arianna Dalla Pozza
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Maria Carrara
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Sara Bogialli
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Annarosa Floreani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Maria Guido
- Department of Medicine, General Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- * E-mail:
| |
Collapse
|
23
|
Luo L, Aubrecht J, Li D, Warner RL, Johnson KJ, Kenny J, Colangelo JL. Assessment of serum bile acid profiles as biomarkers of liver injury and liver disease in humans. PLoS One 2018. [PMID: 29513725 PMCID: PMC5841799 DOI: 10.1371/journal.pone.0193824] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To assess the potential of individual bile acids (IBA) and their profiles as mechanistic biomarkers of liver injury for humans in real world situations, we interrogated samples collected under minimum controlled conditions (ie subjects were not fasted). Total bile acids (TBA) have been considered to be biomarkers of liver injury for decades, and more recently, monitoring of IBA has been proposed for differentiation of variety of etiologies of liver injury. We established a LC-MS/MS methodology to analyze nine IBA, generated reference ranges, and examined effects of age, gender, and ethnicity for each IBA. Furthermore, we evaluated the ability of IBA and their profiles to detect hepatic injury in subjects with a broad range of liver impairments. To date, our study utilized the largest total cohort of samples (N = 645) that were divided into 2 groups, healthy or liver impaired, to evaluate IBA as biomarkers. The TBA serum levels in the Asian ethnic group trended higher when compared to other ethnic groups, and the serum concentrations of IBA, such as glycocholic acid (GCA), glycochenodeoxycholic acid (GCDCA), chenodeoxycholic acid (CDCA), and taurochenoxycholic acid (TCDCA) were significantly increased. To our knowledge, this report is the first to describe ethnic differences in serum concentrations of IBAs. In patients with hepatic impairments, with the exception of deoxycholic acid (DCA), the concentrations of IBAs were significantly elevated when compared with healthy subjects. The conjugated bile acids displayed greater differences between healthy subjects and subjects with hepatic impairments than non-conjugated bile acids. Furthermore, the subjects with hepatic impairments exhibited distinct profiles (signatures) of IBAs that clustered subjects according the nature of their liver impairments. Although additional studies are needed, our data suggested that the analysis of IBA has the potential to become useful for differentiation of various forms of liver injury.
Collapse
Affiliation(s)
- Lina Luo
- Pharmacokinetics, Dynamics & Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut, United States of America
- * E-mail:
| | - Jiri Aubrecht
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Dingzhou Li
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Roscoe L. Warner
- University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kent J. Johnson
- University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Julia Kenny
- University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Jennifer L. Colangelo
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut, United States of America
| |
Collapse
|
24
|
Di Guida F, Pirozzi C, Magliocca S, Santoro A, Lama A, Russo R, Nieddu M, Burrai L, Boatto G, Mollica MP, Sodano F, Lazzarato L, Chegaev K, Meli R, Raso GM, Rimoli MG. Galactosylated Pro-Drug of Ursodeoxycholic Acid: Design, Synthesis, Characterization, and Pharmacological Effects in a Rat Model of Estrogen-Induced Cholestasis. Mol Pharm 2017; 15:21-30. [PMID: 29140706 DOI: 10.1021/acs.molpharmaceut.7b00626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ursodeoxycholic acid (UDCA) is considered the first-choice therapy for cholestatic disorders. To enhance solubility and exploit specific transporters in liver, we synthesized a new galactosyl pro-drug of UDCA (UDCAgal). Ethinylestradiol (EE)-induced cholestasis was used to study and compare the effects of UDCAgal with UDCA on bile flow, hepatic canalicular efflux transporter expression, and inflammation. UDCAgal resulted quite stable both at pH 7.4 and 1.2 and regenerated the parent drug after incubation in human plasma. Its solubility, higher than UDCA, was pH- and temperature-independent. UDCAgal displayed a higher cell permeation compared to UDCA in liver HepG2 cells. Moreover, in cholestatic rats, UDCAgal showed a higher potency compared to UDCA in reducing serum biomarkers (AST, ALT, and ALP) and cytokines (TNF-α and IL-1β). The higher effect of UDCAgal on the increase in bile salt export pump and multidrug resistance-associated protein 2 transcription indicated an improved spillover of bile acids from the liver. UDCAgal showed a reduction in CCL2, as well as TNF-α, IL-1β, and cyclooxygeanse-2 mRNAs, indicating a reduction in hepatic neutrophil accumulation and inflammation. Moreover, UDCAgal, similarly to UDCA, heightens bile flow and modulates biliary acids secretion. These results indicate that UDCAgal has a potential in the treatment of cholestatic disease.
Collapse
Affiliation(s)
- Francesca Di Guida
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| | - Salvatore Magliocca
- Department of Chemistry and Pharmacy, University of Sassari , Sassari 07100, Italy
| | - Anna Santoro
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| | - Adriano Lama
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| | - Maria Nieddu
- Department of Chemistry and Pharmacy, University of Sassari , Sassari 07100, Italy
| | - Lucia Burrai
- Department of Chemistry and Pharmacy, University of Sassari , Sassari 07100, Italy
| | - Gianpiero Boatto
- Department of Chemistry and Pharmacy, University of Sassari , Sassari 07100, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II , Naples 80138, Italy
| | - Federica Sodano
- Department of Drug Science and Technology, University of Turin , Turin 10124, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin , Turin 10124, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin , Turin 10124, Italy
| | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| | | | - Maria Grazia Rimoli
- Department of Pharmacy, University of Naples Federico II , Naples 80138, Italy
| |
Collapse
|
25
|
Marchionatti AM, Pérez A, Rivoira MA, Rodríguez VA, Tolosa de Talamoni NG. Lithocholic acid: a new emergent protector of intestinal calcium absorption under oxidant conditions. Biochem Cell Biol 2017; 95:273-279. [PMID: 28318299 DOI: 10.1139/bcb-2016-0164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
LCA and 1,25(OH)2D3 are vitamin D receptor ligands with different binding affinity. The secosteroid stimulates intestinal Ca2+ absorption. Whether LCA alters this process remains unknown. The aim of our work was to determine the effect of LCA on intestinal Ca2+ absorption in the absence or presence of NaDOC, bile acid that inhibits the cation transport. The data show that LCA by itself did not alter intestinal Ca2+ absorption, but prevented the inhibitory effect of NaDOC. The concomitant administration of LCA avoided the reduction of intestinal alkaline phosphatase activity caused by NaDOC. In addition, LCA blocked a decrease caused by NaDOC on gene and protein expression of molecules involved in the transcellular pathway of intestinal Ca2+ absorption. The oxidative stress and apoptosis triggered by NaDOC were abrogated by LCA co-treatment. In conclusion, LCA placed in the intestinal lumen protects intestinal Ca2+ absorption against the inhibitory effects caused by NaDOC. LCA avoids the reduction of the transcellular Ca2+ movement, apparently by blocking the oxidative stress and apoptosis triggered by NaDOC, normalizing the gene and protein expression of molecules involved in Ca2+ movement. Therefore, LCA might become a possible treatment to improve intestinal calcium absorption under oxidant conditions.
Collapse
Affiliation(s)
- Ana M Marchionatti
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina.,Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina
| | - Adriana Pérez
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina.,Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina
| | - María A Rivoira
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina.,Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina
| | - Valeria A Rodríguez
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina.,Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina
| | - Nori G Tolosa de Talamoni
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina.,Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba, Argentina
| |
Collapse
|
26
|
|
27
|
Gomez-Quiroz LE, Seo D, Lee YH, Kitade M, Gaiser T, Gillen M, Lee SB, Gutierrez-Ruiz MC, Conner EA, Factor VM, Thorgeirsson SS, Marquardt JU. Loss of c-Met signaling sensitizes hepatocytes to lipotoxicity and induces cholestatic liver damage by aggravating oxidative stress. Toxicology 2016; 361-362:39-48. [PMID: 27394961 DOI: 10.1016/j.tox.2016.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/18/2016] [Accepted: 07/05/2016] [Indexed: 02/08/2023]
Abstract
Recent studies confirmed a critical importance of c-Met signaling for liver regeneration by modulating redox balance. Here we used liver-specific conditional knockout mice (MetKO) and a nutritional model of hepatic steatosis to address the role of c-Met in cholesterol-mediated liver toxicity. Liver injury was assessed by histopathology and plasma enzymes levels. Global transcriptomic changes were examined by gene expression microarray, and key molecules involved in liver damage and lipid homeostasis were evaluated by Western blotting. Loss of c-Met signaling amplified the extent of liver injury in MetKO mice fed with high-cholesterol diet for 30days as evidenced by upregulation of liver enzymes and increased synthesis of total bile acids, aggravated inflammatory response and enhanced intrahepatic lipid deposition. Global transcriptomic changes confirmed the enrichment of networks involved in steatosis and cholestasis. In addition, signaling pathways related to glutathione and lipid metabolism, oxidative stress and mitochondria dysfunction were significantly affected by the loss of c-Met function. Mechanistically, exacerbation of oxidative stress in MetKO livers was corroborated by increased lipid and protein oxidation. Western blot analysis further revealed suppression of Erk, NF-kB and Nrf2 survival pathways and downstream target genes (e.g. cyclin D1, SOD1, gamma-GCS), as well as up-regulation of proapoptotic signaling (e.g. p53, caspase 3). Consistent with the observed steatotic and cholestatic phenotype, nuclear receptors RAR, RXR showed increased activation while expression levels of CAR, FXR and PPAR-alpha were decreased in MetKO. Collectively, our data provide evidence for the critical involvement of c-Met signaling in cholesterol and bile acids toxicity.
Collapse
Affiliation(s)
- Luis E Gomez-Quiroz
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA; Departamento de Ciencias de la Salud, Universidad Autonoma Metropolitana Iztapalapa, Mexico, DF, Mexico
| | - Daekwan Seo
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yun-Han Lee
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA; Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Mitsuteru Kitade
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Timo Gaiser
- Institute of Pathology, University Medical Center Mannheim, Mannheim, Germany
| | - Matthew Gillen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Seung-Bum Lee
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Elizabeth A Conner
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Valentina M Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Jens U Marquardt
- 1st Department of Medicine, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany.
| |
Collapse
|